ClickCease
+1-915-850-0900 spinedoctors@gmail.com
Select Page

Functional Medicine

Back Clinic Functional Medicine Team. Functional medicine is an evolution in the practice of medicine that better addresses the healthcare needs of the 21st century. By shifting the traditional disease-centered focus of medical practice to a more patient-centered approach, functional medicine addresses the whole person, not just an isolated set of symptoms.

Practitioners spend time with their patients, listening to their histories and looking at the interactions among genetic, environmental, and lifestyle factors that can influence long-term health and complex, chronic disease. In this way, functional medicine supports the unique expression of health and vitality for each individual.

By changing the disease-centered focus of medical practice to this patient-centered approach, our physicians are able to support the healing process by viewing health and illness as part of a cycle in which all components of the human biological system interact dynamically with the environment. This process helps to seek and identify genetic, lifestyle, and environmental factors that may shift a person’s health from illness to well-being.


Essential Mineral Intake for Cardiovascular Disease | Wellness Clinic

Essential Mineral Intake for Cardiovascular Disease | Wellness Clinic

An improper diet and nutrition can often lead to a variety of health issues, such as cardiovascular disease and hypertension. Additionally, other food-related risk factors can include, high blood pressure, or BP, obesity and type 2 diabetes. Saturated and trans fats, for example, can increase cholesterol in the blood. It’s this build-up of fatty deposits in the coronary arteries, which can lead to cardiovascular disease, hypertension and even heart attacks.

 

Which vitamins and minerals can improve cardiovascular disease and hypertension?

 

A healthy balance of vitamins and minerals can help manage and maintain heart health. According to research studies, the right intake of potassium, magnesium, calcium and zinc can substantially improve cardiovascular disease and hypertension. A proper diet and nutrition can have similar effects as taking cardiovascular disease and hypertension drugs and medications, but through a much more natural approach.

Potassium

The average U.S. dietary intake of potassium (K+) is 45 mmol/d with a potassium to sodium (K+/Na+) ratio of less than 1:2. The suggested intake of K+ is 4700 mg/d (120 mmol) with a K+/Na+ ratio of about 4-5 to 1. Several clinical and observational trials have demonstrated a substantial decrease in BP with greater K+ intake in hypertensive patients. The normal blood pressure reduction with a K+ supplementation of 60 to 120 mmol/d is 4.4/2.5 mmHg in hypertensive patients but may be as far as 8/4.1 mmHg with 120 mmol/d (4700 mg). In hypertensive patients, the linear dose response relationship is 1.0 mmHg reduction in systolic blood pressure, or SBP, and 0.52 mmHg decrease in diastolic blood pressure, or DBP, that a 0.6 g/d growth in dietary fiber intake. The solution can involve race (black > white), sodium, magnesium and calcium intake. Those on a higher sodium intake have a greater decrease in BP. Alteration of this K+/Na+ ratio is very important to the two polyunsaturated and outcomes. High potassium intake reduces the prevalence of cardiovascular disease independent of their BP reduction. Furthermore, If the serum potassium is less than 4.0 meq/dL, there is a higher risk of CVD mortality, ventricular tachycardia, and ventricular fibrillation. Red blood cell potassium is a sign of overall body stores and CVD risk in comparison to the serum potassium. Gu et al discovered that potassium supplementation in 60 mmol of KCl Daily for 12 wk significantly reduced SBP -5.0 mmHg (range -2.13 into -7.88 mmHg) (p < 0.001) in 150 Chinese men and women aged 35 to 64 decades.

 

Insulin raises natriuresis, modulates sensitivity, vasodilates, reduces the sensitivity to catecholamines and Angiotensin II, raises nitric oxide ATPase and DNA synthesis in the vascular smooth muscle cells and decreases sympathetic nervous system activity. In addition, potassium increases bradykinin and prostate kallikrein, decreases NADPH oxidase, which reduces oxidative stress and inflammation, improves insulin sensitivity, reduces ADMA, reduces intracellular sodium and reduces production of TGF-?.Each 1000 mg increase in potassium intake per day reduces all cause mortality by approximately 20 percent. Potassium intake of 4.7 g/d is estimated to decrease CVA by 8 percent to 15 percent and MI by 6 percent to 11 percent. Numerous SNP’s, such as nuclear receptor subfamily 3 group C, angiotensin IItype receptor and hydroxysteroid 11 beta dehydrogenase (HSD11B1 and B2) determine an individual’s reaction to dietary potassium intake towards their overall health and wellness.

 

Each 1000 mg drop in sodium intake daily will reduce all cause mortality. A recent study indicated a dose related response to CVA. There has been a RRR of CVA of 23 percent at 1.5-1.99 gram, 27% at 2.0-2.49 g, 29 percent at 2.5-3 g and 32 percent more than 3 g/d of potassium urinary excretion. The recommended daily dietary intake for individuals with hypertension is 4.7 to 5.0 g of potassium and less than 1500 milligrams of sodium. Potassium used out of supplementation should be decreased with care in patients with renal impairment or those ARB, DRI and serum aldosterone receptor antagonists.

 

Magnesium

 

A high dietary intake of magnesium of at least 500-1000 mg/d reduces BP in the majority of the reported observational epidemiologic and clinical trials, but the outcomes are much less consistent than those seen with K + and Na +. There’s an inverse relationship between BP and dietary magnesium intake. A report on 60 essential hypertensive subjects given magnesium supplements showed a substantial decrease in blood pressure in an eight week interval reported by 24 h ambulatory BP, office and home BP. The maximum decrease in clinical trials has been 5.6/2.8 mmHg but some studies have shown no change in BP. The blend of high potassium and low sodium intake with increased magnesium intake had.

 

Magnesium also raises the effectiveness of all anti-hypertensive drugs and medications, according to research studies. Magnesium competes with Na+ for binding sites on vascular smooth muscle and also functions as a direct vasodilator, . Magnesium increases prostaglandin E (PGE), modulates intracellular sodium, potassium, calcium and pH, increases nitric oxide, improves adrenal function, reduces oxLDL, reduces HS-CRP, TBxA2, A-II, and norepinephrine. Magnesium also enhances insulin resistance, glucose and MS, binds at a necessary cooperative manner with potassium, causing EDV and BP reduction, reduces CVD and cardiac arrhythmias, reduces carotid IMT, reduces cholesterol, reduces cytokine production, inhibits nuclear factor Kb, reduces oxidative stress and inhibits platelet aggregation to reduce thrombosis. Magnesium is an essential co-factor because of its delta-6-desaturase enzyme that for conversion of linoleic acid (LA) to gamma linolenic acid (GLA) required for synthesis of this vasodilator and platelet inhibitor PGE1.

 

A meta-analysis of all 241378 patients utilizing 6477 strokes showed a reverse relationship of dietary magnesium to the incidence of stroke. For each 100 milligrams of magnesium intake, stroke diminished. The mechanism comprise inhibition of induced glutamate release, NMDA receptor blockade, CCB actions reduction in vasodilation and ATP depletion of the arteries. A meta-analysis showed discounts mmHg in 22 trials of 1173 patients. Intracellular level of calcium (RBC) is more indicative of overall body shops and should be quantified along with serum and urinary magnesium. Magnesium might be supplemented in doses of 500. Magnesium formulations may improve absorption and reduce the incidence of diarrhea. Adding taurine in 1000 increases the ramifications of magnesium. Magnesium supplements should be avoided or used with caution in individuals with renal insufficiency.

 

Calcium

 

Population studies reveal a link between hypertension and calcium, but clinical trials that handled calcium supplements have shown consequences on blood pressure. The heterogeneous responses to calcium supplementation have been clarified through research studies. This is really the “ionic hypothesis” of hypertension, cardiovascular disease and associated cognitive, cognitive and functional disorders. Calcium supplementation is not recommended at this time as an effective method to decrease blood pressure due to insufficient research studies on its use.

 

Zinc

 

Low serum zinc levels in observational research and hypertension correlate as well as CHD, type II DM, hyperlipidemia, elevated lipoprotein that a [Lp(a)], increased 2 h post-prandial plasma glucose levels and insulin resistance. Zinc is hauled to vascular and cardiac muscle and cells by metallothionein. Deficiencies of metallothionein with intramuscular zinc deficiencies can lead to cardiomyocyte oxidative stress , mitochondrial dysfunction, dysfunction and apoptosis with cardiac remodeling hypertension, cardiovascular disease, heart failure, or fibrosis. Intracellular calcium increases oxidative.

 

Bergomi et al assessed Zinc (Zn++) status in 60 hypertensive subjects compared to 60 normotensive control subjects. A reverse correlation of serum Zn++ and BP has been observed. The BP was inversely associated with a Zn++ dependent enzyme lysyl oxidase activity. Zn++ inhibits gene expression and transcription through NF-?Band activated protein-1 and is now a significant co-factor for SOD. These impacts plus those on insulin resistance and SNS consequences, membrane ion exchange, RAAS might account for Zn++ antihypertensive effects. Intake needs to be 50 mg/d.

 

Individuals with cardiovascular disease and hypertension can benefit from the proper diet and nutrition. Essential vitamins and minerals found in a balanced, healthy nutrition, such as potassium, magnesium, calcium and zing, among others, can help improve heart health. Deficiencies in these and a diet full of saturated and trans fats can increase the prevalence of cardiovascular disease. While diagnosis and drugs/medications can be prescribed to treat cardiovascular disease and hypertension, a balanced diet and nutrition can have similar effects.� The scope of our information is limited to chiropractic and spinal injuries and conditions. To discuss the subject matter, please feel free to ask Dr. Jimenez or contact us at 915-850-0900 .

By Dr. Alex Jimenez

Green-Call-Now-Button-24H-150x150-2-3.png

 

Additional Topics: Wellness

 

Overall health and wellness are essential towards maintaining the proper mental and physical balance in the body. From eating a balanced nutrition as well as exercising and participating in physical activities, to sleeping a healthy amount of time on a regular basis, following the best health and wellness tips can ultimately help maintain overall well-being. Eating plenty of fruits and vegetables can go a long way towards helping people become healthy.

 

blog picture of cartoon paperboy big news

 

TRENDING TOPIC: EXTRA EXTRA: About Chiropractic

 

 

GM Crops: The  Limitations, Risks, And Alternatives

GM Crops: The Limitations, Risks, And Alternatives

GM Crops: Proponents claim that genetically modified (GM) crops:

 

  • are safe to eat and more nutritious
  • beneft the environment
  • reduce use of herbicides and insecticides
  • increase crop yields, thereby helping farmers and solving the food crisis
  • create a more affuent, stable economy
  • are just an extension of natural breeding, and have no risks different from naturally bred crops.

However, a large and growing body of scientifc research and on-the-ground experience indicate that GMOs fail to live

up to these claims. Instead, GM crops:

 

  • can be toxic, allergenic or less nutritious than their natural counterparts
  • can disrupt the ecosystem, damage vulnerable wild plant and animal populations and harm biodiversity
  • increase chemical inputs (pesticides, herbicides) over the long term
  • deliver yields that are no better, and often worse, than conventional crops
  • cause or exacerbate a range of social and economic problems
  • are laboratory-made and, once released, harmful GMOs cannot be recalled from the environment.

The scientifically demonstrated risks and clear absence of real benefits have led experts to see GM as a clumsy, outdated technology. They present risks that we need not incur, given the availability of effective, scientifically proven, energy-efficient and safe ways of meeting current and future global food needs.

This paper presents the key scientific evidence � 114 research studies and other authoritative documents � documenting the limitations and risks of GM crops and the many safer, more effective alternatives available today.

Is GM An Extension Of Natural Plant Breeding?

Natural reproduction or breeding can only occur between closely related forms of life (cats with cats, not cats with dogs; wheat with wheat, not wheat with tomatoes or fish). In this way, the genes that offspring inherit from parents, which carry information for all parts of the body, are passed down the generations in an orderly way.

GM is not like natural plant breeding. GM uses laboratory techniques to insert artificial gene units to re-program the DNA blueprint of the plant with completely new properties. This process would never happen in nature. The artificial gene units are created in the laboratory by joining fragments of DNA, usually derived from multiple organisms, including viruses, bacteria, plants and animals. For example, the GM gene in the most common herbicide resistant soya beans was pieced together from a plant virus, a soil bacterium and a petunia plant.

The GM transformation process of plants is crude, imprecise, and causes widespread mutations, resulting in major changes to the plant�s DNA blueprint1. These mutations unnaturally alter the genes� functioning in unpredictable and potentially harmful ways2, as detailed below. Adverse effects include poorer crop performance, toxic effects, allergic reactions, and damage to the environment.Are GM foods safe to eat?Contrary to industry claims, GM foods are not properly tested for human safety before they are released for sale3 4. In fact, the only published study directly testing the safety of a GM food on humans found potential problems5. To date, this study has not been followed up. Typically the response to the safety question is that people have been eating GM foods in the United States and elsewhere for more than ten years without ill effects and that this proves that the products are safe. But GM foods are not labelled in the US and other nations where they are widely eaten and consumers are not monitored for health effects.

Because of this, any health effects from a GM food would have to meet unusual conditions before they would be noticed. The health effects would have to:

� occur immediately after eating a food that was known to be GM (in spite of its not being labeled). This kind of response is called acute toxicity.

� cause symptoms that are completely different from common diseases. If GM foods caused a rise in common or slow-onset diseases like allergies or cancer, nobody would know what caused the rise.

� be dramatic and obvious to the naked eye. Nobody examines a person�s body tissues with a microscope for harm after they eat a GM food. But just this type of examination is needed to give early warning of problems such as pre-cancerous changes.

To detect important but more subtle effects on health, or effects that take time to appear (chronic effects), long-term controlled studies on larger populations are required.

Under current conditions, moderate or slow-onset health effects of GM foods could take decades to become known, just as it took decades for the damaging effects of trans-fats (another type of artificial food) to be recognized. �Slow poison� effects from trans-fats have caused millions of premature deaths across the world6 .

Another reason why any harmful effects of GM foods will be slow to surface and less obvious is because, even in the United States, which has the longest history of GM crop consumption, GM foods account for only a small part of the US diet (maize is less than 15% and soya bean products are less than 5%).

Nevertheless, there are signs that all is not well with the US food supply. A report by the US Centers for Disease Control shows that food-related illnesses increased 2- to 10-fold in the years between 1994 (just before GM food was commercialized) and 19997 . Is there a link with GM food? No one knows, because studies on humans have not been done.

Animal Studies On GM Foods Give Cause For Concern

Although studies on humans have not been done, scientists are reporting a growing number of studies that examine the effects of GM foods on laboratory animals. These studies, summarized below, raise serious concerns regarding the safety of GM foods for humans as well as animals.

Small Animal Feeding Studies

� Rats fed GM tomatoes developed stomach ulcerations8

� Liver, pancreas and testes function was disturbed in mice fed GM soya9 10 11

� GM peas caused allergic reactions in mice12

� Rats fed GM oilseed rape developed enlarged livers, often a sign of toxicity13

� GM potatoes fed to rats caused excessive growth of the lining of the gut similar to a pre-cancerous condition14 15

� Rats fed insecticide-producing GM maize grew more slowly, suffered problems with liver and kidney function, and showed higher levels of certain fats in their blood16

� Rats fed GM insecticide-producing maize over three generations suffered damage to liver and kidneys and showed alterations in blood biochemistry17

� Old and young mice fed with GM insecticide-producing maize showed a marked disturbance in immune system cell populations and in biochemical activity18

� Mice fed GM insecticide-producing maize over four generations showed a buildup of abnormal structural changes in various organs (liver, spleen, pancreas), major changes in the pattern of gene function in the gut, reflecting disturbances in the chemistry of this organ system (e.g. in cholesterol production, protein production and breakdown), and, most significantly, reduced fertility19

� Mice fed GM soya over their entire lifetime (24 months) showed more acute signs of aging in their liver20

� Rabbits fed GM soya showed enzyme function disturbances in kidney and heart21.

Feeding Studies With Farm Animals

Farm animals have been fed GM feed for many years. Does this mean that GM feed is safe for livestock? Certainly it means that effects are not acute and do not show up immediately. However, longer-term studies, designed to assess slow-onset and more subtle health effects of GM feed, indicate that GM feed does have adverse effects, confirming the results described above for laboratory animals.

The following problems have been found:

� Sheep fed Bt insecticide-producing GM maize over three generations showed disturbances in the functioning of the digestive system of ewes and in the liver and pancreas of their lambs22.

� GM DNA was found to survive processing and to be detectable in the digestive tract of sheep fed GM feed. This raises the possibility that antibiotic resistance and Bt insecticide genes can move into gut bacteria23, a process known as horizontal gene transfer. Horizontal gene transfer can lead to antibiotic resistant disease causing bacteria (�superbugs�) and may lead to Bt insecticide being produced in the gut with potentially harmful consequences. For years, regulators and the biotech industry claimed that horizontal gene transfer would not occur with GM DNA, but this research challenges this claim

� GM DNA in feed is taken up by the animal�s organs. Small amounts of GM DNA appear in the milk and meat that people eat24 25 26. The effects on the health of the animals and the people who eat them have not been researched.

Do Animal Feeding Studies Highlight Potential Health Problems For People?

Before food additives and new medicines can be tested on human subjects, they have to be tested on mice or rats. If harmful effects were to be found in these initial animal experiments, then the drug would likely be disqualified for human use. Only if animal studies reveal no harmful effects can the drug be further tested on human volunteers.

But GM crops that caused ill effects in experimental animals have been approved for commercialization in many countries. This suggests that less rigorous standards are being used to evaluate the safety of GM crops than for new medicines.

In fact, in at least one country � the United States � safety assessment of GMOs is voluntary and not required by law, although, to date, all GMOs have undergone voluntary review. In virtually all countries, safety assessment is not scientifically rigorous. For instance, the animal feeding studies that GM crop developers routinely conduct to demonstrate the safety of their products are too short in duration and use too few subjects to reliably detect important harmful effects.27

While industry conducts less than rigorous studies on its own GM products, 28 it has, in parallel, systematically and persistently interfered with the ability of independent scientists to conduct more rigorous and incisive independent research on GMOs. Comparative and basic agronomic studies on GMOs, assessments of safety and composition, and assessments of environmental impact have all been restricted and suppressed by the biotechnology industry.29 30

Patent rights linked with contracts are used to restrict access of independent researchers to commercialized GM seed. Permission to study patented GM crops is either withheld or made so difficult to obtain that research is effectively blocked. In cases where permission is finally given, biotech companies keep the right to block publication, resulting in much significant research never being published.31 32

The industry and its allies also use a range of public relations strategies to discredit and/or muzzle scientists who do publish research that is critical of GM crops.33

Are GM Foods More Nutritious?

There are no commercially available GM foods with improved nutritional value. Currently available GM foods are no better and in some cases are less nutritious than natural foods. Some have been proven in tests to be toxic or allergenic.

Examples include:

� GM soya had 12�14% lower amounts of cancer-fighting isoflavones than non-GM soya34

� Oilseed rape engineered to have vitamin A in its oil had much reduced vitamin E and altered oil-fat composition35

� Human volunteers fed a single GM soya bean meal showed that GM DNA can survive processing and is detectable in the digestive tract. There was evidence of horizontal gene transfer to gut bacteria36 37. Horizontal gene transfer of antibiotic resistance and Bt insecticide genes from GM foods into gut bacteria is an extremely serious issue. This is because the modified gut bacteria could become resistant to antibiotics or become factories for Bt insecticide. While Bt in its natural form has been safely used for years as an insecticide in farming, Bt toxin genetically engineered into plant crops has been found to have potential ill health effects on laboratory animals38 39 40

� In the late 1980s, a food supplement produced using GM bacteria was toxic41, initially killing 37 Americans and making more than 5,000 others seriously ill.

� Several experimental GM food products (not commercialized) were found to be harmful:

� People allergic to Brazil nuts had allergic reactions to soya beans modified with a Brazil nut gene42

� The GM process itself can cause harmful effects. GM potatoes caused toxic reactions in multiple organ systems43 44. GM peas caused a 2-fold allergic reaction � the GM protein was allergenic and stimulated an allergic reaction to other food components45. This raises the question of whether GM foods cause an increase in allergies to other substances.

Can GM Foods Help Alleviate The World Food Crisis?

The root cause of hunger is not a lack of food, but a lack of access to food. The poor have no money to buy food and increasingly, no land on which to grow it. Hunger is fundamentally a social, political, and economic problem, which GM technology cannot address.

gm crops GM-quote.jpg

Recent reports from the World Bank and the United Nations Food and Agriculture Organization have identified the biofuels boom as the main cause of the current food crisis46 47. But GM crop producers and distributors continue to promote the expansion of biofuels. This suggests that their priority is to make a profit, not to feed the world.

GM companies focus on producing cash crops for animal feed and biofuels for affluent countries, not food for people.

GM crops contribute to the expansion of industrial agriculture and the decline of the small farmer around the world. This is a serious development as there is abundant evidence that small farms are more efficient than large ones, producing more crops per hectare of land48 49 50 51 52.

Do GM Crops Increase Yield Potential?

At best, GM crops have performed no better than their non-GM counterparts, with GM soya beans giving consistently lower yields for over a decade54. Controlled comparative field trials of GM/non-GM soya suggest that 50% of the drop in yield is due to the genetic disruptive effect of the GM transformation process55. Similarly, field tests of Bt insecticide-producing maize hybrids showed that they took longer to reach maturity and produced up to 12% lower yields than their non-GM counterpart56.

A US Department of Agriculture report confirms the poor yield performance of GM crops, saying, �GE crops available for commercial use do not increase the yield potential of a variety. In fact, yield may even decrease…. Perhaps the biggest issue raised by these results is how to explain the rapid adoption of GE crops when farm financial impacts appear to be mixed or even negative57.�

The failure of GM to increase yield potential was emphasized in 2008 by the United Nations International Assessment of Agricultural Knowledge, Science and Technology for Development (IAASTD) report58. This report on the future of farming, authored by 400 scientists and backed by 58 governments, stated that yields of GM crops were �highly variable� and in some cases, �yields declined�. The report noted, �Assessment of the technology lags behind its development, information is anecdotal and contradictory, and uncertainty about possible benefits and damage is unavoidable.�

Failure To Yield

The definitive study to date on GM crops and yield is �Failure to Yield: Evaluating the Performance of Genetically Engineered Crops�. Published in 2009, the study is authored by former US EPA and Center for Food Safety scientist, Dr Doug Gurian-Sherman. It is based on published, peer-reviewed studies conducted by academic scientists and using adequate experimental controls.

In the study, Dr Gurian-Sherman distinguishes between intrinsic yield (also called potential yield), defined as the highest yield which can be achieved under ideal conditions, with operational yield, the yield achieved under normal field conditions when the farmer factors in crop reductions due to pests, drought, or other environmental stresses.

The study also distinguishes between effects on yield caused by conventional breeding methods and those caused by GM traits. It has become common for biotech companies to use conventional breeding and marker assisted breeding to produce higher-yielding crops and then finally to engineer in a gene for herbicide tolerance or insect resistance. In such cases, higher yields are not due to genetic engineering but to conventional breeding. �Failure to Yield� teases out these distinctions and analyses what contributions genetic engineering and conventional breeding make to increasing yield.

Based on studies on corn and soybeans, the two most commonly grown GM crops in the United States, the study concludes that genetically engineering herbicide-tolerant soybeans and herbicide-tolerant corn has not increased yields. Insect-resistant corn, meanwhile, has improved yields only marginally. The increase in yields for both crops over the last 13 years, the report finds, was largely due to traditional breeding or improvements in agricultural practices.

The author concludes: �commercial GE crops have made no inroads so far into raising the intrinsic or potential yield of any crop. By contrast, traditional breeding has been spectacularly successful in this regard; it can be solely credited with the intrinsic yield increases in the United States and other parts of the world that characterized the agriculture of the twentieth century.�59

Critics of the study have objected that it does not use data from developing countries. The Union of Concerned Scientists responds that there are few peer-reviewed papers evaluating the yield contribution of GM crops in developing countries � not enough to draw clear and reliable conclusions. However, the most widely grown food/feed crop in developing countries, herbicide-tolerant soybeans, offers some hints. Data from Argentina, which has grown more GM soybeans than any other developing country, suggest that yields for GM varieties are the same or lower than for conventional non-GE soybeans.60

�If we are going to make headway in combating hunger due to overpopulation and climate change, we will need to increase crop yields,� says Dr Gurian-Sherman. �Traditional breeding outperforms genetic engineering hands down.�61

If GM cannot improve intrinsic (potential) yield even in the affluent United States, where high-input, irrigated, heavily subsidized farming is the norm, it would seem irresponsible to assume that it would improve yields in the developing world, where increased food production is most needed. Initiatives promoting GM crops for the developing world are experimental and appear to be founded on expectations that are not consistent with data obtained in the West.

In the West, crop failure is often underwritten by governments, which bail out farmers with compensation. Such support systems are rare in the developing world. There, farmers may literally bet their farms and their entire livelihoods on a crop. Failure can have severe consequences.

Three GM Crops For Africa

GM sweet potato The virus-resistant sweet potato has been the ultimate GM showcase project for Africa, generating a vast amount of global media coverage. Florence Wambugu, the Monsanto trained scientist fronting the project, has been proclaimed an African heroine and the savior of millions, based on her claims about the GM sweet potato doubling output in Kenya. Forbes magazine even declared her one of a tiny handful of people around the globe who would �reinvent the future�.62 It eventually emerged, however, that the claims being made for the GM sweet potato were untrue, with field trial results showing the GM crop to be a failure.63 64

In contrast with the unproven GM sweet potato variety, a successful conventional breeding program in Uganda had produced a new high-yielding variety which is virus resistant and has �raised yields by roughly 100%�. The Ugandan project achieved success at a small cost and in just a few years. The GM sweet potato, in contrast, in over 12 years in the making, consumed funding from Monsanto, the World Bank, and USAID to the tune of $6 million.65

GM Cassava

The potential of genetic engineering to massively boost the production of cassava � one of Africa�s most important foods � by defeating a devastating virus has been heavily promoted since the mid-1990s. There has even been talk of GM solving hunger in Africa by increasing cassava yields as much as tenfold.66 But almost nothing appears to have been achieved. Even after it became clear that the GM cassava had suffered a major technical failure67, media stories continued to appear about its curing hunger in Africa.68 69 Meanwhile, conventional (non-GM) plant breeding has quietly produced virus resistant cassavas that are already making a remarkable difference in farmers� fields, even under drought conditions.70

Bt Cotton

In Makhatini, South Africa, often cited as the showcase Bt cotton project for small farmers, 100,000 hectares were planted with Bt cotton in 1998. By 2002, that had crashed to 22,500 hectares, an 80% reduction in 4 years. By 2004, 85% of farmers who used to grow Bt cotton had given up. The farmers found pest problems and no increase in yield. Those farmers who still grew the crop did so at a loss, continuing only because the South African government subsidized the project and there was a guaranteed market for the cotton.71

A study published in Crop Protection journal concluded, �cropping Bt cotton in Makhathini Flats did not generate sufficient income to expect a tangible and sustainable socioeconomic improvement due to the way the crop is currently managed. Adoption of an innovation like Bt cotton seems to pay only in an agro-system with a sufficient level of intensification.�72

How Will Climate Change Impact Agriculture?

Industrial agriculture is a major contributor to global warming, producing up to 20 per cent of greenhouse gas emissions, and some methods of increasing yield can exacerbate this negative impact. For example, crops that achieve higher intrinsic yield often need more fossil fuel based nitrogen fertilizer, some of which is converted by soil microbes into nitrous oxide, a greenhouse gas nearly 300 times more potent than carbon dioxide. Minimizing global agriculture�s future climate impact will require investment in systems of agriculture less dependent on industrial fertilizers and agroecological methods of improving soil water-holding capacity and resilience.

GM seeds are created by agrochemical companies and are heavily dependent on costly external inputs such as synthetic fertilizer, herbicides, and pesticides. It would seem risky to promote such crops in the face of climate change.

Peak Oil & Agriculture

According to some analysts, peak oil, when the maximum rate of global petroleum extraction is reached, has already arrived. This will have drastic effects on the type of agriculture we practice. GM crops are designed to be used with synthetic herbicides and fertilizers. But synthetic pesticides are made from oil and synthetic fertilizer from natural gas. Both these fossil fuels are running out fast, as are phosphates, a major ingredient of synthetic fertilizers.

Farming based on the current US GM and chemical model that depends on these fossil fuel-based inputs will become increasingly expensive and unsustainable. The statistics tell the story:

In the US food system, 10 kcal of fossil energy is required for every kcal of food consumed.73

� Approximately 7.2 quads of fossil energy are consumed in the production of crops and livestock in the U.S. each year.74 75

� Approximately 8 million kcal/ha are required to produce an average corn crop and other similar crops.76

� Two-thirds of the energy used in crop production is for fertilizers and mechanization.77

Proven technologies that can reduce the amount of fossil energy used in farming include reducing fertilizer applications, selecting farm machinery appropriate for each task, managing soil for conservation, limiting irrigation, and organic farming techniques.78

In the Rodale Institute Farming Systems Trial (FST), a comparative analysis of energy inputs conducted by Dr David Pimentel of Cornell University found that organic farming systems use just 63% of the energy required by conventional farming systems, largely because of the massive amounts of energy required to synthesize nitrogen fertilizer, followed by herbicide production.79

Studies show that the low-input organic model of farming works well in African countries. The Tigray project in Ethiopia, part-funded by the UN Food and Agriculture Organisation (FAO), compared yields from the application of compost and chemical fertilizer in farmers� fields over six years. The results showed that compost can replace chemical fertilizers and that it increased yields by more than 30 percent on average. As side-benefits to using compost, the farmers noticed that the crops had better resistance to pests and disease and that there was a reduction in �difficult weeds�.80

GM Crops & Climate Change

Climate change brings sudden, extreme, and unpredictable changes in weather. If we are to survive, the crop base needs to be as flexible, resilient and diverse as possible. GM technology offers just the opposite � a narrowing of crop diversity and an inflexible technology that requires years and millions of dollars in investment for each new variety.

Each GM crop is tailor-made to fit a particular niche. With climate change, no one knows what kind of niches will exist and where. The best way to insure against the destructive effects of climate change is to plant a wide variety of highperforming crops that are genetically diverse.

GM companies have patented plant genes that they believe are involved in tolerance to drought, heat, flooding, and salinity � but have not succeeded in using these genes to produce a single new crop with these properties. This is because these functions are highly complex and involve many different genes working together in a precisely regulated way. It is beyond existing GM technology to engineer crops with these sophisticated, delicately regulated gene networks for improved tolerance traits.

Conventional natural cross-breeding, which works holistically, is much better adapted to achieving this aim, using the many varieties of virtually every common crop that tolerate drought, heat, flooding, and salinity.

In addition, advances in plant breeding have been made using marker-assisted selection (MAS), a largely uncontroversial branch of biotechnology that can speed up the natural breeding process by identifying important genes. MAS does not involve the risks and uncertainties of genetic engineering.

The controversies that exist around MAS relate to gene patenting issues. It is important for developing countries to consider the implications of patent ownership relating to such crops.

Non-GM Successes For Niche Crops

If it is accepted that niche speciality crops may be useful in helping adaptation to climate change, there are better ways of creating them than genetic engineering. Conventional breeding and marker-assisted selection have produced many advances in breeding speciality crops, though these have garnered only a fraction of the publicity given to often speculative claims of GM miracles.

An example of such a non-GM success is the �Snorkel� rice that adapts to flooding by growing longer stems, preventing the crop from drowning.81 While genetic engineering was used as a research tool to identify the desirable genes, only conventional breeding � guided by Marker Assisted Selection � was used to generate the Snorkel rice line. Snorkel rice is entirely non-GM. This is an excellent example of how the whole range of biotechnology tools, including GM, can be used most effectively to work with the natural breeding process to develop new crops that meet the critical needs of today.

Are GM Crops Environmentally Friendly?

Two kinds of GM crops dominate the marketplace:

� Crops that resist broad-spectrum (kill-all) herbicides such as Roundup. These are claimed to enable farmers to spray herbicide less frequently to kill weeds but without killing the crop

� Crops that produce the insecticide Bt toxin. These are claimed to reduce farmers� need for chemical insecticide sprays.

Both claims require further analysis.

GM Crops & Herbicide Use

The most commonly grown herbicide-resistant GM crops are engineered to be resistant to Roundup. But the increasing use of Roundup has led to the appearance of numerous weeds resistant to this herbicide82. Roundup resistant weeds are now common and include pigweed83, ryegrass84, and marestail85. As a result, in the US, an initial drop in average herbicide use after GM crops were introduced has been followed by a large increase as farmers were forced to change their farming practices to kill weeds that had developed resistance to Roundup86 87. Farmers have increased radically the amounts of Roundup applied to their fields and are being advised to use increasingly powerful mixtures of multiple herbicides and not Roundup alone88 89.

All of these chemicals are toxic and a threat to both the farmers who apply them and the people and livestock that eat the produce. This is the case even for Roundup, which has been shown to have a range of damaging cellular effects indicating toxicity at levels similar to those found on crops engineered to be resistant to the herbicide90.

A Canadian government study in 2001 showed that after just 4-5 years of commercial growing, herbicide-resistant GM oilseed rape (canola) had cross-pollinated to create �superweeds� resistant to up to three different broadspectrum herbicides. These superweeds have become a serious problem for farmers both within91 92 and outside their fields93.

In addition, GM oilseed rape has also been found to crosspollinate with and pass on its herbicide resistant genes to related wild plants, for example, charlock and wild radish/turnip. This raises the possibility that these too may become superweeds and difficult for farmers to control94. The industry�s response has been to recommend use of higher amounts and complex mixtures of herbicides95 96 and to start developing crops resistant to additional or multiple herbicides. These developments are clearly creating a chemical treadmill that would be especially undesirable for farmers in developing countries.

Insecticide-Producing GM Crops

Bt insecticide-producing GM crops have led to resistance in pests, resulting in rising chemical applications97 98 99.

In China and India, Bt cotton was initially effective in suppressing the boll weevil. But secondary pests, especially mirids and mealy bugs, that are highly resistant to Bt toxin, soon took its place. The farmers suffered massive crop losses and had to apply costly pesticides, wiping out their profit margins100 101 102 103. Such developments are likely to be more damaging to farmers in developing countries, who cannot afford expensive inputs.

The claim that Bt GM crops reduce pesticide use is disingenuous, since Bt crops are in themselves pesticides. Prof Gilles-Eric S�ralini of the University of Caen, France states: �Bt plants, in fact, are designed to produce toxins to repel pests. Bt brinjal (eggplant/aubergine) produces a very high quantity of 16-17mg toxin per kg. They affect animals. Unfortunately, tests to ascertain their effect on humans have not been conducted.�104

GM Crops & Wildlife

Farm-scale trials sponsored by the UK government showed that the growing of herbicide-resistant GM crops (sugar beet, oilseed rape) can reduce wildlife populations105 106.

The Case Of Argentina

In Argentina, the massive conversion of agriculture to GM soya production has had disastrous effects on rural social and economic structures. It has damaged food security and caused a range of environmental problems, including the spread of herbicide-resistant weeds, soil depletion, and increased pests and diseases107 108.

GM Crops, Non-Target Insects & Organisms

Bt insecticide-producing GM crops harm non-target insect populations, including butterflies109 110 111 and beneficial pest predators112. Bt insecticide released from GM crops can also be toxic to water life113 and soil organisms114. One study reveals more negative than positive impacts on beneficial insects from GM Bt insecticide-producing crops.115

Can GM & Non-GM Crops Co-Exist?

The biotech industry argues that farmers should be able to choose to plant GM crops if they wish. It says GM and non-GM crops can peacefully �co-exist�. But experience in North America has shown that �coexistence� of GM and non-GM crops rapidly results in widespread contamination of non-GM crops.

This not only has significant agroecological effects, but also serious economic effects, damaging the ability of organic farmers to receive premiums, and blocking export markets to countries that have strict regulations regarding GM contamination.

Contamination occurs through cross-pollination, spread of GM seed by farm machinery, and inadvertent mixing during storage. The entry of GM crops into a country removes choice � everyone is gradually forced to grow GM crops or to have their non-GM crop contaminated.

Here are a few examples of GM contamination incidents:

� In 2006 GM rice grown for only one year in field trials was found to have widely contaminated the US rice supply and seed stocks116. Contaminated rice was found as far away as Africa, Europe, and Central America. In March 2007 Reuters reported that US rice export sales were down by around 20 percent from those of the previous year as a result of the GM contamination117.

� In Canada, contamination from GM oilseed rape has made it virtually impossible to cultivate organic, nonGM oilseed rape118

� US courts reversed the approval of GM alfalfa because it threatened the existence of non-GM alfalfa through cross-pollination119

� Organic maize production in Spain has dropped significantly as the acreage of GM maize production has increased, because of cross-pollination problems120

� In 2009, the Canadian flax seed export market to Europe collapsed following the discovery of widespread contamination with an unauthorized GM variety121.

� In 2007 alone, there were 39 new instances of GM contamination in 23 countries, and 216 incidents have been reported since 2005122.

Alternatives To GM

Many authoritative sources, including the IAASTD report on the future of agriculture123, have found that GM crops have little to offer global agriculture and the challenges of poverty, hunger and climate change, because better alternatives are available. These go by many names, including integrated pest management (IPM), organic, sustainable, low-input, non-chemical pest management (NPM) and agroecological farming, but extend beyond the boundaries of any particular category. Projects employing these sustainable strategies in the developing world have produced dramatic increases in yields and food security124 125 126 127 128 129.

Strategies employed include:

� Sustainable, low-input, energy-saving practices that conserve and build soil, conserve water, and enhance natural pest resistance and resilience in crops

� Innovative farming methods that minimize or eliminate costly chemical pesticides and fertilizers

� Use of thousands of traditional varieties of each major food crop, which are naturally adapted to stresses such as drought, heat, harsh weather conditions, flooding, salinity, poor soil, and pests and diseases130

� Use of existing crops and their wild relatives in traditional breeding programs to develop varieties with useful traits

� Programs that enable farmers to cooperatively preserve and improve traditional seeds

� Use of beneficial and holistic aspects of modern biotechnology, such as Marker Assisted Selection (MAS), which uses the latest genetic knowledge to speed up traditional breeding131. Unlike GM technology, MAS can safely produce new varieties of crops with valuable, genetically complex properties such as enhanced nutrition, taste, yield potential, resistance to pests and diseases, and tolerance to drought, heat, salinity, and flooding132.

Organic & Low-Input Methods Improve Yields In Africa

There seems little reason to gamble with the livelihoods of poor farmers by persuading them to grow experimental GM crops when tried-and-tested, inexpensive methods of increasing food production are readily available. Several recent studies have shown that low-input methods such as organic can dramatically improve yields in African countries, along with other benefits. Such methods have the advantage of being knowledge-based rather than costly input-based. As a result they are more accessible to poor farmers than the more expensive technologies (which often have not helped in the past).

A 2008 United Nations report, �Organic Agriculture and Food Security in Africa�, looked at 114 farming projects in 24 African countries and found that organic or near-organic practices resulted in a yield increase of more than 100 percent. In East Africa, a yield increase of 128 percent was found.133 The Foreword to the study states: �The evidence presented in this study supports the argument that organic agriculture can be more conducive to food security in Africa than most conventional production systems, and that it is more likely to be sustainable in the long term.�134

Organic & Low-Input Methods Improve Farmer Incomes In Developing Countries

Poverty is a major contributory factor to food insecurity. According to the 2008 United Nations report, �Organic Agriculture and Food Security in Africa�, organic farming has a positive impact on poverty in a variety of ways. Farmers benefit from:

� cash savings, as organic farming does not require costly pesticides and fertilizers;

� extra incomes gained by selling the surplus produce (resulting from the change to organic);

� premium prices for certified organic produce, obtained primarily in Africa for export but also for domestic markets; and

� added value to organic products through processing activities.

These findings are backed up by studies from Asia and Latin America that concluded that organic farming can reduce poverty in an environmentally friendly way.135

A recent study found that certified organic farms involved in production for export were significantly more profitable than those involved in conventional production (in terms of net farm income earnings).136 Of these cases, 87 per cent showed increases in farmer and household incomes as a result of becoming organic, which contributed to reducing poverty levels and to increasing regional food security.

Who Owns The Technology?

In considering which agricultural technologies will most benefit the developing world, it is crucial to ask who owns those technologies. The �Gene Revolution� that is proposed for Africa will be rolled out via public-private partnerships. The public side of such partnerships will be provided by Africa, whereas the private side will be provided by biotechnology companies based in the United States and Europe.

The transgenes used in creating GM crops are patented and owned by biotech companies. In the United States and Canada, companies have launched lawsuits against farmers whose crops were alleged to contain a company�s patented GM genes. Farmers� claims that they have not intentionally planted GM crops have proved no defense in court against large fines being imposed.

When farmers buy GM seed, they sign a technology agreement promising not to save and replant seed. They have to buy new seed each year from the biotech company, thus transferring control of food production from farmers to seed companies. Consolidation of the seed industry increasingly means that farmers have little choice but to buy GM seed. Centuries of farmer knowledge that went into creating locally adapted and varied seed stocks are wiped out.

In contrast, low-input and organic farming methods do not involve patented technologies. Control of food production remains in the hands of farmers, keeping farmer skills alive and favoring food security.

Conclusion

GM crop technologies do not offer significant benefits. On the contrary, they present risks to human and animal health, the environment, farmers, food security, and export markets. There is no convincing reason to take such risks with the livelihoods of farmers when proven successful and widely acceptable alternatives are readily and cheaply available. These alternatives will maintain the independence of the food supply from foreign multinational control and offer the best insurance against the challenges of climate change.

Blank
References:

1. The Mutational Consequences of Plant Transformation. Latham
J.R. et al. J Biomed Biotech. 2006, Article ID 25376, 1-7, 2006.
2. Transformation-induced mutations in transgenic plants: Analysis
and biosafety implications. Wilson A.K. et al. Biotechnol Genet Eng
Rev., 23: 209-234, 2006.
3. Safety testing and regulation of genetically engineered foods. Freese
W and Schubert D. Biotechnol Genet Eng Rev., 21: 299-324, 2004.
4. GMO in animal nutrition: potential benefits and risks. Pusztai A.
and Bardocz S. In: Biology of Nutrition in Growing Animals, eds. R.
Mosenthin, J. Zentek and T. Zebrowska, Elsevier Limited, pp. 513-
540, 2006.
5. Assessing the survival of transgenic plant DNA in the human
gastrointestinal tract. Netherwood T. et al. Nat Biotech., 22: 204-
209, 2004.
6. Experts Weigh In: Will Trans Fat Bans Affect Obesity Trends? Meir
Stampfer. DOC News, Volume 4 (Number 5): p. 1, 1 May 2007.
7. Food related illness and death in the United States. Mead P.S. et al.
Emerging Infectious Diseases, 5: 607-625, 1999.
8. Food Safety – Contaminants and Toxins. Unpublished study
reviewed in J.P.F. D�Mello, CABI Publishing, 2003.
9. Fine structural analysis of pancreatic acinar cell nuclei from mice fed
on GM soybean. Malatesta M. et al. Eur J Histochem., 47: 385-388, 2003.
10. Ultrastructural morphometrical and immunocytochemical
analyses of hepatocyte nuclei from mice fed on genetically modified
soybean. Malatesta M et al. Cell Struct Funct., 27: 173-180, 2002.
11. Ultrastructural analysis of testes from mice fed on genetically
modified soybean. Vecchio L. et al. Eur J Histochem., 48: 448-454, 2004.
12. Transgenic expression of bean alpha-amylase inhibitor in peas
results in altered structure and immunogenicity. Prescott V.E. et al. J
Agric Food Chem., 53: 9023-9030, 2005.
13. Biotechnology Consultation Note to the File BNF No 00077.
Office of Food Additive Safety, Center for Food Safety and Applied
Nutrition, US Food and Drug Administration, 4 September 2002.
14. GMO in animal nutrition: potential benefits and risks. Pusztai
A. and Bardocz S. In: Biology of Nutrition in Growing Animals, eds.
R. Mosenthin, J. Zentek and T. Zebrowska, Elsevier Limited, pp. 513-
540, 2006.
15. Effects of diets containing genetically modified potatoes
expressing Galanthus nivalis lectin on rat small intestine. Ewen S.W.
and Pusztai A. The Lancet, 354: 1353-1354, 1999.
16. New analysis of a rat feeding study with a genetically modified
maize reveals signs of hepatorenal toxicity. S�ralini, G.-E. et al. Arch.
Environ Contam Toxicol., 52: 596-602, 2007.
17. A three generation study with genetically modified Bt corn in
rats: Biochemical and histopathological investigation. Kilic A and
Akay MT. Food and Chemical Toxicology, 46: 1164-1170, 2008.
18. Intestinal and Peripheral Immune Response to MON810 Maize
Ingestion in Weaning and Old Mice. Finamore A et al. J. Agric. Food
Chem., 56: 11533-11539, 2008.
19. Biological effects of transgenic maize NK603xMON810 fed
in long term reproduction studies in mice. Velimirov A et al.
Bundesministerium f�r Gesundheit, Familie und Jugend Report,
Forschungsberichte der Sektion IV Band 3/2008, Austria, 2008.
bmgfj.cms.apa.at/cms/site/attachments/3/2/9/ CH0810/
CMS1226492832306/forschungsbericht_3-2008_letztfassung.pdf
20. A long-term study on female mice fed on a genetically modified
soybean: effects on liver ageing. Malatesta M. et al. Histochem Cell
Biol., 130: 967-977, 2008.
21. Genetically modified soya bean in rabbit feeding: detection of
DNA fragments and evaluation of metabolic effects by enzymatic
analysis. R. Tudisco et al. Animal Science, 82: 193-199, 2006.
22. A three-year longitudinal study on the effects of a diet
containing genetically modified Bt176 maize on the health status
and performance of sheep. Trabalza-Marinucci M. et al. Livestock
Science, 113: 178-190, 2008.
23. Fate of genetically modified maize DNA in the oral cavity and
rumen of sheep. Duggan P.S. et al. Br J Nutr., 89: 159-166, 2003.
24. Detection of genetically modified DNA sequences in milk from the
Italian market. Agodi A. et al. Int J Hyg Environ Health, 209: 81-88, 2006.25. Assessing the transfer of genetically modified DNA from feed to
animal tissues. Mazza R. et al. Transgenic Res., 14: 775-784, 2005.
26. Detection of Transgenic and Endogenous Plant DNA in Digesta
and Tissues of Sheep and Pigs Fed Roundup Ready Canola Meal.
Mazza R. et al. J Agric Food Chem. 54: 1699-1709, 2006.
27. How Subchronic and Chronic Health Effects can be Neglected
for GMOs, Pesticides or Chemicals. S�ralini, G-E, et al. International
Journal of Biological Sciences, 2009; 5(5):438-443.
28. How Subchronic and Chronic Health Effects can be Neglected
for GMOs, Pesticides or Chemicals. S�ralini, G-E, et al. International
Journal of Biological Sciences, 2009; 5(5):438-443.
29. Under wraps � Are the crop industry�s strong-arm tactics and
close-fisted attitude to sharing seeds holding back independent
research and undermining public acceptance of transgenic crops?
Waltz, E., Nature Biotechnology, Vol. 27, No. 10, October 2009.
30. Crop Scientists Say Biotechnology Seed Companies Are
Thwarting Research. Pollack, A., New York Times, 20 February 2009.
31. The Genetic Engineering of Food and the Failure of Science �
Part 1: The Development of a Flawed Enterprise. Lotter, D., Int. Jrnl.
of Soc. of Agr. & Food, Vol. 16, No. 1, 2007, pp. 31�49.
32. The Genetic Engineering of Food and the Failure of Science
� Part 2: Academic Capitalism and the Loss of Scientific Integrity.
Lotter, D., Int. Jrnl. of Soc. of Agr. & Food, Vol. 16, No. 1, 2008, pp.
50�68.
33. Biotech proponents aggressively attack independent research
papers: GM crops: Battlefield. Waltz, E., Nature 461, 2009, 27�32.
34. Alterations in clinically important phytoestrogens in genetically
modified, herbicide-tolerant soybeans. Lappe M.A. et al. J Med Food,
1: 241-245, 1999.
35. Seed-specific overexpression of phytoene synthase: increase in
carotenoids and other metabolic effects. Shewmaker CK et al. Plant
J, 20: 401-412, 1999.
36. Assessing the survival of transgenic plant DNA in the human
gastrointestinal tract. Netherwood T. et al. Nat Biotech., 22: 204-
209, 2004.
37. The fate of transgenes in the human gut. Heritage J. Nat Biotech.,
22: 170-172, 2004.
38. Bacillus thuringiensis Cry1Ac Protoxin is a Potent Systemic and
Mucosal Adjuvant. V�zquez RI et al. Scand J Immunol., 49: 578-584,
1999.
39. Intragastric and intraperitoneal administration of Cry1Ac
protoxin from Bacillus thuringiensis induces systemic and mucosal
antibody responses in mice. V�zquez-Padr�n, RI et al. Life Sci., 64:
1897-1912, 1999.
40. Cry1Ac Protoxin from Bacillus thuringiensis sp. kurstaki HD73
Binds to Surface Proteins in the Mouse Small Intestine. V�zquezPadr�n,
RI et al. Biochem Biophys Res Comm., 271: 54-58, 2000.
41. Eosinophilia-myalgia syndrome and tryptophan production: a
cautionary tale. Mayeno A.N and Gleich G.J. Tibtech, 12: 346-352,
1994.
42. Identification of a Brazil-nut allergen in transgenic soybeans.
Nordlee J.E. et al. N England J Med., 334: 688-692, 1996.
43. GMO in animal nutrition: potential benefits and risks. Pusztai
A. and Bardocz S. In: Biology of Nutrition in Growing Animals, eds.
R. Mosenthin, J. Zentek and T. Zebrowska, Elsevier Limited, pp. 513-
540, 2006.
44. Effects of diets containing genetically modified potatoes
expressing Galanthus nivalis lectin on rat small intestine. Ewen S.W.
and Pusztai A. The Lancet, 354: 1353-1354, 1999.
45. Transgenic expression of bean alpha-amylase inhibitor in peas
results in altered structure and immunogenicity. Prescott V.E. et al. J
Agric Food Chem., 53: 9023-9030, 2005.
46. A Note on Rising Food Prices. Donald Mitchell. World Bank
report, 2008. image.guardian.co.uk/sys-files/Environment/
documents/2008/07/10/Biofuels.PDF
47. Soaring Food Prices: Facts, Perspectives, Impacts and Actions
Required. United Nations Food and Agriculture Organisation
conference and report, Rome, 3-5 June 2008. www.fao.org/
fileadmin/user_upload/foodclimate/HLCdocs/HLC08-inf-1-E.pdf
48. Small Is Beautiful: Evidence of Inverse Size Yield Relationship
in Rural Turkey. �nal, FG. The Levy Economics Institute of Bard
College, October 2006, updated December 2008. www.levy.
org/pubs/wp_551.pdf.
49. Farm Size, Land Yields and the Agricultural Production function:
An Analysis for Fifteen Developing Countries. Cornia, G. World
Development, 13: 513-34, 1985.
50. Rural market imperfections and the farm size-productivity
relationship: Evidence from Pakistan. Heltberg, R. World
Development 26: 1807-1826, 1998.
51. Is there a future for small farms? Hazell, P. Agricultural
Economics, 32: 93-101, 2005.
52. Is Small Beautiful? Farm Size, Productivity and Poverty in Asian
Agriculture. Fan S and Chan-Kang C. Agricultural Economics, 32:
135-146, 2005.
53. Hope for Africa lies in political reforms. Daniel Howden, Africa
correspondent, The Independent (London), 8 September 2008,
www.independent.co.uk/opinion/commentators/danielhowden-hope-for-africa-lies-in-political-reforms-922487.html
54. Evidence of the Magnitude and Consequences of the Roundup
Ready Soybean Yield Drag from University-Based Varietal Trials in
1998. Benbrook C. Benbrook Consulting Services Sandpoint, Idaho.
Ag BioTech InfoNet Technical Paper, Number 1, 13 Jul 1999. http://
www.mindfully.org/GE/RRS-Yield-Drag.htm
55. Glyphosate-resistant soyabean cultivar yields compared with
sister lines. Elmore R.W. et al. Agronomy Journal, 93: 408-412, 2001.
56. Development, yield, grain moisture and nitrogen uptake of
Bt corn hybrids and their conventional near-isolines. Ma B.L. and
Subedi K.D. Field Crops Research, 93: 199-211, 2005.
57. The Adoption of Bioengineered Crops. US Department of
Agriculture Report, May 2002, www.ers.usda.gov/publications/
aer810/aer810.pdf.
58. International Assessment of Agricultural Knowledge, Science and
Technology for Development: Global Summary for Decision Makers
(IAASTD); Beintema, N. et al., 2008. www.agassessment.org/
index.cfm?Page=IAASTD%20Reports&ItemID=2713
59. Failure to Yield: Evaluating the Performance of Genetically
Engineered Crops. Doug Gurian-Sherman. Union of Concerned
Scientists, April 2009, p. 13
60. Roundup ready Soybeans in Argentina: farm level and aggregate
welfare effects. Qaim, M. and G. Traxler. 2005. Agricultural Economics
32: 73�86.
61. Doug Gurian-Sherman, quoted on Union of Concerned
Scientists website, www.ucsusa.org/food_and_agriculture/
science_and_impacts/science/failure-to-yield.html.
62. Millions served. Lynn J. Cook. Forbes magazine, 23 December
2002.
63. GM technology fails local potatoes. Gatonye Gathura. The Daily
Nation (Kenya), 29 January 2004.
64. Monsanto�s showcase project in Africa fails. New Scientist, Vol.
181, No. 2433, 7 February 2004.
65. Genetically modified crops and sustainable poverty alleviation
in sub-Saharan Africa: An assessment of current evidence. Aaron
deGrassi. Third World Network-Africa, June 2003.66. Plant Researchers Offer Bumper Crop of Humanity. Martha
Groves. LA Times, 26 December 1997.
67. Danforth Center cassava viral resistance update. Donald
Danforth Plant Science Center, 30 June 2006.
68. Can biotech from St. Louis solve hunger in Africa? Kurt
Greenbaum. St. Louis Post-Dispatch, 9 December 2006.
69. St. Louis team fights crop killer in Africa. Eric Hand. St. Louis
Post-Dispatch, 10 December 2006.
70. Farmers get better yields from new drought-tolerant cassava.
IITA, 3 November 2008; Cassava�s comeback. United Nations Food
and Agriculture Organisation, 13 November 2008.
71. A Disaster in Search of Success: Bt Cotton in Global South. Film
by Community Media Trust, Pastapur, and Deccan Development
Society, Hyderabad, India, February 2007.
72. Impact of Bt cotton adoption on pesticide use by smallholders:
A 2-year survey in Makhatini Flats (South Africa). Hofs, J-L, et al.
Crop Protection, Volume 25, Issue 9, September 2006, pp. 984�988.
73. Food, energy and society. Pimentel, D., and M. Pimentel. Niwot:
Colorado University Press, 1996. Cited in Energy efficiency and
conservation for individual Americans. D. Pimentel, Environ Dev
Sustain, 1996.
74. Energy and economic inputs in crop production: Comparison
of developed, developing countries. Pimentel, D., Doughty, R.,
Carothers, C., Lamberson, S., Bora, N., & Lee, K. In L. Lal, D. Hansen,
N. Uphoff, & S. Slack (Eds.), Food security & environmental quality in
the developing world (pp. 129�151). Boca Raton: CRC Press, 2002.
75. U.S. energy conservation and efficiency: Benefits and costs.
Pimentel, D., Pleasant, A., Barron, J., Gaudioso, J., Pollock, N., Chae,
E., Kim, Y., Lassiter, A., Schiavoni, C., Jackson, A., Lee, M., & Eaton, A.
Environment Development and Sustainability, 6, 279�305, 2004.
76. Ethanol production using corn, switchgrass, and wood; and
biodiesel production using soybean and sunflower. Pimentel, D., &
Patzek, T. Natural Resources Research, 14(1), 65�76, 2005.
77. Energy and economic inputs in crop production: Comparison
of developed, developing countries. Pimentel, D., Doughty, R.,
Carothers, C., Lamberson, S., Bora, N., & Lee, K. In L. Lal, D. Hansen,
N. Uphoff, & S. Slack (Eds.), Food security & environmental quality in
the developing world (pp. 129�151). Boca Raton: CRC Press, 2002.
78. Energy efficiency and conservation for individual Americans. D.
Pimentel et al. Environ Dev Sustain., Vol. 11, No. 3, June 2009.
79. Environmental, Energetic, and Economic Comparisons of
Organic and Conventional Farming Systems. Pimentel, D. et al.
Bioscience, Vol. 55, No. 7, July 2005, pp. 573�582, www.bioone.
org/doi/full/10.1641/0006-3568(2005)055%5B0573%3AEEAECO%5
D2.0.CO%3B2#references
80. The impact of compost use on crop yields in Tigray,
Ethiopia. Institute for Sustainable Development (ISD). Edwards,
S. Proceedings of the International Conference on Organic
Agriculture and Food Security. FAO, Rom, 2007, ftp.fao.org/paia/
organicag/ofs/02-Edwards.pdf.
81. The ethylene response factors SNORKEL1 and SNORKEL2
allow rice to adapt to deep water. Hattori, Y. et al. Nature, Vol 460,
20 August 2009: 1026�1030.
82. Glyphosate-Resistant Weeds: Current Status and Future
Outlook. Nandula V.K. et al. Outlooks on Pest Management, August
2005: 183�187.
83. Syngenta module helps manage glyphosate-resistant weeds.
Delta Farm Press, 30 May 2008, deltafarmpress.com/mag/
farming_syngenta_module_helps/index.html.
84. Resistant ryegrass populations rise in Mississippi. Robinson R.
Delta Farm Press, Oct 30, 2008. deltafarmpress.com/wheat/
resistant-ryegrass-1030/
85. Glyphosate Resistant Horseweed (Marestail) Found in 9 More
Indiana Counties. Johnson B and Vince Davis V. Pest & Crop, 13
May 2005. extension.entm.purdue.edu/pestcrop/2005/issue8/
index.html
86. Genetically Engineered Crops and Pesticide Use in the United
States: The First Nine Years. Benbrook CM. BioTech InfoNet
Technical Paper Number 7, October 2004. www.biotech-info.
net/Full_version_first_nine.pdf
87. Agricultural Pesticide Use in US Agriculture. Center for Food
Safety, May 2008, www.centerforfoodsafety.org/pubs/USDA%20
NASS%20Backgrounder-FINAL.pdf.
88. A Little Burndown Madness. Nice G et al. Pest & Crop, 7 Mar
2008. extension.entm.purdue.edu/pestcrop/2008/issue1/
index.html
89. To slow the spread of glyphosate resistant marestail, always
apply with 2,4-D. Pest & Crop, issue 23, 2006. extension.entm.
purdue.edu/pestcrop/2006/issue23/table1.html
90. Glyphosate Formulations Induce Apoptosis and Necrosis in
Human Umbilical, Embryonic, and Placental Cells. Benachour, N. and
Gilles-Eric S�ralini. Chem. Res. Toxicol., 2009, 22 (1), pp 97�105.
91. Genetically-modified superweeds �not uncommon�. Randerson
J. New Scientist, 05 February 2002. www.newscientist.com/
article/dn1882-geneticallymodified-superweeds-not-uncommon.html
92. Elements of Precaution: Recommendations for the Regulation
of Food Biotechnology in Canada. An Expert Panel Report on the
Future of Food Biotechnology prepared by The Royal Society of
Canada at the request of Health Canada Canadian Food Inspection
Agency and Environment Canada, 2001. www.rsc.ca//files/
publications/ expert_panels/foodbiotechnology/GMreportEN.pdf
93. Gene Flow and Multiple Herbicide Resistance in Escaped Canola
Populations. Knispel AL et al. Weed Science, 56: 72-80, 2008.
94. Do escaped transgenes persist in nature? The case of an
herbicide resistance transgene in a weedy Brassica rapa population.
Warwick SI et al. Molecular Ecology, 17: 1387-1395, 2008.
95. A Little Burndown Madness. Nice G et al. Pest & Crop, 7 Mar
2008. extension.entm.purdue.edu/pestcrop/2008/issue1/
index.html
96. To slow the spread of glyphosate resistant marestail, always
apply with 2,4-D. Pest & Crop, issue 23, 2006. extension.entm.
purdue.edu/pestcrop/2006/issue23/table1.html
97. First report of field resistance by the stem borer, Busseola fusca
(Fuller) to Bt-transgenic maize. Rensburg J.B.J. S. Afr J Plant Soil., 24:
147-151, 2007.
98. Resistance of sugarcane borer to Bacillus thuringiensis Cry1Ab
toxin. Huang F et al. Entomologia Experimentalis et Applicata 124:
117-123, 2007.
99. Insect resistance to Bt crops: evidence versus theory. Tabashnik
BE et al. Nat Biotech., 26: 199-202, 2008.
100. Transgenic cotton drives insect boom. Pearson H. NatureNews.
Published online 25 July 2006. www.nature.com/
news/2006/060724/full/news060724-5.html
101. Bt-cotton and secondary pests. Wang S et al. Int. J.
Biotechnology, 10: 113-121, 2008.
102. India: Bt cotton devastated by secondary pests. Bhaskar
Goswami. Grain, 01 Sept 2007. www.grain.org/
btcotton/?id=398
103. Bt cotton not pest resistant. Gur Kirpal Singh Ashk. The Times
of India, 24 Aug 2007, timesofindia.indiatimes.com/Chandigarh/
Bt_cotton_not_pest_resistant/articleshow/2305806.cms
104. Prof Gilles-Eric S�ralini, in an interview with Savvy Soumya
Misra, Down to Earth, 15 April 2009, downtoearth.org.in/full6.
asp?foldername=20091031&filename=inv&sec_id=14&sid=1105. Transgenic crops take another knock. Giles J. NatureNews,
published online: 21 March 2005. www.nature.com/
news/2005/050321/full/050321-2.html
106. Effects on weed and invertebrate abundance and diversity of
herbicide management in genetically modified herbicide-tolerant
winter-sown oilseed rape. Bohan DA et al. Proc R Soc B, 272: 463-
474, 2005.
107. Argentina�s bitter harvest. Branford S. New Scientist, 17 April
2004.
108. Rust, resistance, run down soils, and rising costs � Problems
facing soybean producers in Argentina. Benbrook C.M. AgBioTech
InfoNet, Technical Paper No 8, Jan 2005.
109. Transgenic pollen harms monarch larvae. Losey J.E. et al.
Nature, 399: 214, 1999.
110. Field deposition of Bt transgenic corn pollen: lethal effects on
the monarch butterfly. Hansen L. C. and J. Obrycki J. Oecologia, 125:
241-245, 2000.
111. The effects of pollen consumption of transgenic Bt maize
on the common swallowtail, Papilio machaon L. (Lepidoptera,
Papilionidae). Lang A and Vojtech E. Basic and Applied Ecology, 7:
296-306, 2006.
112. A meta-analysis of effects of Bt cotton and maize on nontarget
invertebrates. Marvier M. et al. Science, 316: 1475-1477, 2007.
113. Toxins in transgenic crop byproducts may affect headwater
stream ecosystems. Rosi-Marshall E.J. et al. Proc. Natl. Acad. Sci.
USA, 104: 16204-16208, 2007.
114. Impact of Bt Corn on Rhizospheric and Soil Eubacterial
Communities and on Beneficial Mycorrhizal Symbiosis in
Experimental Microcosms. M. Castaldini M. et al. Appl Environ
Microbiol., 71: 6719-6729, 2005.
115. The impact of transgenic plants on natural enemies: a critical
review of laboratory studies. L�vei, G.L. and S. Arpaia, 2004.
Entomologia Experimentalis et Applicata vol. 114: 1�14.
116. Risky business: Economic and regulatory impacts from the
unintended release of genetically engineered rice varieties into the
rice merchandising system of the US. Report for Greenpeace, 2007.
117. Mexico Halts US Rice Over GMO Certification. Reuters, 16
March 2007.
118. Organic farmers seek Supreme Court hearing. Press release,
Organic Agriculture Protection Fund Committee, Saskatoon,
Canada, 1 August 2007.
119. The United States District Court for the Northern District
of California. Case 3:06-cv-01075-CRB Document 199 Filed
05/03/2007: Memorandum and Order Re: Permanent Injunction.
120. Coexistence of plants and coexistence of farmers: Is an
individual choice possible? Binimelis, R., Journal of Agricultural and
Environmental Ethics, 21: 437-457, 2008.
121. CDC Triffid Flax Scare Threatens Access To No. 1 EU Market.
Allan Dawson. Manitoba Co-operator, 17 September 2009; Changes
Likely For Flax Industry. Allan Dawson. Manitoba Cooperator, 24
September 2009.
122. Biotech companies fuel GM contamination spread. Greenpeace
International, 29 February 2008. www.greenpeace.org/
international/news/gm-ge-contamination-report290208
123. International Assessment of Agricultural Knowledge,
Science and Technology for Development: Global Summary
for Decision Makers (IAASTD); Beintema, N. et al., 2008.
www.agassessment.org/index.cfm?Page=IAASTD%20
Reports&ItemID=2713
124. Applying Agroecology to Enhance the Productivity of Peasant
Farming Systems in Latin America. Altieri M.A. Environment,
Development and Sustainability, 1: 197-217, 1999.
125. More Productivity with Fewer External Inputs: Central
American Case Studies of Agroecological Development and their
Broader Implications. Bunch R. Environment, Development and
Sustainability, 1: 219-233, 1999.
126. Can Sustainable Agriculture Feed Africa? New Evidence
on Progress, Processes and Impacts. Pretty J. Environment,
Development and Sustainability, 1: 253-274, 1999.
127. Organic Agriculture and Food Security in Africa. United
Nations Conference on Trade and Development, United Nations
Environment Programme, 2008. www.unep-unctad.org/cbtf/
publications/UNCTAD_DITC_TED_2007_15.pdf
128. Ecologising rice-based systems in Bangladesh. Barzman M. &
Das L. ILEIA Newsletter, 2: 16-17, 2000. www.leisa.info/index.
php?url=magazine-details.tpl&p[_id]=12434
129. Genetic diversity and disease control in rice. Zhu Y et al.
Nature, 406: 718-722, 2000.
130. Lost Crops of Africa, Vol.1: Grains. National Research
Council (Washington DC, USA) Report, 1996. www7.
nationalacademies.org/dsc/LostCropsGrains_Brief.pdf
131. Marker-assisted selection: an approach for precision plant
breeding in the twenty-first century. Collard BCY and Mackill DJ.
Phil Trans R Soc B, 363: 557-572, 2008.
132. Breeding for abiotic stresses for sustainable agriculture.
Witcombe J.R. et al. Phil Trans R Soc B, 363: 703-716, 2008.
133. �Organic Agriculture and Food Security in Africa�. Foreword by
Supachai Panitchpakdi, Secretary-General of UNCTAD, and Achim
Steiner, Executive Director of UNEP. United Nations Environment
Programme (UNEP) and United Nations Conference on Trade and
Development (UNCTAD), 2008, p. 16, www.unep-unctad.org/
cbtf/publications/UNCTAD_DITC_TED_2007_15.pdf
134. �Organic Agriculture and Food Security in Africa�. Foreword by
Supachai Panitchpakdi, Secretary-General of UNCTAD, and Achim
Steiner, Executive Director of UNEP. United Nations Environment
Programme (UNEP) and United Nations Conference on Trade and
Development (UNCTAD), 2008, www.unep-unctad.org/cbtf/
publications/UNCTAD_DITC_TED_2007_15.pdf
135. Certified organic export production. Implications for economic
welfare and gender equity among smallholder farmers in tropical
Africa. UNCTAD. 2008, www.unctad.org/trade_env/test1/
publications/UNCTAD_DITC_TED_2007_7.pdf; The economics
of certified organic farming in tropical Africa: A preliminary
analysis. Gibbon P and Bolwig S. 2007. SIDA DIIS Working Paper
no 2007/3, Subseries on Standards and Agro-Food-Exports (SAFE)
No. 7; Organic Agriculture: A Trade and Sustainable Development
Opportunity for Developing Countries. Twarog. 2006. In UNCTAD.
2006. Trade and Environment Review, UN, 2006, www.unctad.
org/en/docs/ditcted200512_en.pdf.
136. The economics of certified organic farming in tropical Africa: A
preliminary analysis. Gibbon P and Bolwig S. 2007. SIDA DIIS Working
Paper no 2007/3, Subseries on Standards and Agro-Food-Exports
(SAFE) No. 7; Certified organic export production. Implications for
economic welfare and gender equity among smallholder farmers in
tropical Africa. UNCTAD. 2008, www.unctad.org/trade_env/
test1/publications/UNCTAD_DITC_TED_2007_7.pdf.

Close Accordion
Hypertension Associated with Sodium Loss | Wellness Clinic

Hypertension Associated with Sodium Loss | Wellness Clinic

Cardiovascular disease and hypertension can often occur due to a variety of factors, however, an improper diet and nutrition has been appointed to be one of the most prevalent causes behind the onset of cardiovascular disease and hypertension. While an improper diet and nutrition can lead to these issues, a balanced, healthy diet and nutrition can help prevent cardiovascular disease and hypertension, even treat the existing conditions.

 

What’s the best dietary and nutritional approach to treat cardiovascular disease and hypertension?

 

Many of the substances in food, particularly in nutraceutical supplements, antioxidants, vitamins or nutritional supplements, function in a manner that is similar to a category of drugs and medications to improve cardiovascular disease and hypertension. The effect is synergistic when used in combination with other nutritional supplements even though the drug might not be less than the potency of the compounds. These natural compounds have been outlined to the significant antihypertensive drug classes, such as diuretics, beta blockers, central alpha agonists, direct vasodilators, calcium channel blockers (CCB’s), angiotensin converting enzyme inhibitors (ACEI’s), angiotensin receptor blockers (ARB’s) and direct renin inhibitors (DRI).

 

Dietary Approaches to Stop Hypertension

 

The Dietary Approaches to Stop Hypertension (DASH) Iand II diets conclusively demonstrated significant reductions in BP in borderline and stageIhypertensive patients. Back in DASH I, untreated hypertensive subjects with SBP < 160 mmHg and DBP 80-95 mmHg were placed on one of three diets for 4 weeks, controled diet, vegetable and fruit diet (F + V) and combined diet, which added F + V and low fat milk. Sodium restriction was added by DASH II in every group. The control diet consisted of sodium at 3 g/d, potassium, calcium and magnesium in 25% of the US average, macronutrients at US average of 4 portions every day, a sodium/potassium ratio of 1.7 and fiber at 9 g/d. The F + V diet raised the potassium, calcium and magnesium to 75 percent, macronutrients compared to the US average, a sodium potassium ratio of 0.7, 31 gram of fiber and 8.5 portions of vegetables and fruits per day. The joint diet was similar to the F + V diet but additional fat milk. At 2 wk the BP was decreased by 10.7/5.2 mmHg from the hypertensive patients in DASHIand 11.5/6.8 mmHg from the hypertensive patients in DASH II. These reductions persisted provided that the patients were on the diet. The DASH diet increases plasma renin activity (PRA) and serum aldosterone levels in response to this BP reductions) The increase in PRA was 37 ng/mL every day. There has been an associated of reaction with the polymorphism of beta 2 adrenergic receptor. The A allele of G46A had blunted PRA and a higher BP reduction and aldosterone. The arachidonic acid (AA) genotype had the best response along with the GG genotype had no response. Adding an ARB, DRI or ACEI improved BP reaction due to blockade of the increase in PRA to the DASH diet at the GG group. A very low sodium DASH diet reduces oxidative stress (urine F2-isoprostanes), enhances vascular function (enhancement indicator) and lowers BP in salt sensitive areas. In addition, plasma nitrite increased and pulse wave velocity decreased on the DASH diet at week two.

 

Sodium (Na+) Loss and Hypertension

 

The average sodium intake in the US is 5000 mg/d with some regions of the nation consuming 15000-20000 mg/d. On the other hand, the minimal requirement for sodium is most likely roughly 500 mg/d. Epidemiologic, observational and controlled clinical trials reveal that an increased sodium intake is associated with increased risk for proteinuria, renal insufficiency, CVD LVH, CHD, MI and of the SNS as well as BP. A decrease in sodium intake in hypertensive patients the salt sensitive patients, will significantly lower BP by 4-6/2-3 mmHg that’s proportional to the level of sodium restriction and might stop or delay hypertension in high risk patients and decrease CV events.

 

Salt sensitivity (? 10% increase in MAP with salt loading) occurs in about 51 percent of hypertensive patients and is a vital variable in determining the cardiovascular, cerebrovascular, renal and BP responses to dietary salt intake. Cardiovascular events are prevalent in the salt patients than ones, independent of BP. An increased sodium intake has a direct positive correlation with BP and the risk of CHD and CVA. The risk is independent of BP to get CVA with a relative risk of 1.04 to 1.25 in the lowest to the highest quartile. In addition, patients may convert into a BP routine with increases in nocturnal BP as the sodium intake increases.

 

Increased sodium intake has a direct impact on endothelial cells. Sodium promotes cutaneous lymphangiogenesis, increases endothelial cell stiffness, reduces dimensions, surface area, volume, cytoskeleton, deformability and pliability, reduces eNOS and NO generation, raises asymmetric dimethyl arginine (ADMA), oxidative stress and TGF-?. Every one of these vascular responses are increased in the presence of aldosterone. These modifications occur independent of BP and may be partially counteract by potassium. The endothelial cells act as vascular salt sensors. Endothelial cells are targets for aldosterone which activate epithelial sodium channels (ENaCs) and also have a negative effects on discharge of NO and on endothelial function. The mechanical stimulation of the cell plasma membrane along with the submembranous actin network (endothelial glcyocalyx) (“shell”) serve as a “firewall” to protect the endothelial cells and are regulated by serum sodium, potassium and aldosterone within the physiological selection. Changes in shear-stress-dependent activity of the endothelial NO synthase located in the caveolae regulate the viscosity in this “shell”. High plasma sodium gelates the casing of the cell, whereas the casing is fluidized by high fructose. These communications between intracellular enzymes and extracellular ions happen in the plasma membrane barrier, whereas 90 percent of the cell mass remains uninvolved in such changes. Blockade of the ENaC using spironolactone (100%) or amiloride (84%) minimizes or prevent many of these vascular endothelial responses and boost NO. Nitric oxide release follows not vice versa and nanomechanics and decreases vascular endothelial cell stiffness which enhances circulation conducive vasodilation that is dependent. In the presence of HS-CRP that was increased and vascular inflammation, the effects of aldosterone on the ENaC is enhanced further raising vascular stiffness and BP. High sodium intake also abolishes the AT2R-mediated vasodilation immediately with complete abolition of endothelial vasodilation (EDV) within 30 d. Thus, it is now clear that high dietary sodium has adverse effects on the circulatory system, BP and CVD by changing the endothelial glycocalyx, which is a negatively charged biopolymer that lines the blood vessels and also serves as a protective barrier against sodium over-load, increased sodium permeability and sodium-induced TOD. Certain SNP’s of salt inducible kinaseIwhich alter Na+/K+ ATPase, determine LVH and sodium caused hypertension.

 

The sodium intake every day in patients must be between 1500. BP reduction improves in people on patients which are on treatment and the decrease in BP is additive with limitation of refined carbohydrates. Reducing sodium consumption may reduce damage to the brain, heart, kidney and vasculature through mechanisms dependent on the BP reduction that is little as well as those independent of the BP. A balance of sodium with nutrients, particularly calcium, magnesium and potassium is important, not just in reducing and controlling BP, but also in decreasing cerebrovascular and cardiovascular events. An increase in the sodium to potassium ratio is associated with risk of all-cause mortality and CVD. The Yanomamo Indians consume and excrete only 1 meq of sodium from 24 h and consume and excrete 152 meq of potassium in 24 h. BP doesn’t rise with age and is related to elevated PRA, although the Na + to K + percentage is 1/152. Currently 50 the BP in the Yanomamo is 100-108/64-69 mmHg.

 

In conclusion,�Cardiovascular disease and hypertension can often occur due to a variety of factors, however, an improper diet and nutrition has been appointed to be one of the most prevalent causes behind the onset of cardiovascular disease and hypertension. According to the above research studies, an imbalance in the intake of sodium can lead to cardiovascular disease and hypertension.� The scope of our information is limited to chiropractic and spinal injuries and conditions. To discuss options on the subject matter, please feel free to ask Dr. Jimenez or contact us at 915-850-0900 .

Green-Call-Now-Button-24H-150x150-2-3.png

By Dr. Alex Jimenez

 

Additional Topics: Wellness

 

Overall health and wellness are essential towards maintaining the proper mental and physical balance in the body. From eating a balanced nutrition as well as exercising and participating in physical activities, to sleeping a healthy amount of time on a regular basis, following the best health and wellness tips can ultimately help maintain overall well-being. Eating plenty of fruits and vegetables can go a long way towards helping people become healthy.

 

blog picture of cartoon paperboy big news

 

TRENDING TOPIC: EXTRA EXTRA: About Chiropractic

 

 

Neuroinflammation And Psychiatric Illness

Neuroinflammation And Psychiatric Illness

Neuroinflammation:

Abstract

Multiple lines of evidence support the pathogenic role of neuroinflammation in psychiatric illness. While systemic autoimmune diseases are well-documented causes of neuropsychiatric disorders, synaptic autoimmune encephalitides with psychotic symptoms often go under-recognized. Parallel to the link between psychiatric symptoms and autoimmunity in autoimmune diseases, neuroimmunological abnormalities occur in classical psychiatric disorders (for example, major depressive, bipolar, schizophrenia, and obsessive-compulsive disorders). Investigations into the pathophysiology of these conditions traditionally stressed dysregulation of the glutamatergic and monoaminergic systems, but the mechanisms causing these neurotransmitter abnormalities remained elusive. We review the link between autoimmunity and neuropsychiatric disorders, and the human and experimental evidence supporting the pathogenic role of neuroinflammation in selected classical psychiatric disorders. Understanding how psychosocial, genetic, immunological and neurotransmitter systems interact can reveal pathogenic clues and help target new preventive and symptomatic therapies.

Keywords:

  • Neuroinflammation,
  • Psychoneuroimmunology,
  • Astrocyte,
  • Microglia,
  • Cytokines,
  • Oxidative stress,
  • Depression,
  • Obsessive-compulsive disorder,
  • Bipolar disorder, Schizophrenia

Introduction

As biological abnormalities are increasingly identified among patients with psychiatric disorders, the distinction between neurological and psychiatric illness fades. In addition to systemic autoimmune diseases associated with psychiatric manifestations (for example, lupus) [1], more recently, patients with acute isolated psychosis were identified with synaptic autoimmune encephalitides (Table 1) [2-6]. These patients are often erroneously diagnosed with refractory primary psych- otic disorders, delaying initiation of effective immune therapy (Table 1). Additionally, growing evidence supports the pathogenic role of anti-neuronal antibodies in neuropsychiatric disorders [7].

neuroinflammation table-1-3.jpg

Separation of neurological and psychiatric disorders, supported by Descartes�s conception of the �mind� as an ontologically distinct entity and by the reproducibility of neuropathological abnormalities, dominated medicine in�the 19th and early 20th centuries [8]. Since then, an expanding collection of reproducible biological causes, from neurosyphilis, head trauma, stroke, tumor, demyelination and many others caused symptom complexes that overlapped with classic psychiatric disorders [9-11]. More recently, neuroinflammatory and immunological abnormalities have been documented in patients with classical psychiatric disorders.

Peripheral immune modulators can induce psychiatric symptoms in animal models and humans [12-19]. Healthy animals injected with pro-inflammatory IL-1? and tumor necrosis factor alpha (TNF-?) cytokines demonstrate �sick- ness behavior� associated with social withdrawal [12]. In humans, injections of low-dose endotoxin deactivate the ventral striatum, a region critical for reward processing, producing anhedonia a debilitating depressive symptom [14]. Approximately 45% of non-depressed hepatitis C and cancer patients treated with IFN-? develop depressive symptoms associated with increased serum IL-6 levels [12,15,17,18].

Medical conditions associated with chronic inflammatory and immunological abnormalities, including obesity, diabetes, malignancies, rheumatoid arthritis, and multiple sclerosis, are risk factors for depression and bipolar disorder [10,12,13,15,17,18]. The positive�correlation between these medical conditions and psychiatric illness suggests the presence of a widespread underlying inflammatory process affecting the brain among other organs [10,19,20]. A 30-year population- based study showed that having an autoimmune disease or a prior hospitalization for serious infection increased the risk of developing schizophrenia by 29% and 60%, respectively [16]. Further, herpes simplex virus, Toxoplasma gondii, cytomegalovirus, and influenza during pregnancy increase the risk of developing schizophrenia [16].

Peripheral cellular [21,22] (Table 2), and humoral immunological abnormalities [13,21-23] are more prevalent in psychiatric patients relative to healthy controls. In both pilot (n = 34 patients with major depressive disorder (MDD), n = 43 healthy controls) and replication studies (n = 36 MDD, n = 43 healthy controls), a serum assay comprising nine serum biomarkers distinguished MDD subjects from healthy controls with 91.7% sensitivity and 81.3% specificity; significantly elevated biomarkers for neuropsychiatric symptoms were the immunological molecules alpha 1 antitrypsin, myeloperoxidase, and soluble TNF-? receptor II [23].

neuroinflammation table 2We first review the association between autoimmunity and neuropsychiatric disorders, including: 1) systemic lupus erythematosus (SLE) as a prototype of systemic auto- immune disease; 2) autoimmune encephalitides associated with serum anti-synaptic and glutamic acid decarboxylase (GAD) autoantibodies; and 3) pediatric neuropsychiatric autoimmune disorders associated with streptococcal infections (PANDAS) and pure obsessive-compulsive dis- order (OCD) associated with anti-basal ganglia/thalamic autoantibodies. We then discuss the role of innate inflammation/autoimmunity in classical psychiatric disorders, including MDD, bipolar disorder (BPD), schizophrenia, and OCD.

Neuropsychiatric Disorders Associated With Autoimmunity

Systemic Lupus Erythematosus

Between 25% to 75% of SLE patients have central nervous system (CNS) involvement, with psychiatric symptoms typically occurring within the first two years of disease on- set. Psychiatric symptoms may include anxiety, mood and psychotic disturbances [97]. Brain magnetic resonance imaging (MRI) is normal in approximately 42% of neuropsychiatric SLE cases [97]. Microangiopathy and blood� brain barrier (BBB) breakdown may permit entry of autoantibodies into the brain [97]. These antibodies include anti-ribosomal P (positive in 90% of psychotic SLE patients) [1], anti-endothelial cell, anti-ganglioside, anti- dsDNA, anti-2A/2B subunits of N-methyl-D-aspartate receptors (NMDAR) and anti-phospholipid antibodies [97]. Pro-inflammatory cytokines�principally IL-6 [97], S100B�[97], intra-cellular adhesion molecule 1 [97] and matrix- metalloproteinase-9 [98] are also elevated in SLE. Psychiatric manifestations of SLE, Sjo?gren�s disease, Susac�s syndrome, CNS vasculitis, CNS Whipple�s disease, and Behc?et�s disease were recently reviewed [1].

Neuropsychiatric Autoimmune Encephalitides Associated With Serum Anti-Synaptic & Glutamic Acid Decarboxylase

Autoantibodies

Autoimmune encephalitides are characterized by an acute onset of temporal lobe seizures, psychiatric features, and cognitive deficits [2,3,99-108]. The pathophysiology is typically mediated by autoantibodies targeting synaptic or intracellular autoantigens in association with a paraneo plastic or nonparaneoplastic origin [3]. Anti-synaptic autoantibodies target NR1 subunits of the NMDAR [100,108,109], voltage-gated potassium channel (VGKC) complexes (Kv1 subunit, leucine-rich glioma inactivated (LGI1) and contactin associated protein 2 (CASPR2)) [101,102,106], GluR1 and GluR2 subunits of the amino-3- hydroxy-5-methyl-l-4-isoxazolepropionic acid receptor (AMPAR) [6,110,111] and B1 subunits of the ?-aminobu- tyric acid B receptors (GABABR) [3,99,103]. Anti-intracellular autoantibodies target onconeuronal and GAD-65 autoantigens [2,3].

The inflammation associated with anti-synaptic autoantibodies, particularly NMDAR-autoantibodies, is typically much milder than that associated with GAD-autoantibodies or anti-neuronal autoantibodies related to systemic auto- immune disorders or paraneoplastic syndromes [2,107].

Although neurological symptoms eventually emerge, psychiatric manifestations, ranging from anxiety [2,3] to psychosis mimicking schizophrenia [2-6], can initially dominate or precede neurological features. Up to two- thirds of patients with anti-NMDAR autoimmune encephalitis, initially present to psychiatric services [5]. Anti-synaptic antibodies-mediated autoimmune encephalitides must be considered in the differential of acute psychosis [2-6]. Psychiatric presentations can include normal brain MRI and cerebrospinal fluid (CSF) ana- lysis, without encephalopathy or seizures [2,3,5,6,107]. We reported a case of seropositive GAD autoantibodies associated with biopsy-proven neuroinflammation, despite normal brain MRI and CSF analyses, where the patient presented with isolated psychosis diagnosed as schizophrenia by Diagnostic and Statistical Manual of Mental Disorders, 4th Edition (DSM-IV) criteria [2]. Further, seronegative autoimmune encephalitides can also present with prominent neuropsychiatric disturbances, making diagnosis more elusive [107,112,113]. Psychiatric and neurological features associated with anti- synaptic and GAD autoantibodies are summarized in Table 1 [1-6,99-108,114].

Serum anti-synaptic and GAD autoantibodies may occur in patients with pure psychiatric disorders [2,4,5,112,115-121]. In a prospective cohort of 29 subjects who met the DSM-IV criteria for schizophrenia, serum anti-NMDAR autoantibodies were found in three subjects, and anti-VGKC-complex autoantibodies were found in one subject [5]. Using more sensitive techniques to detect immunoglobulin G (IgG) NR1 auto- antibodies in 100 patients with definite schizophrenia, no autoantibodies were identified [122]. However, this study did not assess autoantibodies targeting the NR2 subunit of NMDAR. Other studies reported significantly increased odds of elevated (?90th percentile non-psychiatric control levels) NR2 antibody levels (odds ratio (OR) 2.78, 95% confidence interval (CI) 1.26 to 6.14, P = 0.012) among individuals with acute mania (n = 43), but not in chronic mania or schizophrenia [116].

PANDAS & Pure Obsessive-Compulsive Disorder Associated With Anti-Basal Ganglia/Thalamic Autoantibodies

OCD often complicates neurological disorders involving the basal ganglia including Sydenham�s chorea, Huntington�s disease and Parkinson�s disease. Anti- basal ganglia antibodies are implicated in Sydenham�s chorea [123]. PANDAS is characterized by acute exacerbations of OCD symptoms and/or motor/phonic tics following a prodromal group A ?-hemolytic streptococcal infection. The pathophysiology is thought to involve cross-reactivity between anti-streptococcal antibodies and basal ganglia proteins [124]. The clinical overlap between the PANDAS and pure OCD suggests a common etiological mechanism [125].

Among a random cohort of 21 pure OCD patients, 91.3% had CSF anti-basal ganglia (P <0.05) and anti-thalamic autoantibodies (P <0.005) at 43 kDa [88], paralleling functional abnormalities in the cortico-striatal-thalamo-cortico circuitry of OCD subjects [84]. Another study documented that 42% (n = 21) of OCD pediatric and adolescent subjects had serum anti-basal ganglia autoantibodies at 40, 45, and 60 kDa compared to 2% to 10% of controls (P = 0.001) [7]. Anti�basal ganglia autoantibodies were detected in the sera of 64% of PANDAS subjects (n = 14), compared to only 9% (n = 2) of streptococcal-positive/OCD-negative controls (P <0.001) [126]. One study found no difference between the prevalence of anti-basal ganglia autoantibodies in OCD (5.4%, n = 4) versus MDD controls (0%) [127]; however, a limitation was the random use of rat cortex and bovine basal ganglia and cortex that might have limited the identification of seropositive cases.

The basal ganglia autoantigens are aldolase C (40 kDa), neuronal-specific/non-neuronal enolase (45 kDa doublet) and pyruvate kinase M1 (60 kDa)�neuronal glycolytic enzymes�involved in neurotransmission, neuronal metabolism

Page 3 of 24 and cell signaling [128]. These enzymes exhibit substantial structural homology to streptococcal proteins [129]. The latest study (96 OCD, 33 MDD, 17 schizophrenia subjects) tested patient serum against pyruvate kinase, aldolase C and enolase, specifically; a greater pro- portion of OCD subjects were sero-positive relative to controls (19.8% (n = 19) versus 4% [n = 2], P = 0.012) [130].

Yet, in the same study only one of 19 sero-positive OCD subjects also had positive anti-streptolysin O antibody ti- ters, suggesting that in pure OCD the anti-streptolysin O antibody seronegativity does not exclude the presence of anti-basal ganglia autoantibodies.

In pure OCD, sero-positivity for anti-basal ganglia/ thalamic antibodies is associated with increased levels of CSF glycine (P = 0.03) [88], suggesting that these anti- bodies contribute to hyperglutamatergia observed in OCD [84,88,131]. The improvement of infection-provoked OCD with immune therapies supports the pathogenicity of these autoantibodies [132]. A large NIH trial assessing the efficacy of intravenous immunoglobulin (IVIG) for children with acute onset OCD and anti-streptococcal antibodies is ongoing (ClinicalTrials.gov: NCT01281969). However, the finding of slightly higher CSF glutamate levels in OCD patients with negative CSF anti-basal ganglia/thalamic anti- bodies as compared to those with positive CSF antibodies suggests that non-immunological mechanisms may play role in OCD [84]. Other mechanisms, including cytokine- mediated inflammation (Table 2), are also hypothesized.

Psychiatric Disorders Associated With Innate Inflammation

Disorders of innate inflammation/autoimmunity occur in some patients with classical psychiatric disorders. We discuss innate inflammation-related CNS abnormalities� including glial pathology, elevated cytokines levels, cyclo-oxygenase activation, glutamate dysregulation, increased S100B levels, increased oxidative stress, and BBB dysfunction�in MDD, BPD, schizophrenia, and OCD. We also describe how innate inflammation may be mechanistically linked to the traditional monoaminergic and glutamatergic abnormalities reported in these disorders (Figures 1 and 2). The therapeutic role of antiinflammatory agents in psychiatric disorders is also reviewed.

neuroinflammation fig 1

neuroinflammation fig 2Astroglial & Oligodendroglial Histopathology

Astroglia and oligodendroglia are essential to neural metabolic homeostasis, behavior and higher cognitive functions [54-56,133-136]. Normal quiescent astroglia provide energy and trophic support to neurons, regulate synaptic neurotransmission (Figure 2), synaptogenesis, cerebral blood flow, and maintain BBB integrity [134,136,137]. Mature oligodendroglia provide energy and trophic support to neurons and maintain BBB integrity, and regulate axonal repair�and myelination of white matter tracts providing inter- and intra-hemispheric connectivity [54-56]. Both astroglia and oligodendroglia produce anti-inflammatory cytokines that can down-regulate harmful inflammation [52,55].

In MDD, astroglial loss is a consistent post-mortem finding in functionally relevant areas, including the anterior cingulate cortex, prefrontal cortex, amygdala, and white matter [35-38,42-46,55,138-147], with few exceptions [42,43]. Post-mortem studies revealed reduced glial fibrillary acidic protein (GFAP)-positive astroglial density primarily in the prefrontal cortex [37,38] and amygdala [36]. A large proteomic analysis of frontal cortices from depressed patients showed significant reductions in three GFAP isoforms [39]. Although in one study that reported no significant glial loss, subgroup analysis revealed a significant decrease (75%) in GFAP-positive astroglial density among study subjects younger than 45 years of age [35]. A morphometric study similarly showed no changes in glial density in late-life MDD brains [148]. We hypothesize that the apparent absence of astroglial loss among older MDD patients may reflect secondary astrogliosis [35] that is associated with older age [42,50] rather than a true negative.

Animal studies are consistent with human studies showing astroglial loss in MDD. Wistar-Kyoto rats� known to exhibit depressive-like behaviors�revealed reduced astroglial density in the same areas as observed in humans [40]. Administration of the astroglial-toxic agent, L-alpha-aminoadipic acid, induces depressive- like symptoms in rats, suggesting that astroglial loss is pathogenic in MDD [41].

Post-mortem studies of MDD subjects documented reduced oligodendroglial density in the prefrontal cortex and amygdala [54-57,66], which may correlate with brain MRI focal white matter changes occasionally noted in some MDD patients [57]. However, microvascular abnormalities may also contribute to these changes [57].

In BPD, some studies demonstrate significant glial loss [138,143,149,150], while others do not [37,44-46]. These inconsistent findings may result from lack of control for: 1) treatment with mood stabilizers, because post-hoc ana- lysis reported by some studies showed significant reduction in glial loss only after controlling for treatment with lithium and valproic acid [46]; 2) familial forms of BPD, as glial loss is particularly prominent among BPD patients with a strong family history [143]; and/or, 3) the predominant state of depression versus mania, as glial loss is frequent in MDD [35-38,42-46,55,138-147]. Whether astroglia or oligodendroglia account for the majority of glial loss is unclear; while proteomic analysis revealed a significant decrease in one astroglial GFAP isoform [39], several other post-mortem studies found either unchanged [36,37] or reduced GFAP-positive astroglial expression in the orbitrofrontal cortex [47], or reduced oligodendroglial density [54-56,58,59].

In schizophrenia, astroglial loss is an inconsistent finding [48,150]. While some studies showed no significant astroglial loss [42,50,51], several others found reduced astroglial density [37,38,43,44,48,49,151] and significant reductions in two GFAP isoforms [39]. Inconsistent findings may result from: 1) MDD comorbidity, which is often associated with glial loss; 2) age variation, as older patients have increased GFAP-positive astroglia [35,42,50]; 3) regional [150] and cortical layer variability [48]; 4) treatment with antipsychotic drugs, as experimental studies show both reduced [152] and increased [153] astroglial-density related to chronic antipsychotic treatment [70]; and 5) disease state (for example, suicidal versus non-suicidal behavior) [154]. Post-mortem studies documented oligodendroglial loss [54,56,60-65,148,155,156], particularly in the prefrontal cortex, anterior cingulate cortex, and hippocampus [148]. Ultrastructural examination of the prefrontal region showed abnormally myelinated fibers in both gray and white matter; both age and duration of illness were positively correlated with the white matter abnormalities [157].

In contrast to neurodegenerative disorders that are commonly associated with astroglial proliferation [136], psychiatric disorders are instead associated with either reduced or unchanged astroglial density [138]. The lack of increased glial density in early-onset psychiatric disorders [44,138] may reflect the slower rate of degenerative progression in psychiatric illnesses [138].

We postulate that degenerative changes associated with psychiatric disorders are subtler and not severe enough to provoke astroglial intracellular transcription factors that positively regulate astrogliosis, including signal transducer activator of transcription 3 and nuclear factor kappa B (NF-?B) [136].

While the majority of post-mortem studies focused on the alteration of glial density in MDD, BPD, and schizophrenia, others described alteration of glial cell morphology, with mixed findings. In MDD and BPD, glial size is either increased or unchanged [55]. One study found reduced glial size in BPD and schizophrenia but not in MDD [43]. A post-mortem study of depressed patients who committed suicide found increased astroglial size in the anterior cingulate white matter but not in the cortex [158]. One study in schizophrenic subjects found markedly decreased astroglial size in layer V of the dorsolateral prefrontal cortex, notwithstanding that astroglial density is double that of controls in the same layer [48]. The mixed results may partially reflect earlier studies of glial alterations in psychiatric illnesses that did not specify astroglia versus oligodendroglia [148].

Glial loss in psychiatric illnesses may contribute to neuroinflammation through several mechanisms, including abnormal cytokine levels (see Cytokine section), dysregulated glutamate metabolism (see Glutamate section), elevated S100B protein (see S100B section),�and altered BBB function (see Blood brain barrier section), resulting in impaired cognition and behavior [44,45,54,133,159].

Microglial Histopathology

Microglia are the resident immune cells of the CNS. They provide ongoing immune surveillance and regulate developmental synaptic pruning [160,161]. CNS injury transforms ramified resting microglia into activated elongated rod-shaped and macrophage-like phagocytic amoeboid cells that proliferate and migrate towards the site of injury along chemotactic gradients (that is, micro- glial activation and proliferation (MAP)) [161]. Human microglial cells express NMDARs that may mediate MAP leading to neuronal injury [162].

In MDD, BPD and schizophrenia, the results of post- mortem studies investigating the presence of MAP are mixed. Post-mortem studies revealed elevated MAP in only one out of five MDD subjects [67]. In some BPD disorder patients, increased human leukocyte antigen-DR-positive microglia displaying thickened processes were documented in the frontal cortex [69]. In schizophrenia, while some studies reported elevated MAP relative to controls, others showed no difference between groups [22,67,70]. In a post-mortem study assessing MAP in MDD and BPD; quinolinic acid-positive microglial cell density was in- creased in the subgenual anterior cingulate cortex and anterior midcingulate cortex of MDD and BPD patients who committed suicide relative to controls [53]. Post-hoc ana- lysis revealed this increased MAP was solely attributable to MDD and not BPD, since the positive microglial immuno-staining in MDD subjects was significantly greater than that in the BPD subgroup in both the subgenual anterior cingulate and midcingulate cortices, and since the microglia density was similar in both BPD and control groups [53]. A study comparing all three disorders (nine MDD, five BPD, fourteen schizophrenia, ten healthy controls) demonstrated no significant difference in microglial density across the four groups [68].

These mixed results may be attributed to variable microglial immunological markers used among different studies [70] and/or the failure to control for disease severity [22,53,68]. Notably, three post-mortem studies of MDD and schizophrenic subjects documented a strong positive correlation between MAP and suicidality in the anterior cingulate cortex and mediodorsal thalamus, in- dependent of psychiatric diagnosis [22,53,68]. Thus, MAP may be a state rather than a trait marker for MDD and schizophrenia.

In OCD, animal models suggest that dysfunction and reduction of certain microglial phenotypes, such as those expressing the Hoxb8 gene, which encodes homeobox transcription factor, can cause OCD-like behavior [71,72].

Hoxb8 knockout mice exhibit excessive grooming behavior and anxiety in association with reduced microglial density [71,72]. This excessive grooming behavior resembles the behavioral characteristics of human OCD. Hoxb8 injection in adult Hoxb8 knockout mice reverses microglial loss and restores normal behavior [71,72]. The role of these specific microglial phenotypes in human OCD is unclear.

Experimental data suggest that MAP comprises distinctive harmful and neuroprotective phenotypes (Figure 2). Harmful microglia do not express major histocompatibility complex II (MHC-II) and, therefore, cannot act as antigen presenting cells (APC) [163,164]; they promote deleterious effects [17,69,165] through proinflammatory cytokine production, nitric oxide synthase signaling [17,166], promoting glial and BBB-pericyte/endothelial cyclooxygenase- 2 (COX-2) expression [167], inducing astroglial S100B secretion (see S100B section), and microglial glutamate release [17,136,168,169]. Harmful microglia also secrete prostaglandin E-2 (PGE-2) that promotes proinflammatory cytokines production, which in turn increases PGE-2 levels in a feed-forward cycle [29]. Further, PGE-2 stimulates COX-2 expression, which mediates the conversion of arachidonic acid to PGE-2, setting up another feed-forward cycle [29].

Neuroprotective microglia by contrast can: 1) express MHC-II in vivo and in vitro [163,166] and act as cognate APC (Figure 2) [163,164,166]; 2) facilitate healing and limit neuronal injury by promoting secretion of antiinflammatory cytokines [17], brain-derived neurotrophic factor [17], and insulin-like growth factor-1 [166]; and 3) express excitatory amino acid transporter-2 (EAAT2) that eliminates excess extracellular glutamate [163,166], and promotes neuroprotective T lymphocytic autoimmunity (Figure 2) [163,164]. However, more studies are needed to confirm the contributory role of neuroprotective microglia to neuropsychiatric disorders in humans.

 

In vitro animal studies suggest that the ratio of harmful versus neuroprotective microglia can be influenced by the net effect of inflammatory counter-regulatory mechanisms [15,74,164,166]. These mechanisms include the number of neuroprotective CD4+CD25+FOXP3+ T regulatory cells ((T regs) Figure 1) [15,74,164,166] and brain cytokine levels; low IFN-? levels may promote neuroprotective microglia (Figure 2) [166], whereas high levels can promote the harmful phenotype [166].

The Role Of Cytokines

Proinflammatory cytokines include IL-1?, IL-2, IL-6, TNF-? and IFN-?. They are secreted primarily by micro- glia, Th1 lymphocytes and M1 phenotype monocytes/ macrophages (Figure 1) [15,170]. They promote harmful inflammation. Antiinflammatory cytokines include IL-4, IL-5 and IL-10. They are primarily secreted by astroglia,�Th2 lymphocytes, T regs and M2 phenotype monocytes/ macrophages [15,52,74]. They can limit harmful inflammation [15,74] by converting the proinflammatory M1-pheno- type into the beneficial antiinflammatory M2-phenotype [15], and potentially by promoting the neuroprotective microglial phenotype [15,17,74,163,166]. The role of proinflammatory/antiinflammatory cytokines in psychiatric dis- orders is supported by several lines of evidence (Figure 1, Table 2) [15,17,29,52,74].

In MDD, the most recent meta-analysis (29 studies, 822 MDD, 726 healthy controls) of serum proinflammatory cytokines confirmed that soluble IL-2 receptor, IL-6 and TNF-? levels are increased in MDD (trait markers) [91], while, IL-1?, IL-2, IL-4, IL-8 and IL-10, are not statistically different from controls [91]. In a primary cytokine study comparing MDD subgroups (47 suicidal- MDD, 17 non-suicidal MDD, 16 health controls), both sera IL-6 and TNF-? were significantly higher, while IL-2 levels were significantly lower in MDD subjects who committed suicide relative to both other groups [96]. This finding suggests that IL-6 and TNF-? are also state markers of MDD [96]. The decrease of serum IL-2 levels associated with acute suicidal behavior may reflect increased binding to its upregulated receptor in the brain; parallel to the aforementioned meta-analysis showing increased soluble IL-2 receptor in MDD [91]. Studies investigating the clinical significance of cytokines in MDD showed that serum cytokine levels are elevated during acute depressive episodes [171,172] and normalized following successful, but not failed, treatment with antidepressants [17] and electro- convulsive therapy [29]; these findings suggest a possible pathogenic role for cytokines.

In BPD, serum cytokine alterations were summarized in a recent review; TNF-?, IL-6 and IL-8 are elevated during manic and depressive phases, whereas IL-2, IL-4 and IL-6 are elevated during mania [92]. Other studies showed that sera IL-1? and IL-1 receptor levels are not statistically different from healthy controls [92], although tissue studies documented increased levels of IL-1? and IL-1 receptor in the BPD frontal cortex [69].

In schizophrenia, results from studies investigating cytokine abnormalities are conflicting (Table 2). While some studies found both decreased serum proinflammatory (IL-2, IFN-?) and increased serum and CSF antiinflammatory cytokines (IL-10) [52], others found elevated serum pro- and antiinflammatory cytokines, with a proinflammatory type dominance [22,173,174]. One cytokine meta-analysis (62 studies, 2,298 schizophrenia, 858 healthy controls) showed increased levels of IL-1R antagonist, sIL-2R and IL-6 [174]. However, this study did not account for the use of antipsychotics, which is thought to enhance proinflammatory cytokine production [52]. A more recent cytokine meta-analysis (40 studies, 2,572 schizophrenics,�4,401 controls) that accounted for antipsychotics, found that TNF-?, IFN-?, IL-12 and sIL-2R are consistently elevated in chronic schizophrenia independent of disease activity (trait markers), while IL-1?, IL-6 and transforming growth factor beta positively correlate with disease activity (state markers)[173]. Cell cultures of peripheral blood mononuclear cells (PBMC) obtained from schizophrenic patients produced higher levels of IL-8 and IL-1? spontaneously as well as after stimulation by LPS, suggesting a role for activated monocytes/macrophages in schizophrenia pathology [175].

In OCD, results from a random survey of sera and CSF cytokines, and LPS-stimulated PBMC studies, are inconsistent [93-95,176-179]. There is a correlation between OCD and a functional polymorphism in the promoter region of the TNF-? gene [34], although low-powered studies did not confirm this association [180]. Therefore, the mixed results from studies documenting either increased or decreased TNF-? cytokine levels [93,176-178] may reflect their variable inclusion of the subset of OCD subjects with this particular polymorphism in their cohorts.

Cytokine Response Polarization In Major Depression & Schizophrenia

Cytokine response phenotypes are classified as either proinflammatory Th1 (IL-2, IFN-?) or antiinflammatory Th2 (IL-4, IL-5, IL-10) according to the immune functions they regulate. While Th1 cytokines regulate cell-mediated immunity directed against intra-cellular antigens, Th2 cytokines regulate humoral immunity directed against extra- cellular antigens [29,52]. Th1 cytokines are produced by Th1 lymphocytes and M1 monocytes whereas Th2 cytokines are produced by Th2 lymphocytes and M2 monocytes [29,52]. In the brain, microglia predominantly secrete Th1 cytokines, whereas astroglia predominately secrete Th2 cytokines [29,52]. The reciprocal ratio of Th1:Th2 cytokines, henceforth �Th1-Th2 seesaw,� is influenced by the proportion of activated microglia (excess Th1) to astroglia (excess Th2) and the interplay between activated T cells and excessive CNS glutamate levels that we hypothesized to favor Th1 response (Figure 2) [29,163,166].

The Th1-Th2 seesaw imbalance can influence trypto- phan metabolism by altering its enzymes [21,52] thereby shifting tryptophan catabolism towards kynurenine (KYN) and KYN catabolism towards either of its two down- stream metabolites; microglia quinolinic acid that is Th1 response-mediated or astroglial kynurenic acid (KYNA) (Figure 1) that is Th2 response-mediated [21,29,170].

Tryptophan metabolism enzymes affected by Th1-Th2 seesaw include (Figure 1): indoleamine 2,3-dioxygenase (IDO) expressed by microglia and astroglia, the rate-limiting enzymes that mediate the conversion of trypto- phan to KYN and serotonin to 5-hydroxyindoleacetic acid�[21,29]. Kynurenine 3-monooxygenase (KMO), solely expressed by microglia, is the rate-limiting enzyme that converts KYN to 3-hydroxykynurenine (3-OH-KYN), which is further metabolized to quinolinic acid [21,29]. Tryptophan-2,3-dioxygenase (TDO), expressed solely by astroglia, is the rate-limiting enzyme that converts�tryptophan to KYN [21,29]. Kynurenine aminotransferase (KAT), expressed primarily in astroglial processes, is the rate-limiting enzyme that mediates the conversion of KYN to KYNA [21,29].

Th1 cytokines activate microglial IDO and KMO, shifting microglial KYN catabolism towards quinolinic�acid (NMDAR agonist) synthesis, while Th2 cytokines in- activate microglial IDO and KMO, shifting astroglial KYN catabolism towards TDO- and KAT-mediated KYNA (NMDAR antagonist) synthesis (Figure 1) [21,29].

Th1 and Th2 predominant immunophenotypes have been proposed for MDD and schizophrenia, respectively, based on peripheral, rather than CNS, cytokines patterns [52,173]. We believe that peripheral cytokines patterns are unreliable surrogate markers of those in the CNS. Indeed, peripheral cytokine levels can be influenced by many extra-CNS variables, which are not consistently controlled for in several of the peripheral cytokines studies, including: 1) age, body mass index, psychotropic medications, smoking, stress and circadian fluctuations; 2) the influence of�disease activity/state on the production of selected cytokines synthesis [95,173]; and 3) the effects of psychotropic agents on cytokines production [52]. The short half-lives and the rapid turnover of serum cytokines [181] (for ex- ample, 18 minutes for TNF-? [182] versus 60 minutes for IL-10 [183]), may further limit the reliability of interpreting their levels measured from random sera sampling.

In MDD, there is a consensus that a proinflammatory Th1 immunophenotype response predominates (Table 2) [17,29]. High levels of quinolinic acid in post-mortem MDD brains [53], suggest the presence of an upregulated Th1 response (Figure 1) [21,29]. Elevated CNS quinolinic acid can promote calcium influx mediated apoptosis of human astroglia [184], which hypothetically may blunt the�astroglia-derived Th2 response [29], tipping Th1 versus Th2 seesaw balance in favor of the microglial Th1 response. CNS hyposerotonergia [29] adds further support to an excess Th1 response, which is shown to reduce CNS serotonin synthesis [185] and to increase its degradation (Figure 1) [21,29].

CNS hyperglutamatergia may also contribute to an excess Th1 response in the brain (Figure 2). An in vitro study suggests that the peripheral resting T lymphocytes constitutively express metabotropic glutamate receptor 5 (mGluR5) [164], whose binding to glutamate inhibits lymphocytic IL-6 release, thereby downregulating auto- reactive T-effector cell proliferation [164]. Activated T lymphocytes, but not resting T lymphocytes, can cross the BBB [37].

Experimental data suggest that the interaction between T cell receptors of activated T lymphocytes and their cognate antigen presenting cells can downregulate mGluR5 and induce mGluR1 expressions [164]. In animal models, binding of excess glutamate to lymphocytic mGluR1 receptors promotes production of Th1 cytokines, including IFN-? [164].

We hypothesize that in some MDD patients, parallel to experimental data [164], the binding of excess CNS glutamate to induced lymphocytic mGluR1 receptors may contribute to an excess Th1 response, including IFN-? (Figure 2). We speculate that IFN-? in a small quantity, similar to its in vitro effects on microglia [166], may induce microglial expression of MHC-II and EAAT2 [163,166], allowing microglia to serve as cognate antigen presenting cells and to provide glutamate reuptake function [163,164,166], thereby transforming harmful microglia into neuroprotective phenotype [163,166] that participate in eliminating excess extracellular glutamate [163,164,166]. Therefore, we also hypothesize that excess Th1 response in subgroups of MDD patients is a double-edged sword, promoting harmful inflammation and serving as a beneficial counter- regulatory mechanism that may limit excess glutamate- related neuroexcitotoxicity (Figure 2).

In schizophrenia, while some peripheral cytokine studies suggest the predominance of an antiinflammatory Th2 immunophenotype/response [52], others refute this [173,174]. However, we agree with the authors who hypothesized that the Th2 response is the dominant phenotype in schizophrenia [52]. Elevated brain, CSF, and serum levels of KYNA [21,52] suggest downregulation of micro-glial IDO and KMO, which is a function of Th2 response that shifts astroglial KYN catabolism towards KYNA synthesis (Figure 1) [21,52]. Reduced KMO activity and KMO mRNA expression in post-mortem schizophrenic brains [73] is consistent with excess Th2 response (Figure 1). Increased prevalence of Th2-mediated humoral immunity abnormalities in subgroups of schizophrenia patients�as evidenced by increased B cell counts [21,76], increased�production of autoantibodies including antiviral antibodies [76] and increased immunoglobulin E [52]�adds further support to the Th2 response dominance hypothesis.

Neuroinflammation & CNS Glutamate Dysregulation

Glutamate mediates cognition and behavior [186]. Syn- aptic glutamate levels are regulated by high-affinity sodium-dependent glial and neuronal EAATs, namely, the XAG- system responsible for glutamate reuptake/ aspartate release [137,164] and the sodium-independent astroglial glutamate/cystine antiporter system (Xc-) responsible for glutamate release/cystine reuptake [164]. Astroglial EAAT1 and EAAT2 provide more than 90% of glutamate re-uptake [79].

Neuroinflammation can alter glutamate metabolism and the function of its transporters [15,29,187,188], producing cognitive, behavioral, and psychiatric impairments [15,21,29,79,186,188,189]. Abnormalities of EAATs function/expression and glutamate metabolism in MDD, BPD, schizophrenia, and OCD are summarized in Table 2.

In MDD, there is evidence for cortical hyperglutamatergia (Table 2). Cortical glutamate levels correlated positively with the severity of depressive symptoms, and a five-week course of antidepressants decreased serum glutamate concentrations [85,86]. A single dose of ketamine, a potent NMDAR antagonist, can reverse refractory MDD for a week [17,21,29,85]. Excess CNS glutamate levels can induce neurotoxicity-mediated inflammation [163,164,188], including a proinflammatory Th1 response (Figure 2) [164].

Limited in vitro evidence suggests that inflammation/ proinflammatory cytokines can increase CNS glutamate levels [188] in a feed-forward cycle through several potential mechanisms: 1) proinflammatory cytokines can inhibit [15,17,168] and reverse [45,137] astroglial EAAT-mediated glutamate reuptake function; 2) proinflammatory cytokines can enhance microglial quinolinic acid synthesis [53], which has been experimentally shown to promote synaptosomal glutamate release [15,17,29,190]; 3) increased COX-2/PGE-2 and TNF-? levels can induce calcium influx [137], which, based on in vitro data, may increase astroglial glutamate and D-serine release [191]; and 4) activated microglia can express excess Xc- antiporter systems that mediate glutamate release [164,192].

In schizophrenia, prefrontal cortical hypoglutamatergia [87,90,193,194] (Table 2) and reduced NMDAR functionality are found [5]. Recent H1 magnetic resonance spectroscopy (MRS) meta-analysis (28 studies, 647 schizophrenia, 608 control) confirmed decreased glutamate and increased glutamine levels in the medial frontal cortex [90]. The contributory role of inflammation to hypoglutamatergia is not proven. Elevated KYNA synthesis in schizophrenia brains [21,52], typically a function of Th2 response (Figure 1), can inhibit NR1 subunit of NMDAR and alpha 7 nicotinic�acetylcholine receptor (?7nAchR) [195], leading to decreased NMDAR function and reduced ?7nAchR-mediated glutamate release [195].

In BPD and OCD, data suggest CNS cortical hyper- glutamatergia in both disorders (Table 2) [78,84,88,131]. The contribution of inflammation (BPD and OCD) and autoantibodies (OCD)[7,77,84,88,130] to increased CNS glutamate levels requires further investigation.

The Role Of S100B

S100B is a 10 kDa calcium-binding protein produced by astroglia, oligodendroglia, and choroid plexus ependymal cells [196]. It mediates its effects on the surrounding neurons and glia via the receptor for advanced glycation end-product [196]. Nanomolar extracellular S100B levels provide beneficial neurotrophic effects, limit stress-related neuronal injury, inhibit microglial TNF-? release, and increase astroglial glutamate reuptake [196]. Micromolar S100B concentrations, predominantly produced by activated astroglia and lymphocytes [196,197], have harmful effects transduced by receptor for advanced glycation end product that include neuronal apoptosis, production of COX-2/PGE-2, IL-1? and inducible nitric oxide species, and upregulation of monocytic/microglial TNF-? secretion [21,196,198].

Serum and, particularly, CSF and brain tissue S100B levels are indicators of glial (predominantly astroglial) activation [199]. In MDD and psychosis, serum S100B levels positively correlate with the severity of suicidality, independent of psychiatric diagnosis [200]. Post-mortem analysis of S100B showed decreased levels in the dorso- lateral prefrontal cortex of MDD and BPD, and in- creased levels in the parietal cortex of BPD [196].

Meta-analysis (193 mood disorder, 132 healthy controls) confirmed elevated serum and CSF S100B levels in mood disorders, particularly during acute depressive episodes and mania [201].

In schizophrenia, brain, CSF and serum S100B levels are elevated [199,202]. Meta-analysis (12 studies, 380 schizophrenia, 358 healthy controls) confirmed elevated serum S100B levels in schizophrenia [203]. In post-mortem brains of schizophrenia subjects, S100B-immunoreactive astroglia are found in areas implicated in schizophrenia, including anterior cingulate cortex, dorsolateral prefrontal cortex, orbitofrontal cortex and hippocampi [154]. Elevated S100B levels correlate with paranoid [154] and negativistic psychosis [204], impaired cognition, poor therapeutic response and duration of illness [202]. Genetic polymorphisms in S100B [32] and receptor for advanced glycation end-product genes in schizophrenia cohorts (Table 2) [32,33,205] suggest these abnormalities are likely primary/ pathogenic rather than secondary/biomarkers. Indeed, the decrease in serum S100B levels following treatment with antidepressants [201] and antipsychotics [196] suggests�some clinical relevance of S100B to the pathophysiology of psychiatric disorders.

Neuroinflammation & Increased Oxidative Stress

Oxidative stress is a condition in which an excess of oxidants damages or modifies biological macromolecules such as lipids, proteins and DNA [206-209]. This excess results from increased oxidant production, decreased oxidant elimination, defective antioxidant defenses, or some combination thereof [206-209]. The brain is particularly vulnerable to oxidative stress due to: 1) elevated amounts of peroxidizable polyunsaturated fatty acids; 2) relatively high content of trace minerals that induce lipid peroxidation and oxygen radicals (for example, iron, copper); 3) high oxygen utilization; and 3) limited anti-oxidation mechanisms [206,207].

Excess oxidative stress can occur in MDD [206], BPD [206,207], schizophrenia [207,209], and OCD [206,208]. Peripheral markers of oxidative disturbances include increased lipid peroxidation products (for example, malondialdehyde and 4-hydroxy-2-nonenal), increased nitric oxide (NO) metabolites, decreased antioxidants (for example, glutathione) and altered antioxidant enzyme levels [206,207].

In MDD, increased superoxide radical anion production correlates with increased oxidation-mediated neutrophil apoptosis [206]. Serum levels of antioxidant enzymes (for example, superoxide dismutase-1) are elevated during acute depressive episodes and normalize after selective serotonin reuptake inhibitors (SSRIs) treatment [206]. This suggests that in MDD, serum antioxidant enzyme levels are a state marker, which may reflect a compensatory mechanism that counteracts acute increases in oxidative stress. [206]. In schizophrenia by contrast, CSF soluble superoxide dismutase-1 levels are substantially decreased in early-onset schizophrenic patients relative to chronic schizophrenic patients and healthy controls. This suggests that reduced brain antioxidant enzyme levels may contribute to oxidative damage in acute schizophrenia [210], though larger studies are needed to confirm this finding.

Several additional experimental and human studies examined in more detail the mechanisms underlying the pathophysiology of increased oxidative stress in psychiatric disorders [206-262]. In animal models of depression, brain levels of glutathione are reduced while lipid peroxidation and NO levels are increased [206,262].

Postmortem studies show reduced brain levels of total glutathione in MDD, BPD [206] and schizophrenic subjects [206,207]. Fibroblasts cultured from MDD patients show increased oxidative stress independent of glutathione levels [262], arguing against a primary role of glutathione depletion as the major mechanism of oxidative stress in depression.

Microglial activation may increase oxidative stress through its production of proinflammatory cytokines and NO [206-209]. Proinflammatory cytokines and high NO levels may promote reactive oxygen species (ROS) formation, which in turn accelerates lipid peroxidation, damaging membrane phospholipids and their membrane-bound monoamine neurotransmitter receptors and depleting endogenous antioxidants. Increased ROS products can enhance microglial activation and increase proinflammatory production via stimulating NF-?B [208], which in turn perpetuates oxidative injury [208], creating the potential for a pathological positive feedback loop in some psychiatric disorders [206-209]. Although neuroinflammation can increase brain glutamate levels [85,86], the role of glutamatergic hyperactivity as a cause of oxidative stress remains unsubstantiated [207].

Mitochondrial dysfunction may contribute to increased oxidative stress in MDD, BPD and schizophrenia [206]. Postmortem studies in these disorders reveal abnormalities in mitochondrial DNA, consistent with the high prevalence of psychiatric disturbances in primary mitochondrial disorders [206]. In vitro animal studies show that proinflammatory cytokines, such as TNF-?, can reduce mitochondrial density and impair mitochondrial oxidative metabolism [211,212], leading to increased ROS production [206,213]. These experimental findings may imply mechanistic links among neuroinflammation, mitochondrial dysfunction and oxidative stress [206,213], meriting further investigation of these intersecting pathogenic pathways in human psychiatric disorders.

The vulnerability of neural tissue to oxidative damage varies among different psychiatric disorders based on the neuroanatomical, neurochemical and molecular pathways involved in the specific disorder [207]. Treatment effects may also be critical, as preliminary evidence suggests that antipsychotics, SSRIs and mood stabilizers possess antioxidant properties [206,207,262]. The therapeutic role of adjuvant antioxidants (for example, vitamins C and E) in psychiatric disorder remains to be substantiated by high- powered randomized clinical trials. N-acetylcysteine shows the most promising results to-date, with several randomized placebo-controlled trials demonstrating its efficacy in MDD, BPD and schizophrenia [207].

Blood�Brain Barrier Dysfunction

The BBB secures the brain�s immune-privileged status by restricting the entry of peripheral inflammatory mediators, including cytokines and antibodies that can impair neurotransmission [214,215]. The hypothesis of BBB breakdown and its role in some psychiatric patients [60,214,216,217] is consistent with the increased prevalence of psychiatric comorbidity in diseases associated with its dysfunction, including SLE [97], stroke [11],�epilepsy [218] and autoimmune encephalitides (Table 1). An elevated �CSF:serum albumin ratio� in patients with MDD and schizophrenia suggests increased BBB permeability [214].

In one study (63 psychiatric subjects, 4,100 controls), CSF abnormalities indicative of BBB-damage were detected in 41% of psychiatric subjects (14 MDD and BPD, 14 schizophrenia), including intrathecal synthesis of IgG, IgM, and/or IgA, mild CSF pleocytosis (5 to 8 cells per mm3) and the presence of up to four IgG oligoclonal bands [216]. One post-mortem ultrastructural study in schizophrenia revealed BBB ultrasructural abnormalities in the prefrontal and visual cortices, which included vacuolar degeneration of endothelial cells, astroglial-end-foot- processes, and thickening and irregularity of the basal lamina [60]. However, in this study, the authors did not comment on the potential contribution of postmortem changes to their findings. Another study investigating transcriptomics of BBB endothelial cells in schizophrenic brains identified significant differences among genes influencing immunological function, which were not detected in controls [217].

Oxidation-mediated endothelial dysfunction may con- tribute to the pathophysiology of BBB dysfunction in psychiatric disorders. Indirect evidence from clinical and experimental studies in depression [219] and, to a lesser extent, in schizophrenia [220] suggests that increased oxidation may contribute to endothelial dysfunction. Endothelial dysfunction may represent a shared mechanism accounting for the known association between depression and cardiovascular disease [219,221], which may be related to decreased levels of vasodilator NO [221-223]. Experimental studies suggest that reduced endothelial NO levels are mechanistically linked to the uncoupling of endothelial nitric oxide synthase (eNOS) from its essential co-factor tetrahydrobiopterin (BH4), shifting its substrate from L- arginine to oxygen [224-226]. Uncoupled eNOS promotes synthesis of ROS (for example, superoxide) and reactive nitrogen species (RNS) (for example, peroxynitrite; a product of the interaction of superoxide with NO) [227] rather than NO, leading to oxidation-mediated endothelial dysfunction [224-226].

Animal data showed that SSRIs could restore deficient endothelial NO levels [219], suggesting that anti-oxidative mechanisms may contribute to their antidepressant effects. In humans, L-methylfolate may potentiate anti- depressant effects of SSRIs [228], putatively by increasing levels of BH4, which is an essential cofactor for eNOS re- coupling-mediated anti-oxidation [229], as well as for the rate-limiting enzymes of monoamine (that is, serotonin, norepinephrine, dopamine) synthesis [228].

Taken together, both the recent work emphasizing the role of uncoupled eNOS-induced oxidative stress in the pathogenesis of vascular diseases [230,231] and the�epidemiological studies establishing depression as an in- dependent risk factor for vascular pathologies, such as stroke and heart disease [219,221], add further support to the clinical relevance of uncoupled eNOS-mediated endothelial oxidative damage in depression. Despite abundant evidence for cytokine abnormalities in human psychiatric illnesses and the experimental data showing that proinflammatory cytokines can reduce eNOS expression [212] and increase BBB permeability [215], human evidence that directly links excess proinflammatory cytokines to eNOS dysfunction and/or BBB impairment is lacking.

Imaging & Treating Inflammation In Psychiatric Illness

Imaging Neuroinflammation In Situ

Clinically, neuroinflammation imaging may prove to be crucial for identifying the subgroup of psychiatric patients with neuroinflammation who would be most likely to respond favorably to immunomodulatory therapies. Additionally, such imaging may allow clinicians to monitor neuroinflammation-related disease activity and its response to immune therapy in psychiatric patients. Imaging inflammation in the human brain has traditionally depended upon MRI or CT visualization of extravagated intravenous contrast agents, indicating localized breakdown of the BBB. Gadolinium-enhanced MRI occasionally demonstrates such breakdown in the limbic regions associated with emotional processing in patients with psychiatric dis- orders attributable to paraneoplastic or other encephalitides [107,109,113]. To our knowledge, however, abnormal enhancement has never been demonstrated in any classical psychiatric disorder [21,214,232], despite functional [214,216] and ultrastructural BBB abnormalities [60].

Whether or not subtler neuroinflammation can be visualized in vivo in classical psychiatric disorders remains unknown. One promising technique is positron emission tomography (PET) using radiotracers, such as C11- PK11195, which bind to the translocator protein, previously known as the peripheral benzodiazepine receptor, expressed by activated microglia [233,234].

Using this method, patients with schizophrenia were shown to have greater microglial activation throughout the cortex [235] and in the hippocampus during acute psychosis [236]. One study (14 schizophrenia, 14 controls) found no significant difference between [11C] DAA1106 binding in schizophrenia versus controls, but a direct correlation between [11C] DAA1106 binding and the severity of positive symptoms and illness duration in schizophrenia [236].

Investigators from our institution utilized C11-PK11195 PET to demonstrate bi-hippocampal inflammation in a patient with neuropsychiatric dysfunction, including psychotic MDD, epilepsy, and anterograde amnesia, associated with anti-GAD antibodies [237]. However, PK11195 PET has�low signal-to-noise properties and requires an on-site cyclotron.

Accordingly, research is being devoted to developing improved translocator protein ligands for PET and SPECT. Future high-powered post-mortem brain tissues studies utilizing protein quantification aimed at elucidating metabolic and inflammatory pathways, CNS cytokines and their binding receptors, in psychiatric disorders are needed to advance our understanding of the autoimmune pathophysiology.

Role Of Antiinflammatory Drugs In Psychiatric Disorders

Several human and animal studies suggest that certain antiinflammatory drugs may play an important adjunctive role in the treatment of psychiatric disorders (Table 3). Common drugs are cyclooxygenase inhibitors (Table 3) [238-245], minocycline (Table 3) [240-245], omega-3 fatty acids [246,247], and neurosteroids [248].

neuroinflammation table 3Several human studies showed that COX-2 inhibitors could ameliorate psychiatric symptoms of MDD, BPD, schizophrenia and OCD (Table 3) [248]. By contrast, adjunctive treatment with non-selective COX-inhibitors (that is, non-steroidal antiinflammatory drugs (NSAIDs)) may reduce the efficacy of SSRIs [249,250]; two large trials reported that exposure to NSAIDs (but not to either selective COX-2 inhibitors or salicylates) was associated with a significant worsening of depression among a sub- set of study participants [249,250].

In the first trial, involving 1,258 depressed patients treated with citalopram for 12 weeks, the rate of remission was significantly lower among those who had taken NSAIDs at least once relative to those who had not (45% versus 55%, OR 0.64, P = 0.0002) [249]. The other trial, involving 1,545 MDD subjects, showed the rate of treatment- resistant depression was significantly higher among those taking NSAIDs (OR 1.55, 95% CI 1.21 to 2.00) [231]. The worsening of depression in the NSAID groups may not be mechanistically linked to NSAID therapy but instead re- lated to co-existing chronic medical conditions [10,12-18] that necessitate long-term NSAIDs and which are known to be independently associated with increased risk of treatment-resistant depression [249,251]. Future studies investigating the impact of NSAIDs on depression and response to antidepressants in humans are needed.

In other experimental studies utilizing acute-stress paradigms to induce a depression-like state in mice, citalopram increased TNF-?, IFN-?, and p11 (molecular factor linked to depressive behavior in animals) in the frontal cortex, while the NSAID ibuprofen decreased these molecules; NSAIDs also attenuated the antidepressant effects of SSRIs but not other antidepressants [249]. These findings suggest that proinflammatory cytokines may paradoxically exert antidepressant effects despite overwhelming evidence from�human studies to the contrary (as reviewed above), which can be attenuated by NSAIDs [249]. At least two considerations may account for this apparent paradox: 1) under some experimental conditions, proinflammatory cytokines have been associated with a neuroprotective role, [251; (for�example, IFN-? in low levels can induce neuroprotective microglia (Figure 2) [163,166,251]); and 2) whether these responses observed in the context of an acute stress paradigm in an animal model are applicable to endogenous MDD in humans remains unclear [251].

The therapeutic effects of COX-2 inhibitors in psychiatric disorders may involve modulation of biosynthesis of COX-2-derived prostaglandins, including proinflammatory PGE2 and antiinflammatory 15-deoxy-?12,14-PGJ2 (15d- PGJ2) [252,253]. COX-2 inhibitors can reduce PGE2- mediated inflammation, which may contribute to the pathophysiology of psychiatric disorders [252,253]. They may also alter the levels 15d-PGJ2, and the activity of its nuclear receptor peroxisome proliferator-activated nuclear receptor gamma (PPAR-?) [252,253].

Several studies suggest that 15d-PGJ2 and its nuclear receptor PPAR-? can serve as biological markers for schizophrenia [253]. In schizophrenic patients, serum PGE2 levels are increased, whereas serum levels of 15d- PGJ2 are decreased, as is the expression of its nuclear receptor PPAR-? in PBMC [252]. While COX-2 inhibitors may limit the potentially beneficial antiinflammatory effects of the COX-2�dependent �15d-PGJ2/PPAR-? path- way�, they may advantageously reduce its harmful effects, including 1) the increased risk for myocardial infarction and certain infections (for example, cytomegalovirus and Toxoplasma gondii) in schizophrenic patients [254] and 2) its pro-apoptotic effects observed in human and ani- mal cancer tissue [255]. Other potential mechanisms of COX-2 inhibitors therapeutic effects may involve their ability to reduce proinflammatory cytokine levels [163], limit quinolinic acid excitotoxicity (as in MDD) and de- crease KYNA levels (as in schizophrenia) [128].

Minocycline can be effective in psychiatric disorders (Table 3) [248]. In vitro data suggest that minocycline inhibits MAP, cytokine secretion,�COX-2/PGE-2 expression,� and inducible nitric oxide synthase [256]. Minocycline may also counteract dysregulated glutamatergic and dopaminergic neurotransmission [256].

Omega-3 fatty acid effectiveness in psychiatric disorders is unclear [248]. In a 2011 meta-analysis of 15 randomized- controlled trials (916 MDD), omega-3 supplements containing eicosapentaenoic acid ?60% (dose range 200 to 2,200 mg/d in excess of the docosahexaenoic acid dose) significantly decreased depressive symptoms as an adjunctive therapy to SRIs (P <0.001) [246]. A subsequent meta- analysis, however, concluded that there is no significant benefit of omega-3 fatty acids in depression and that the purported efficacy is merely a result of publication bias [247]. A 2012 meta-analysis of 5 randomized-controlled trials including 291 BPD participants found that depressive, but not manic, symptoms were significantly improved among those randomized to omega-3 fatty acids relative to those taking placebo (Hedges g 0.34, P = 0.025) [257]. In a randomized controlled trial of schizophrenic subjects followed up to 12 months, both positive and negative symptom scores were significantly decreased among the 66 participants randomized to long-chain omega-3 (1.2 g/day for 12 weeks; P = 0.02 and 0.01, respectively) [258]; the�authors concluded that omega-3 augmentation during the early course of schizophrenia can also prevent relapses and disease progression [258].

A 2012 meta-analysis of seven randomized-controlled trials assessing omega-3 augmentation in 168 schizo- phrenic patients found no benefit of treatment [259]. The authors of this meta-analysis specifically stated that no conclusion could be drawn regarding the relapse prevention or disease progression endpoints [259]. Experimental data suggest that eicosapentaenoic acid and docosahexaenoic acid mediate their antiinflammatory effects by promoting synthesis of resolvins and protectins, which can inhibit leukocyte infiltration and reduce cytokine production [248].

Neurosteroids, including pregnenolone and its down- stream metabolite allopregnanolone, may have a beneficial role in some psychiatric disorders [248,260]. In MDD, several studies found decreased plasma/CSF allopregnanolone levels correlating with symptom severity, which normalized after successful treatment with certain antidepressants (for example, SSRIs), and electroconvulsive therapy [261]. In schizophrenia, brain pregnenolone levels can be altered [248] and serum allopregnanolone levels may increase after some antipsychotic drugs (for example, clozapine and olanzapine) [260]. In three randomized-controlled trials (100 schizophrenia (pooled); treatment duration, approximately nine weeks) positive, negative, and cognitive symptoms, as well as extrapyramidal side effects of antipsy- chotics were significantly improved in one or more trials among those randomized to pregnenolone relative to those receiving placebo [248]. In one trial, the improvement was sustained with long-term pregnenolone treatment [248]. Pregnenolone can regulate cognition and behavior by potentiating the function of NMDA and GABAA receptors [248]. Furthermore, allopregnanolone may exert neuroprotective and antiinflammatory effects [248]. More RCT studies are needed to confirm the beneficial role of neuroactive steroids in early-onset psychiatric disorders in humans.

We are awaiting the results of several ongoing clinical trials investigating the therapeutic effects of other anti-inflammatory agents, including salicylate, an inhibitor of NF-?B (NCT01182727); acetylsalicylic acid (NCT01320982); pravastatin (NCT1082588); and dextromethorphan, a non-competitive NMDAR antagonist that can limit inflammation-induced dopaminergic neuronal injury (NCT01189006).

Future Treatment Strategies

Although current immune therapies (for example, IVIG, plasmapheresis, corticosteroids and immunosuppressive agents) are often effective for treating autoimmune encephalitides wherein inflammation is acute, intense and predominately of adaptive origin, their efficacy in classical psychiatric disorders wherein inflammation is chronic,�much milder, and predominately of innate origin, is limited [2]. Development of novel therapeutics should aim at reversing glial loss [46,138], down-regulating harmful MAP, while optimizing endogenous neuroprotective T regs and beneficial MAP, rather than indiscriminately sup- pressing inflammation as occurs with current immunosuppressive agents. Additionally, development of potent co-adjuvant antioxidants that would reverse oxidative injury in psychiatric disorders is needed.

Conclusions

Autoimmunity can cause a host of neuropsychiatric disorders that may initially present with isolated psychiatric symptoms. Innate inflammation/autoimmunity may be relevant to the pathogenesis of psychiatric symptoms in a subset of patients with classical psychiatric disorders. Innate inflammation may be mechanistically linked to the traditional monoaminergic and glutamatergic abnormalities and increased oxidative injury reported in psychiatric illnesses.

Souhel Najjar1,5*, Daniel M Pearlman2,5, Kenneth Alper4, Amanda Najjar3 and Orrin Devinsky1,4,5

Abbreviations

3-OH-KYN: 3-hydroxy-kynurenine; ?7nAchR: Alpha 7 nicotinic acetylcholine receptors; AMPAR: Amino-3-hydroxy-5-methyl-l-4-isoxazolepropionic acid receptors; APC: Antigen presenting cell; BBB: Blood�brain barrier;
BH4: Tetrahydrobiopterin; BPD: Bipolar disorder; CI: Confidence interval;
CNS: Central nervous system; COX-2: Cyclooxegenase-2; CSF: Cerebrospinal fluid; DSM-IV: Diagnostic and Statistical Manual of Mental Disorders 4th Edition; EAATs: Excitatory amino acid transporters; eNOS: Endothelial nitric oxide synthase; GABAB: Gamma aminobutyric acid-beta; GAD: Glutamic acid decarboxylase; GFAP: Glial fibrillary acidic protein; GLX: 1H MRS detectable glutamate, glutamine, gamma aminobutyric acid composite;
IDO: Indoleamine 2,3-dioxygenase; Ig: Immunoglobulin; IL: Interleukin; IL-1RA: Interleukin 1 receptor antagonist; IFN-?: Interferon gamma;
KAT: Kynurenine aminotransferase; KMO: Kynurenine 3-monooxygenase; KYN: Kynurenine; KYNA: Kynurenic acid; LE: Limbic encephalitis;
LPS: Lipopolysaccharide; MAP: Microglial activation and proliferation;
MDD: Major depressive disorder; mGluR: Metabotropic glutamate receptor; MHC: II Major histocompatibility complex class two; MRI: Magnetic resonance imaging; MRS: Magnetic resonance spectroscopy; NF-?B: Nuclear factor kappa B; NMDAR: N-methyl-D-aspartate receptor; NR1: Glycine site;
OCD: Obsessive-compulsive disorder; OR: Odds ratio; PANDAS: Pediatric neuropsychiatric autoimmune disorders associated with streptococcal infections; PBMC: Peripheral blood mononuclear cells; PET: Positron emission tomography; PFC: Prefrontal cortex; PGE-2: Prostaglandin E2; PPAR-
?: Peroxisome proliferator-activated nuclear receptor gamma; QA: Quinolinic acid; RNS: Reactive nitrogen species; ROS: Reactive oxygen species;
sIL: Soluble interleukin; SLE: Systemic lupus erythematosus; SRI: Serotonin reuptake inhibitor; TNF-?: Tumor necrosis factor alpha; T-regs: CD4+CD25 +FOXP3+ T regulatory cells; TDO: Tryptophan-2,3-dioxygenase; Th: T-helper; VGKC: Voltage-gated potassium channel; XAG-: Glutamate aspartate transporter; Xc-: Sodium-independent astroglial glutamate/cystine
antiporter system

Competing Interests

The authors declare that they have no competing interests.

Authors��Contributions
SN and DMP performed an extensive literature review, interpreted data, prepared the manuscript, figures, and tables. KA prepared the section pertaining to oxidative mechanisms and contributed to the manuscript revisions. AN and OD critically-revised and improved the design and quality of the manuscript. All authors read and approved the final manuscript.

Acknowledgments

We gratefully acknowledge Drs. Josep Dalmau, MD, PhD, Tracy Butler, MD, and David Zazag, MD, PhD, for providing their expertise in autoimmune encephalitides, neuroinflammation imaging, and neuropathology, respectively.

Author�Details

1Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA. 2Geisel School of Medicine at Dartmouth, The Dartmouth Institute for Health Policy and Clinical Practice, 30 Lafayette Street, HB 7252, Lebanon, NH 03766, USA. 3Department of Pathology, Division of Neuropathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA. 4Department of Psychiatry, New York University School of Medicine, New York, NY, USA. 5New York University Comprehensive Epilepsy Center, 550 First Avenue, New York, NY 10016, USA.

Blank
References:

1. Kayser MS, Dalmau J: The emerging link between autoimmune disorders
and neuropsychiatric disease. J Neuropsychiatry Clin Neurosci 2011, 23:90�97.
2. Najjar S, Pearlman D, Zagzag D, Golfinos J, Devinsky O: Glutamic acid
decarboxylase autoantibody syndrome presenting as schizophrenia.
Neurologist 2012, 18:88�91.
3. Graus F, Saiz A, Dalmau J: Antibodies and neuronal autoimmune
disorders of the CNS. J Neurol 2010, 257:509�517.
4. Lennox BR, Coles AJ, Vincent A: Antibody-mediated encephalitis: a
treatable cause of schizophrenia. Br J Psychiatry 2012, 200:92�94.
5. Zandi MS, Irani SR, Lang B, Waters P, Jones PB, McKenna P, Coles AJ, Vincent
A, Lennox BR: Disease-relevant autoantibodies in first episode
schizophrenia. J Neurol 2011, 258:686�688.
6. Bataller L, Kleopa KA, Wu GF, Rossi JE, Rosenfeld MR, Dalmau J:
Autoimmune limbic encephalitis in 39 patients: immunophenotypes and
outcomes. J Neurol Neurosurg Psychiatry 2007, 78:381�385.
7. Dale RC, Heyman I, Giovannoni G, Church AW: Incidence of anti-brain
antibodies in children with obsessive-compulsive disorder. Br J Psychiatry
2005, 187:314�319.
8. Kendler KS: The dappled nature of causes of psychiatric illness: replacing
the organic-functional/hardware-software dichotomy with empirically
based pluralism. Mol Psychiatry 2012, 17:377�388.
9. Keskin G, Sunter G, Midi I, Tuncer N: Neurosyphilis as a cause of cognitive
decline and psychiatric symptoms at younger age. J Neuropsychiatry Clin
Neurosci 2011, 23:E41�E42.
10. Leboyer M, Soreca I, Scott J, Frye M, Henry C, Tamouza R, Kupfer DJ: Can
bipolar disorder be viewed as a multi-system inflammatory disease?
J Affect Disord 2012, 141:1�10.
11. Hackett ML, Yapa C, Parag V, Anderson CS: Frequency of depression after
stroke: a systematic review of observational studies. Stroke 2005, 36:1330�1340.
12. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW: From
inflammation to sickness and depression: when the immune system
subjugates the brain. Nat Rev Neurosci 2008, 9:46�56.
13. Laske C, Zank M, Klein R, Stransky E, Batra A, Buchkremer G, Schott K:
Autoantibody reactivity in serum of patients with major depression,
schizophrenia and healthy controls. Psychiatry Res 2008, 158:83�86.
14. Eisenberger NI, Berkman ET, Inagaki TK, Rameson LT, Mashal NM, Irwin MR:
Inflammation-induced anhedonia: endotoxin reduces ventral striatum
responses to reward. Biol Psychiatry 2010, 68:748�754.
15. Haroon E, Raison CL, Miller AH: Psychoneuroimmunology meets
neuropsychopharmacology: translational implications of the impact of
inflammation on behavior. Neuropsychopharmacology 2012, 37:137�162.
16. Benros ME, Nielsen PR, Nordentoft M, Eaton WW, Dalton SO, Mortensen PB:
Autoimmune diseases and severe infections as risk factors for
schizophrenia: a 30-year population-based register study. Am J Psychiatry
2011, 168:1303�1310.
17. McNally L, Bhagwagar Z, Hannestad J: Inflammation, glutamate, and glia
in depression: a literature review. CNS Spectr 2008, 13:501�510.
18. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD:
Inflammation causes mood changes through alterations in subgenual
cingulate activity and mesolimbic connectivity. Biol Psychiatry 2009,
66:407�414.19. Raison CL, Miller AH: Is depression an inflammatory disorder?
Curr Psychiatry Rep 2011, 13:467�475.
20. Raison CL, Miller AH: The evolutionary significance of depression in
Pathogen Host Defense (PATHOS-D). Mol Psychiatry 2013, 18:15�37.
21. Steiner J, Bogerts B, Sarnyai Z, Walter M, Gos T, Bernstein HG, Myint AM:
Bridging the gap between the immune and glutamate hypotheses of
schizophrenia and major depression: Potential role of glial NMDA
receptor modulators and impaired blood�brain barrier integrity. World J
Biol Psychiatry 2012, 13:482�492.
22. Steiner J, Mawrin C, Ziegeler A, Bielau H, Ullrich O, Bernstein HG, Bogerts B:
Distribution of HLA-DR-positive microglia in schizophrenia reflects
impaired cerebral lateralization. Acta Neuropathol 2006, 112:305�316.
23. Papakostas GI, Shelton RC, Kinrys G, Henry ME, Bakow BR, Lipkin SH, Pi B,
Thurmond L, Bilello JA: Assessment of a multi-assay, serum-based
biological diagnostic test for major depressive disorder: a pilot and
replication study. Mol Psychiatry 2013, 18:332�339.
24. Krishnan R: Unipolar depression in adults: epidemiology, pathogenesis, and
neurobiology. In UpToDate. Edited by Basow DS. Waltham, MA: UpToDate; 2013.
25. Stovall J: Bipolar disorder in adults: epidemiology and diagnosis. In
UpToDate. Edited by Basow DS. UpToDate: Waltham; 2013.
26. Fischer BA, Buchanan RW: Schizophrenia: epidemiology and pathogenesis.
In UpToDate. Edited by Basow DS. Waltham, MA: UpToDate; 2013.
27. Nestadt G, Samuels J, Riddle M, Bienvenu OJ 3rd, Liang KY, LaBuda M,
Walkup J, Grados M, Hoehn-Saric R: A family study of obsessivecompulsive
disorder. Arch Gen Psychiatry 2000, 57:358�363.
28. Stefansson H, Ophoff RA, Steinberg S, Andreassen OA, Cichon S, Rujescu D,
Werge T, Pietilainen OP, Mors O, Mortensen PB, Sigurdsson E, Gustafsson O,
Nyegaard M, Tuulio-Henriksson A, Ingason A, Hansen T, Suvisaari J,
Lonnqvist J, Paunio T, B�rglum AD, Hartmann A, Fink-Jensen A, Nordentoft
M, Hougaard D, Norgaard-Pedersen B, B�ttcher Y, Olesen J, Breuer R, M�ller
HJ, Giegling I, et al: Common variants conferring risk of schizophrenia.
Nature 2009, 460:744�747.
29. M�ller N, Schwarz MJ: The immune-mediated alteration of serotonin and
glutamate: towards an integrated view of depression. Mol Psychiatry
2007, 12:988�1000.
30. Galecki P, Florkowski A, Bienkiewics M, Szemraj J: Functional polymorphism
of cyclooxygenase-2 gene (G-765C) in depressive patients.
Neuropsychobiology 2010, 62:116�120.
31. Levinson DF: The genetics of depression: a review. Biol Psychiatry 2006,
60:84�92.
32. Zhai J, Cheng L, Dong J, Shen Q, Zhang Q, Chen M, Gao L, Chen X, Wang K,
Deng X, Xu Z, Ji F, Liu C, Li J, Dong Q, Chen C: S100B gene
polymorphisms predict prefrontal spatial function in both schizophrenia
patients and healthy individuals. Schizophr Res 2012, 134:89�94.
33. Zhai J, Zhang Q, Cheng L, Chen M, Wang K, Liu Y, Deng X, Chen X, Shen Q,
Xu Z, Ji F, Liu C, Dong Q, Chen C, Li J: Risk variants in the S100B gene,
associated with elevated S100B levels, are also associated with
visuospatial disability of schizophrenia. Behav Brain Res 2011, 217:363�368.
34. Cappi C, Muniz RK, Sampaio AS, Cordeiro Q, Brentani H, Palacios SA,
Marques AH, Vallada H, Miguel EC, Guilherme L, Hounie AG: Association
study between functional polymorphisms in the TNF-alpha gene and
obsessive-compulsive disorder. Arq Neuropsiquiatr 2012, 70:87�90.
35. Miguel-Hidalgo JJ, Baucom C, Dilley G, Overholser JC, Meltzer HY,
Stockmeier CA, Rajkowska G: Glial fibrillary acidic protein
immunoreactivity in the prefrontal cortex distinguishes younger from
older adults in major depressive disorder. Biol Psychiatry 2000, 48:861�873.
36. Altshuler LL, Abulseoud OA, Foland Ross L, Bartzokis G, Chang S, Mintz J,
Hellemann G, Vinters HV: Amygdala astrocyte reduction in subjects with
major depressive disorder but not bipolar disorder. Bipolar Disord 2010,
12:541�549.
37. Webster MJ, Knable MB, Johnston-Wilson N, Nagata K, Inagaki M, Yolken RH:
Immunohistochemical localization of phosphorylated glial fibrillary acidic
protein in the prefrontal cortex and hippocampus from patients with
schizophrenia, bipolar disorder, and depression. Brain Behav Immun 2001,
15:388�400.
38. Doyle C, Deakin JFW: Fewer astrocytes in frontal cortex in schizophrenia,
depression and bipolar disorder. Schizophrenia Res 2002, 53:106.
39. Johnston-Wilson NL, Sims CD, Hofmann JP, Anderson L, Shore AD, Torrey
EF, Yolken RH: Disease-specific alterations in frontal cortex brain proteins
in schizophrenia, bipolar disorder, and major depressive disorder, The
Stanley Neuropathology Consortium. Mol Psychiatry 2000, 5:142�149.
40. Gosselin RD, Gibney S, O’Malley D, Dinan TG, Cryan JF: Region specific
decrease in glial fibrillary acidic protein immunoreactivity in the brain of
a rat model of depression. Neuroscience 2009, 159:915�925.
41. Banasr M, Duman RS: Glial loss in the prefrontal cortex is sufficient to
induce depressive-like behaviors. Biol Psychiatry 2008, 64:863�870.
42. Cotter D, Hudson L, Landau S: Evidence for orbitofrontal pathology in
bipolar disorder and major depression, but not in schizophrenia.
Bipolar Disord 2005, 7:358�369.
43. Brauch RA, Adnan El-Masri M, Parker J Jr, El-Mallakh RS: Glial cell number
and neuron/glial cell ratios in postmortem brains of bipolar individuals.
J Affect Disord 2006, 91:87�90.
44. Cotter DR, Pariante CM, Everall IP: Glial cell abnormalities in major
psychiatric disorders: the evidence and implications. Brain Res Bull 2001,
55:585�595.
45. Cotter D, Mackay D, Landau S, Kerwin R, Everall I: Reduced glial cell density
and neuronal size in the anterior cingulate cortex in major depressive
disorder. Arch Gen Psychiatry 2001, 58:545�553.
46. Bowley MP, Drevets WC, Ong�r D, Price JL: Low glial numbers in the
amygdala in major depressive disorder. Biol Psychiatry 2002, 52:404�412.
47. Toro CT, Hallak JE, Dunham JS, Deakin JF: Glial fibrillary acidic protein and
glutamine synthetase in subregions of prefrontal cortex in schizophrenia
and mood disorder. Neurosci Lett 2006, 404:276�281.
48. Rajkowska G, Miguel-Hidalgo JJ, Makkos Z, Meltzer H, Overholser J,
Stockmeier C: Layer-specific reductions in GFAP-reactive astroglia in the
dorsolateral prefrontal cortex in schizophrenia. Schizophr Res 2002, 57:127�138.
49. Steffek AE, McCullumsmith RE, Haroutunian V, Meador-Woodruff JH: Cortical
expression of glial fibrillary acidic protein and glutamine synthetase is
decreased in schizophrenia. Schizophr Res 2008, 103:71�82.
50. Damadzic R, Bigelow LB, Krimer LS, Goldenson DA, Saunders RC, Kleinman
JE, Herman MM: A quantitative immunohistochemical study of astrocytes in
the entorhinal cortex in schizophrenia, bipolar disorder and major
depression: absence of significant astrocytosis. Brain Res Bull 2001, 55:611�618.
51. Benes FM, McSparren J, Bird ED, SanGiovanni JP, Vincent SL: Deficits in
small interneurons in prefrontal and cingulate cortices of schizophrenic
and schizoaffective patients. Arch Gen Psychiatry 1991, 48:996�1001.
52. M�ller N, Schwarz MJ: Immune system and schizophrenia. Curr Immunol
Rev 2010, 6:213�220.
53. Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai Z, Mawrin C,
Brisch R, Bielau H, Meyer Zu Schwabedissen L, Bogerts B, Myint AM: Severe
depression is associated with increased microglial quinolinic acid in
subregions of the anterior cingulate gyrus: evidence for an immunemodulated
glutamatergic neurotransmission? J Neuroinflammation 2011, 8:94.
54. Vostrikov VM, Uranova NA, Orlovskaya DD: Deficit of perineuronal
oligodendrocytes in the prefrontal cortex in schizophrenia and mood
disorders. Schizophr Res 2007, 94:273�280.
55. Rajkowska G, Miguel-Hidalgo JJ: Gliogenesis and glial pathology in
depression. CNS Neurol Disord Drug Targets 2007, 6:219�233.
56. Uranova NA, Vostrikov VM, Orlovskaya DD, Rachmanova VI:
Oligodendroglial density in the prefrontal cortex in schizophrenia and
mood disorders: a study from the Stanley Neuropathology Consortium.
Schizophr Res 2004, 67:269�275.
57. Uranova N: Damage and loss of oligodendrocytes are crucial in the
pathogenesis of schizophrenia and mood disorders (findings form
postmortem studies). Neuropsychopharmacology 2004, 29:S33.
58. Uranova NA, Orlovskaya DD, Vostrikov VM, Rachmanova VI: Decreased
density of oligodendroglial satellites of pyramidal neurons in layer III in
the prefrontal cortex in schizophrenia and mood disorders. Schizophr Res
2002, 53:107.
59. Vostrikov VM, Uranova NA, Rakhmanova VI, Orlovskaia DD: Lowered
oligodendroglial cell density in the prefrontal cortex in schizophrenia.
Zh Nevrol Psikhiatr Im S S Korsakova 2004, 104:47�51.
60. Uranova NA, Zimina IS, Vikhreva OV, Krukov NO, Rachmanova VI, Orlovskaya
DD: Ultrastructural damage of capillaries in the neocortex in
schizophrenia. World J Biol Psychiatry 2010, 11:567�578.
61. Hof PR, Haroutunian V, Friedrich VL Jr, Byne W, Buitron C, Perl DP, Davis KL:
Loss and altered spatial distribution of oligodendrocytes in the superior
frontal gyrus in schizophrenia. Biol Psychiatry 2003, 53:1075�1085.
62. Davis KL, Stewart DG, Friedman JI, Buchsbaum M, Harvey PD, Hof PR,
Buxbaum J, Haroutunian V: White matter changes in schizophrenia:
evidence for myelin-related dysfunction. Arch Gen Psychiatry 2003,
60:443�456.63. Flynn SW, Lang DJ, Mackay AL, Goghari V, Vavasour IM, Whittall KP, Smith
GN, Arango V, Mann JJ, Dwork AJ, Falkai P, Honer WG: Abnormalities of
myelination in schizophrenia detected in vivo with MRI, and postmortem
with analysis of oligodendrocyte proteins. Mol Psychiatry 2003,
8:811�820.
64. Uranova NA, Vostrikov VM, Vikhreva OV, Zimina IS, Kolomeets NS, Orlovskaya
DD: The role of oligodendrocyte pathology in schizophrenia. Int J
Neuropsychopharmacol 2007, 10:537�545.
65. Byne W, Kidkardnee S, Tatusov A, Yiannoulos G, Buchsbaum MS,
Haroutunian V: Schizophrenia-associated reduction of neuronal and
oligodendrocyte numbers in the anterior principal thalamic nucleus.
Schizophr Res 2006, 85:245�253.
66. Hamidi M, Drevets WC, Price JL: Glial reduction in amygdala in major
depressive disorder is due to oligodendrocytes. Biol Psychiatry 2004,
55:563�569.
67. Bayer TA, Buslei R, Havas L, Falkai P: Evidence for activation of microglia in
patients with psychiatric illnesses. Neurosci Lett 1999, 271:126�128.
68. Steiner J, Bielau H, Brisch R, Danos P, Ullrich O, Mawrin C, Bernstein HG,
Bogerts B: Immunological aspects in the neurobiology of suicide:
elevated microglial density in schizophrenia and depression is
associated with suicide. J Psychiatr Res 2008, 42:151�157.
69. Rao JS, Harry GJ, Rapoport SI, Kim HW: Increased excitotoxicity and
neuroinflammatory markers in postmortem frontal cortex from bipolar
disorder patients. Mol Psychiatry 2010, 15:384�392.
70. Bernstein HG, Steiner J, Bogerts B: Glial cells in schizophrenia:
pathophysiological significance and possible consequences for therapy.
Expert Rev Neurother 2009, 9:1059�1071.
71. Chen SK, Tvrdik P, Peden E, Cho S, Wu S, Spangrude G, Capecchi MR:
Hematopoietic origin of pathological grooming in Hoxb8 mutant mice.
Cell 2010, 141:775�785.
72. Antony JM: Grooming and growing with microglia. Sci Signal 2010, 3:jc8.
73. Wonodi I, Stine OC, Sathyasaikumar KV, Roberts RC, Mitchell BD, Hong LE,
Kajii Y, Thaker GK, Schwarcz R: Downregulated kynurenine 3-
monooxygenase gene expression and enzyme activity in schizophrenia
and genetic association with schizophrenia endophenotypes. Arch Gen
Psychiatry 2011, 68:665�674.
74. Raison CL, Lowry CA, Rook GA: Inflammation, sanitation, and
consternation: loss of contact with coevolved, tolerogenic
microorganisms and the pathophysiology and treatment of major
depression. Arch Gen Psychiatry 2010, 67:1211�1224.
75. Drexhage RC, Hoogenboezem TH, Versnel MA, Berghout A, Nolen WA,
Drexhage HA: The activation of monocyte and T cell networks in patients
with bipolar disorder. Brain Behav Immun 2011, 25:1206�1213.
76. Steiner J, Jacobs R, Panteli B, Brauner M, Schiltz K, Bahn S, Herberth M,
Westphal S, Gos T, Walter M, Bernstein HG, Myint AM, Bogerts B: Acute
schizophrenia is accompanied by reduced T cell and increased B cell
immunity. Eur Arch Psychiatry Clin Neurosci 2010, 260:509�518.
77. Rotge JY, Aouizerate B, Tignol J, Bioulac B, Burbaud P, Guehl D: The
glutamate-based genetic immune hypothesis in obsessive-compulsive
disorder, An integrative approach from genes to symptoms.
Neuroscience 2010, 165:408�417.
78. Y�ksel C, Ong�r D: Magnetic resonance spectroscopy studies of
glutamate-related abnormalities in mood disorders. Biol Psychiatry 2010,
68:785�794.
79. Rao JS, Kellom M, Reese EA, Rapoport SI, Kim HW: Dysregulated glutamate
and dopamine transporters in postmortem frontal cortex from bipolar
and schizophrenic patients. J Affect Disord 2012, 136:63�71.
80. Bauer D, Gupta D, Harotunian V, Meador-Woodruff JH, McCullumsmith RE:
Abnormal expression of glutamate transporter and transporter
interacting molecules in prefrontal cortex in elderly patients with
schizophrenia. Schizophr Res 2008, 104:108�120.
81. Matute C, Melone M, Vallejo-Illarramendi A, Conti F: Increased expression
of the astrocytic glutamate transporter GLT-1 in the prefrontal cortex of
schizophrenics. 2005, 49:451�455.
82. Smith RE, Haroutunian V, Davis KL, Meador-Woodruff JH: Expression of
excitatory amino acid transporter transcripts in the thalamus of subjects
with schizophrenia. Am J Psychiatry 2001, 158:1393�1399.
83. McCullumsmith RE, Meador-Woodruff JH: Striatal excitatory amino acid
transporter transcript expression in schizophrenia, bipolar disorder,
and major depressive disorder. Neuropsychopharmacology 2002,
26:368�375.
84. Pittenger C, Bloch MH, Williams K: Glutamate abnormalities in obsessive
compulsive disorder: neurobiology, pathophysiology, and treatment.
Pharmacol Ther 2011, 132:314�332.
85. Hashimoto K: Emerging role of glutamate in the pathophysiology of
major depressive disorder. Brain Res Rev 2009, 61:105�123.
86. Hashimoto K, Sawa A, Iyo M: Increased levels of glutamate in brains from
patients with mood disorders. Biol Psychiatry 2007, 62:1310�1316.
87. Burbaeva G, Boksha IS, Turishcheva MS, Vorobyeva EA, Savushkina OK,
Tereshkina EB: Glutamine synthetase and glutamate dehydrogenase in
the prefrontal cortex of patients with schizophrenia. Prog
Neuropsychopharmacol Biol Psychiatry 2003, 27:675�680.
88. Bhattacharyya S, Khanna S, Chakrabarty K, Mahadevan A, Christopher R,
Shankar SK: Anti-brain autoantibodies and altered excitatory
neurotransmitters in obsessive-compulsive disorder.
Neuropsychopharmacology 2009, 34:2489�2496.
89. Sanacora G, Gueorguieva R, Epperson CN, Wu YT, Appel M, Rothman DL,
Krystal JH, Mason GF: Subtype-specific alterations of gammaaminobutyric
acid and glutamate in patients with major depression.
Arch Gen Psychiatry 2004, 61:705�713.
90. Marsman A, van den Heuvel MP, Klomp DW, Kahn RS, Luijten PR, Hulshoff
Pol HE: Glutamate in schizophrenia: a focused review and meta-analysis
of 1H-MRS studies. Schizophr Bull 2013, 39:120�129.
91. Liu Y, Ho RC, Mak A: Interleukin (IL)-6, tumour necrosis factor alpha
(TNF-alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in
patients with major depressive disorder: a meta-analysis and metaregression.
J Affect Disord 2012, 139:230�239.
92. Brietzke E, Stabellini R, Grassis-Oliveira R, Lafer B: Cytokines in bipolar
disorder: recent findings, deleterious effects but promise for future
therapeutics. CNS Spectr 2011. www.cnsspectrums.com/aspx/
articledetail.aspx?articleid=3596.
93. Denys D, Fluitman S, Kavelaars A, Heijnen C, Westenberg H: Decreased
TNF-alpha and NK activity in obsessive-compulsive disorder.
Psychoneuroendocrinology 2004, 29:945�952.
94. Brambilla F, Perna G, Bellodi L, Arancio C, Bertani A, Perini G, Carraro C, Gava
F: Plasma interleukin-1 beta and tumor necrosis factor concentrations in
obsessive-compulsive disorders. Biol Psychiatry 1997, 42:976�981.
95. Fluitman S, Denys D, Vulink N, Schutters S, Heijnen C, Westenberg H:
Lipopolysaccharide-induced cytokine production in obsessivecompulsive
disorder and generalized social anxiety disorder. Psychiatry
Res 2010, 178:313�316.
96. Janelidze S, Mattei D, Westrin A, Traskman-Bendz L, Brundin L: Cytokine
levels in the blood may distinguish suicide attempters from depressed
patients. Brain Behav Immun 2011, 25:335�339.
97. Postal M, Costallat LT, Appenzeller S: Neuropsychiatric manifestations in
systemic lupus erythematosus: epidemiology, pathophysiology and
management. CNS Drugs 2011, 25:721�736.
98. Kozora E, Hanly JG, Lapteva L, Filley CM: Cognitive dysfunction in
systemic lupus erythematosus: past, present, and future.
Arthritis Rheum 2008, 58:3286�3298.
99. Lancaster E, Martinez-Hernandez E, Dalmau J: Encephalitis and antibodies to
synaptic and neuronal cell surface proteins. Neurology 2011, 77:179�189.
100. Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon
R: Clinical experience and laboratory investigations in patients with antiNMDAR
encephalitis. Lancet Neurol 2011, 10:63�74.
101. Lai M, Huijbers MG, Lancaster E, Graus F, Bataller L, Balice-Gordon R, Cowell
JK, Dalmau J: Investigation of LGI1 as the antigen in limbic encephalitis
previously attributed to potassium channels: a case series. Lancet Neurol
2010, 9:776�785.
102. Lancaster E, Huijbers MG, Bar V, Boronat A, Wong A, Martinez-Hernandez E,
Wilson C, Jacobs D, Lai M, Walker RW, Graus F, Bataller L, Illa I, Markx S, Strauss
KA, Peles E, Scherer SS, Dalmau J: Investigations of caspr2, an autoantigen of
encephalitis and neuromyotonia. Ann Neurol 2011, 69:303�311.
103. Lancaster E, Lai M, Peng X, Hughes E, Constantinescu R, Raizer J, Friedman
D, Skeen MB, Grisold W, Kimura A, Ohta K, Iizuka T, Guzman M, Graus F,
Moss SJ, Balice-Gordon R, Dalmau J: Antibodies to the GABA(B) receptor in
limbic encephalitis with seizures: case series and characterisation of the
antigen. Lancet Neurol 2010, 9:67�76.
104. Lancaster E, Martinez-Hernandez E, Titulaer MJ, Boulos M, Weaver S, Antoine
JC, Liebers E, Kornblum C, Bien CG, Honnorat J, Wong S, Xu J, Contractor A,
Balice-Gordon R, Dalmau J: Antibodies to metabotropic glutamate
receptor 5 in the Ophelia syndrome. Neurology 2011, 77:1698�1701.105. Ances BM, Vitaliani R, Taylor RA, Liebeskind DS, Voloschin A, Houghton DJ,
Galetta SL, Dichter M, Alavi A, Rosenfeld MR, Dalmau J: Treatmentresponsive
limbic encephalitis identified by neuropil antibodies: MRI and
PET correlates. Brain 2005, 128:1764�1777.
106. Tofaris GK, Irani SR, Cheeran BJ, Baker IW, Cader ZM, Vincent A:
Immunotherapy-responsive chorea as the presenting feature of LGI1-
antibody encephalitis. Neurology 2012, 79:195�196.
107. Najjar S, Pearlman D, Najjar A, Ghiasian V, Zagzag D, Devinsky O:
Extralimbic autoimmune encephalitis associated with glutamic acid
decarboxylase antibodies: an underdiagnosed entity? Epilepsy Behav
2011, 21:306�313.
108. Titulaer MJ, McCracken L, Gabilondo I, Armangue T, Glaser C, Iizuka T, Honig
LS, Benseler SM, Kawachi I, Martinez-Hernandez E, Aguilar E, Gresa-Arribas N,
Ryan-Florance N, Torrents A, Saiz A, Rosenfeld MR, Balice-Gordon R, Graus F,
Dalmau J: Treatment and prognostic factors for long-term outcome in
patients with anti-NMDA receptor encephalitis: an observational cohort
study. Lancet Neurol 2013, 12:157�165.
109. Dalmau J, Gleichman AJ, Hughes EG, Rossi JE, Peng X, Lai M, Dessain SK,
Rosenfeld MR, Balice-Gordon R, Lynch DR: Anti-NMDA-receptor
encephalitis: case series and analysis of the effects of antibodies.
Lancet Neurol 2008, 7:1091�1098.
110. Graus F, Boronat A, Xifro X, Boix M, Svigelj V, Garcia A, Palomino A, Sabater
L, Alberch J, Saiz A: The expanding clinical profile of anti-AMPA receptor
encephalitis. Neurology 2010, 74:857�859.
111. Lai M, Hughes EG, Peng X, Zhou L, Gleichman AJ, Shu H, Mata S, Kremens
D, Vitaliani R, Geschwind MD, Bataller L, Kalb RG, Davis R, Graus F, Lynch DR,
Balice-Gordon R, Dalmau J: AMPA receptor antibodies in limbic
encephalitis alter synaptic receptor location. Ann Neurol 2009, 65:424�434.
112. Najjar S, Pearlman D, Devinsky O, Najjar A, Nadkarni S, Butler T, Zagzag D:
Neuropsychiatric autoimmune encephalitis with negative VGKC-complex,
NMDAR, and GAD autoantibodies: a case report and literature review,
forthcoming. Cogn Behav Neurol. in press.
113. Najjar S, Pearlman D, Zagzag D, Devinsky O: Spontaneously resolving
seronegative autoimmune limbic encephalitis. Cogn Behav Neurol 2011,
24:99�105.
114. Gabilondo I, Saiz A, Galan L, Gonzalez V, Jadraque R, Sabater L, Sans A,
Sempere A, Vela A, Villalobos F, Vi�als M, Villoslada P, Graus F: Analysis of
relapses in anti-NMDAR encephalitis. Neurology 2011, 77:996�999.
115. Barry H, Hardiman O, Healy DG, Keogan M, Moroney J, Molnar PP, Cotter
DR, Murphy KC: Anti-NMDA receptor encephalitis: an important
differential diagnosis in psychosis. Br J Psychiatry 2011, 199:508�509.
116. Dickerson F, Stallings C, Vaughan C, Origoni A, Khushalani S, Yolken R:
Antibodies to the glutamate receptor in mania. Bipolar Disord 2012,
14:547�553.
117. O’Loughlin K, Ruge P, McCauley M: Encephalitis and schizophrenia: a
matter of words. Br J Psychiatry 2012, 201:74.
118. Parratt KL, Allan M, Lewis SJ, Dalmau J, Halmagyi GM, Spies JM: Acute
psychiatric illness in a young woman: an unusual form of encephalitis.
Med J Aust 2009, 191:284�286.
119. Suzuki Y, Kurita T, Sakurai K, Takeda Y, Koyama T: Case report of anti-NMDA
receptor encephalitis suspected of schizophrenia. Seishin Shinkeigaku
Zasshi 2009, 111:1479�1484.
120. Tsutsui K, Kanbayashi T, Tanaka K, Boku S, Ito W, Tokunaga J, Mori A,
Hishikawa Y, Shimizu T, Nishino S: Anti-NMDA-receptor antibody detected
in encephalitis, schizophrenia, and narcolepsy with psychotic features.
BMC Psychiatry 2012, 12:37.
121. Van Putten WK, Hachimi-Idrissi S, Jansen A, Van Gorp V, Huyghens L:
Uncommon cause of psychotic behavior in a 9-year-old girl: a case
report. Case Report Med 2012, 2012:358520.
122. Masdeu JC, Gonzalez-Pinto A, Matute C, Ruiz De Azua S, Palomino A, De
Leon J, Berman KF, Dalmau J: Serum IgG antibodies against the NR1
subunit of the NMDA receptor not detected in schizophrenia. Am J
Psychiatry 2012, 169:1120�1121.
123. Kirvan CA, Swedo SE, Kurahara D, Cunningham MW: Streptococcal mimicry
and antibody-mediated cell signaling in the pathogenesis of
Sydenham’s chorea. Autoimmunity 2006, 39:21�29.
124. Swedo SE: Streptoccocal infection, Tourette syndrome, and OCD: is there
a connection? Pandas: Horse or zebra? Neurology 2010, 74:1397�1398.
125. Morer A, Lazaro L, Sabater L, Massana J, Castro J, Graus F: Antineuronal
antibodies in a group of children with obsessive-compulsive disorder
and Tourette syndrome. J Psychiatr Res 2008, 42:64�68.
126. Pavone P, Bianchini R, Parano E, Incorpora G, Rizzo R, Mazzone L, Trifiletti RR:
Anti-brain antibodies in PANDAS versus uncomplicated streptococcal
infection. Pediatr Neurol 2004, 30:107�110.
127. Maina G, Albert U, Bogetto F, Borghese C, Berro AC, Mutani R, Rossi F,
Vigliani MC: Anti-brain antibodies in adult patients with obsessivecompulsive
disorder. J Affect Disord 2009, 116:192�200.
128. Brimberg L, Benhar I, Mascaro-Blanco A, Alvarez K, Lotan D, Winter C, Klein J,
Moses AE, Somnier FE, Leckman JF, Swedo SE, Cunningham MW, Joel D:
Behavioral, pharmacological, and immunological abnormalities after
streptococcal exposure: a novel rat model of Sydenham chorea and
related neuropsychiatric disorders. Neuropsychopharmacology 2012,
37:2076�2087.
129. Dale RC, Candler PM, Church AJ, Wait R, Pocock JM, Giovannoni G:
Neuronal surface glycolytic enzymes are autoantigen targets in
post-streptococcal autoimmune CNS disease. J Neuroimmunol 2006,
172:187�197.
130. Nicholson TR, Ferdinando S, Krishnaiah RB, Anhoury S, Lennox BR, MataixCols
D, Cleare A, Veale DM, Drummond LM, Fineberg NA, Church AJ,
Giovannoni G, Heyman I: Prevalence of anti-basal ganglia antibodies in
adult obsessive-compulsive disorder: cross-sectional study. Br J Psychiatry
2012, 200:381�386.
131. Wu K, Hanna GL, Rosenberg DR, Arnold PD: The role of glutamate
signaling in the pathogenesis and treatment of obsessive-compulsive
disorder. Pharmacol Biochem Behav 2012, 100:726�735.
132. Perlmutter SJ, Leitman SF, Garvey MA, Hamburger S, Feldman E, Leonard
HL, Swedo SE: Therapeutic plasma exchange and intravenous
immunoglobulin for obsessive-compulsive disorder and tic disorders in
childhood. Lancet 1999, 354:1153�1158.
133. Pereira A Jr, Furlan FA: Astrocytes and human cognition: modeling
information integration and modulation of neuronal activity.
Prog Neurobiol 2010, 92:405�420.
134. Barres BA: The mystery and magic of glia: a perspective on their roles in
health and disease. Neuron 2008, 60:430�440.
135. Verkhratsky A, Parpura V, Rodriguez JJ: Where the thoughts dwell: the
physiology of neuronal-glial “diffuse neural net”. Brain Res Rev 2011,
66:133�151.
136. Sofroniew MV: Molecular dissection of reactive astrogliosis and glial scar
formation. Trends Neurosci 2009, 32:638�647.
137. Hamilton NB, Attwell D: Do astrocytes really exocytose neurotransmitters?
Nat Rev Neurosci 2010, 11:227�238.
138. Rajkowska G: Postmortem studies in mood disorders indicate altered
numbers of neurons and glial cells. Biol Psychiatry 2000, 48:766�777.
139. Coupland NJ, Ogilvie CJ, Hegadoren KM, Seres P, Hanstock CC, Allen PS:
Decreased prefrontal Myo-inositol in major depressive disorder.
Biol Psychiatry 2005, 57:1526�1534.
140. Miguel-Hidalgo JJ, Overholser JC, Jurjus GJ, Meltzer HY, Dieter L, Konick L,
Stockmeier CA, Rajkowska G: Vascular and extravascular immunoreactivity
for intercellular adhesion molecule 1 in the orbitofrontal cortex of
subjects with major depression: age-dependent changes. J Affect Disord
2011, 132:422�431.
141. Miguel-Hidalgo JJ, Wei JR, Andrew M, Overholser JC, Jurjus G, Stockmeier
CA, Rajkowska G: Glia pathology in the prefrontal cortex in alcohol
dependence with and without depressive symptoms. Biol Psychiatry 2002,
52:1121�1133.
142. Stockmeier CA, Mahajan GJ, Konick LC, Overholser JC, Jurjus GJ, Meltzer HY,
Uylings HB, Friedman L, Rajkowska G: Cellular changes in the postmortem
hippocampus in major depression. Biol Psychiatry 2004, 56:640�650.
143. Ong�r D, Drevets WC, Price JL: Glial reduction in the subgenual prefrontal
cortex in mood disorders. Proc Natl Acad Sci USA 1998, 95:13290�13295.
144. Gittins RA, Harrison PJ: A morphometric study of glia and neurons in the
anterior cingulate cortex in mood disorder. J Affect Disord 2011,
133:328�332.
145. Cotter D, Mackay D, Beasley C, Kerwin R, Everall I: Reduced glial density
and neuronal volume in major depressive disorder and schizophrenia in
the anterior cingulate cortex [abstract]. Schizophrenia Res 2000, 41:106.
146. Si X, Miguel-Hidalgo JJ, Rajkowska G: GFAP expression is reduced in the
dorsolateral prefrontal cortex in depression. In Society for Neuroscience; 2003.
Neuroscience Meeting Planne: New Orleans; 2003.
147. Legutko B, Mahajan G, Stockmeier CA, Rajkowska G: White matter astrocytes
are reduced in depression. In Society for Neuroscience. Neuroscience Meeting
Planner: Washington, DC; 2011.148. Edgar N, Sibille E: A putative functional role for oligodendrocytes in
mood regulation. Transl Psychiatry 2012, 2:e109.
149. Rajkowska G, Halaris A, Selemon LD: Reductions in neuronal and glial
density characterize the dorsolateral prefrontal cortex in bipolar
disorder. Biol Psychiatry 2001, 49:741�752.
150. Cotter D, Mackay D, Chana G, Beasley C, Landau S, Everall IP: Reduced
neuronal size and glial cell density in area 9 of the dorsolateral
prefrontal cortex in subjects with major depressive disorder. Cereb Cortex
2002, 12:386�394.
151. Stark AK, Uylings HB, Sanz-Arigita E, Pakkenberg B: Glial cell loss in the
anterior cingulate cortex, a subregion of the prefrontal cortex, in
subjects with schizophrenia. Am J Psychiatry 2004, 161:882�888.
152. Konopaske GT, Dorph-Petersen KA, Sweet RA, Pierri JN, Zhang W, Sampson
AR, Lewis DA: Effect of chronic antipsychotic exposure on astrocyte and
oligodendrocyte numbers in macaque monkeys. Biol Psychiatry 2008,
63:759�765.
153. Selemon LD, Lidow MS, Goldman-Rakic PS: Increased volume and glial
density in primate prefrontal cortex associated with chronic
antipsychotic drug exposure. Biol Psychiatry 1999, 46:161�172.
154. Steiner J, Bernstein HG, Bielau H, Farkas N, Winter J, Dobrowolny H, Brisch R,
Gos T, Mawrin C, Myint AM, Bogerts B: S100B-immunopositive glia is
elevated in paranoid as compared to residual schizophrenia: a
morphometric study. J Psychiatr Res 2008, 42:868�876.
155. Carter CJ: eIF2B and oligodendrocyte survival: where nature and nurture
meet in bipolar disorder and schizophrenia? Schizophr Bull 2007,
33:1343�1353.
156. Hayashi Y, Nihonmatsu-Kikuchi N, Hisanaga S, Yu XJ, Tatebayashi Y:
Neuropathological similarities and differences between schizophrenia
and bipolar disorder: a flow cytometric postmortem brain study.
PLoS One 2012, 7:e33019.
157. Uranova NA, Vikhreva OV, Rachmanova VI, Orlovskaya DD: Ultrastructural
alterations of myelinated fibers and oligodendrocytes in the prefrontal
cortex in schizophrenia: a postmortem morphometric study.
Schizophr Res Treatment 2011, 2011:325789.
158. Torres-Platas SG, Hercher C, Davoli MA, Maussion G, Labonte B, Turecki
G, Mechawar N: Astrocytic hypertrophy in anterior cingulate white
matter of depressed suicides. Neuropsychopharmacology 2011,
36:2650�2658.
159. Pereira A Jr, Furlan FA: On the role of synchrony for neuron-astrocyte
interactions and perceptual conscious processing. J Biol Phys 2009,
35:465�480.
160. Kettenmann H, Hanisch UK, Noda M, Verkhratsky A: Physiology of
microglia. Physiol Rev 2011, 91:461�553.
161. Tremblay ME, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A: The role
of microglia in the healthy brain. J Neurosci 2011, 31:16064�16069.
162. Kaindl AM, Degos V, Peineau S, Gouadon E, Chhor V, Loron G, Le
Charpentier T, Josserand J, Ali C, Vivien D, Collingridge GL, Lombet A, Issa L,
Rene F, Loeffler JP, Kavelaars A, Verney C, Mantz J, Gressens P: Activation of
microglial N-methyl-D-aspartate receptors triggers inflammation and
neuronal cell death in the developing and mature brain. Ann Neurol
2012, 72:536�549.
163. Schwartz M, Shaked I, Fisher J, Mizrahi T, Schori H: Protective
autoimmunity against the enemy within: fighting glutamate toxicity.
Trends Neurosci 2003, 26:297�302.
164. Pacheco R, Gallart T, Lluis C, Franco R: Role of glutamate on T-cell
mediated immunity. J Neuroimmunol 2007, 185:9�19.
165. Najjar S, Pearlman D, Miller DC, Devinsky O: Refractory epilepsy associated
with microglial activation. Neurologist 2011, 17:249�254.
166. Schwartz M, Butovsky O, Bruck W, Hanisch UK: Microglial phenotype: is the
commitment reversible? Trends Neurosci 2006, 29:68�74.
167. Wang F, Wu H, Xu S, Guo X, Yang J, Shen X: Macrophage migration
inhibitory factor activates cyclooxygenase 2-prostaglandin E2 in cultured
spinal microglia. Neurosci Res 2011, 71:210�218.
168. Zhang XY, Xiu MH, Song C, Chenda C, Wu GY, Haile CN, Kosten TA, Kosten
TR: Increased serum S100B in never-medicated and medicated
schizophrenic patients. J Psychiatr Res 2010, 44:1236�1240.
169. Kawasaki Y, Zhang L, Cheng JK, Ji RR: Cytokine mechanisms of central
sensitization: distinct and overlapping role of interleukin-1beta,
interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and
neuronal activity in the superficial spinal cord. J Neurosci 2008,
28:5189�5194.
170. M�ller N, Schwarz MJ: The immunological basis of glutamatergic
disturbance in schizophrenia: towards an integrated view. J Neural
Transm Suppl 2007, 72:269�280.
171. Hestad KA, Tonseth S, Stoen CD, Ueland T, Aukrust P: Raised plasma levels
of tumor necrosis factor alpha in patients with depression: normalization
during electroconvulsive therapy. J ECT 2003, 19:183�188.
172. Kubera M, Kenis G, Bosmans E, Zieba A, Dudek D, Nowak G, Maes M:
Plasma levels of interleukin-6, interleukin-10, and interleukin-1 receptor
antagonist in depression: comparison between the acute state and after
remission. Pol J Pharmacol 2000, 52:237�241.
173. Miller BJ, Buckley P, Seabolt W, Mellor A, Kirkpatrick B: Meta-analysis of
cytokine alterations in schizophrenia: clinical status and antipsychotic
effects. Biol Psychiatry 2011, 70:663�671.
174. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E: Inflammatory
cytokine alterations in schizophrenia: a systematic quantitative review.
Biol Psychiatry 2008, 63:801�808.
175. Reale M, Patruno A, De Lutiis MA, Pesce M, Felaco M, Di Giannantonio M, Di
Nicola M, Grilli A: Dysregulation of chemo-cytokine production in
schizophrenic patients versus healthy controls. BMC Neurosci 2011, 12:13.
176. Fluitman SB, Denys DA, Heijnen CJ, Westenberg HG: Disgust affects TNFalpha,
IL-6 and noradrenalin levels in patients with obsessive-compulsive
disorder. Psychoneuroendocrinology 2010, 35:906�911.
177. Konuk N, Tekin IO, Ozturk U, Atik L, Atasoy N, Bektas S, Erdogan A: Plasma
levels of tumor necrosis factor-alpha and interleukin-6 in obsessive
compulsive disorder. Mediators Inflamm 2007, 2007:65704.
178. Monteleone P, Catapano F, Fabrazzo M, Tortorella A, Maj M: Decreased
blood levels of tumor necrosis factor-alpha in patients with obsessivecompulsive
disorder. Neuropsychobiology 1998, 37:182�185.
179. Marazziti D, Presta S, Pfanner C, Gemignani A, Rossi A, Sbrana S, Rocchi V,
Ambrogi F, Cassano GB: Immunological alterations in adult obsessivecompulsive
disorder. Biol Psychiatry 1999, 46:810�814.
180. Zai G, Arnold PD, Burroughs E, Richter MA, Kennedy JL: Tumor necrosis
factor-alpha gene is not associated with obsessive-compulsive disorder.
Psychiatr Genet 2006, 16:43.
181. Rodr�guez AD, Gonz�lez PA, Garc�a MJ, de la Rosa A, Vargas M, Marrero F:
Circadian variations in proinflammatory cytokine concentrations in acute
myocardial infarction. Rev Esp Cardiol 2003, 56:555�560.
182. Oliver JC, Bland LA, Oettinger CW, Arduino MJ, McAllister SK, Aguero SM,
Favero MS: Cytokine kinetics in an in vitro whole blood model following
an endotoxin challenge. Lymphokine Cytokine Res 1993, 12:115�120.
183. Le T, Leung L, Carroll WL, Schibler KR: Regulation of interleukin-10 gene
expression: possible mechanisms accounting for its upregulation and for
maturational differences in its expression by blood mononuclear cells.
Blood 1997, 89:4112�4119.
184. Lee MC, Ting KK, Adams S, Brew BJ, Chung R, Guillemin GJ:
Characterisation of the expression of NMDA receptors in human
astrocytes. PLoS One 2010, 5:e14123.
185. Myint AM, Kim YK, Verkerk R, Scharpe S, Steinbusch H, Leonard B:
Kynurenine pathway in major depression: evidence of impaired
neuroprotection. J Affect Disord 2007, 98:143�151.
186. Sanacora G, Treccani G, Popoli M: Towards a glutamate hypothesis of
depression: an emerging frontier of neuropsychopharmacology for
mood disorders. Neuropharmacology 2012, 62:63�77.
187. Saleh A, Schroeter M, Jonkmanns C, Hartung HP, Modder U, Jander S: In
vivo MRI of brain inflammation in human ischaemic stroke. Brain 2004,
127:1670�1677.
188. Tilleux S, Hermans E: Neuroinflammation and regulation of glial glutamate
uptake in neurological disorders. J Neurosci Res 2007, 85:2059�2070.
189. Helms HC, Madelung R, Waagepetersen HS, Nielsen CU, Brodin B: In vitro
evidence for the brain glutamate efflux hypothesis: brain endothelial
cells cocultured with astrocytes display a polarized brain-to-blood
transport of glutamate. 2012, 60:882�893.
190. Leonard BE: The concept of depression as a dysfunction of the immune
system. Curr Immunol Rev 2010, 6:205�212.
191. Labrie V, Wong AH, Roder JC: Contributions of the D-serine pathway to
schizophrenia. Neuropharmacology 2012, 62:1484�1503.
192. Gras G, Samah B, Hubert A, Leone C, Porcheray F, Rimaniol AC: EAAT
expression by macrophages and microglia: still more questions than
answers. Amino Acids 2012, 42:221�229.
193. Livingstone PD, Dickinson JA, Srinivasan J, Kew JN, Wonnacott S:
Glutamate-dopamine crosstalk in the rat prefrontal cortex is modulated by Alpha7 nicotinic receptors and potentiated by PNU-120596. J Mol
Neurosci 2010, 40:172�176.194. Kondziella D, Brenner E, Eyjolfsson EM, Sonnewald U: How do glialneuronal
interactions fit into current neurotransmitter hypotheses of
schizophrenia? Neurochem Int 2007, 50:291�301.
195. Wu HQ, Pereira EF, Bruno JP, Pellicciari R, Albuquerque EX, Schwarcz R: The
astrocyte-derived alpha7 nicotinic receptor antagonist kynurenic acid
controls extracellular glutamate levels in the prefrontal cortex. J Mol
Neurosci 2010, 40:204�210.
196. Steiner J, Bogerts B, Schroeter ML, Bernstein HG: S100B protein in
neurodegenerative disorders. Clin Chem Lab Med 2011, 49:409�424.
197. Steiner J, Marquardt N, Pauls I, Schiltz K, Rahmoune H, Bahn S, Bogerts B,
Schmidt RE, Jacobs R: Human CD8(+) T cells and NK cells express and
secrete S100B upon stimulation. Brain Behav Immun 2011, 25:1233�1241.
198. Shanmugam N, Kim YS, Lanting L, Natarajan R: Regulation of
cyclooxygenase-2 expression in monocytes by ligation of the receptor
for advanced glycation end products. J Biol Chem 2003, 278:34834�34844.
199. Rothermundt M, Ohrmann P, Abel S, Siegmund A, Pedersen A, Ponath G,
Suslow T, Peters M, Kaestner F, Heindel W, Arolt V, Pfleiderer B: Glial cell
activation in a subgroup of patients with schizophrenia indicated by
increased S100B serum concentrations and elevated myo-inositol.
Prog Neuropsychopharmacol Biol Psychiatry 2007, 31:361�364.
200. Falcone T, Fazio V, Lee C, Simon B, Franco K, Marchi N, Janigro D: Serum
S100B: a potential biomarker for suicidality in adolescents? PLoS One
2010, 5:e11089.
201. Schroeter ML, Abdul-Khaliq H, Krebs M, Diefenbacher A, Blasig IE: Serum
markers support disease-specific glial pathology in major depression.
J Affect Disord 2008, 111:271�280.
202. Rothermundt M, Ahn JN, Jorgens S: S100B in schizophrenia: an update.
Gen Physiol Biophys 2009, 28 Spec No Focus:F76�F81.
203. Schroeter ML, Abdul-Khaliq H, Krebs M, Diefenbacher A, Blasig IE: Neuronspecific
enolase is unaltered whereas S100B is elevated in serum of
patients with schizophrenia�original research and meta-analysis.
Psychiatry Res 2009, 167:66�72.
204. Rothermundt M, Missler U, Arolt V, Peters M, Leadbeater J, Wiesmann M,
Rudolf S, Wandinger KP, Kirchner H: Increased S100B blood levels in
unmedicated and treated schizophrenic patients are correlated with
negative symptomatology. Mol Psychiatry 2001, 6:445�449.
205. Suchankova P, Klang J, Cavanna C, Holm G, Nilsson S, Jonsson EG, Ekman A:
Is the Gly82Ser polymorphism in the RAGE gene relevant to
schizophrenia and the personality trait psychoticism? J Psychiatry Neurosci
2012, 37:122�128.
206. Scapagnini G, Davinelli S, Drago F, De Lorenzo A, Oriani G: Antioxidants as
antidepressants: fact or fiction? CNS Drugs 2012, 26:477�490.
207. Ng F, Berk M, Dean O, Bush AI: Oxidative stress in psychiatric disorders:
evidence base and therapeutic implications. Int J Neuropsychopharmacol
2008, 11:851�876.
208. Salim S, Chugh G, Asghar M: Inflammation in anxiety. Adv Protein Chem
Struct Biol 2012, 88:1�25.
209. Anderson G, Berk M, Dodd S, Bechter K, Altamura AC, Dell’osso B, Kanba S,
Monji A, Fatemi SH, Buckley P, Debnath M, Das UN, Meyer U, M�ller N,
Kanchanatawan B, Maes M: Immuno-inflammatory, oxidative and nitrosative
stress, and neuroprogressive pathways in the etiology, course and treatment
of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2013, 42:1�42.
210. Coughlin JM, Ishizuka K, Kano SI, Edwards JA, Seifuddin FT, Shimano MA,
Daley EL, et al: Marked reduction of soluble superoxide dismutase-1
(SOD1) in cerebrospinal fluid of patients with recent-onset
schizophrenia. Mol Psychiatry 2012, 18:10�11.
211. Bombaci M, Grifantini R, Mora M, Reguzzi V, Petracca R, Meoni E, Balloni S,
Zingaretti C, Falugi F, Manetti AG, Margarit I, Musser JM, Cardona F, Orefici
G, Grandi G, Bensi G: Protein array profiling of tic patient sera reveals a
broad range and enhanced immune response against Group A
Streptococcus antigens. PLoS One 2009, 4:e6332.
212. Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A, Palomba L,
Cantoni O, Clementi E, Moncada S, Carruba MO, Nisoli E: TNF-alpha
downregulates eNOS expression and mitochondrial biogenesis in fat
and muscle of obese rodents. J Clin Invest 2006, 116:2791�2798.
213. Ott M, Gogvadze V, Orrenius S, Zhivotovsky B: Mitochondria, oxidative
stress and cell death. Apoptosis 2007, 12:913�922.
214. Shalev H, Serlin Y, Friedman A: Breaching the blood�brain barrier as a gate
to psychiatric disorder. Cardiovasc Psychiatry Neurol 2009, 2009:278531.
215. Abbott NJ, Ronnback L, Hansson E: Astrocyte-endothelial interactions at the
blood�brain barrier. Nat Rev Neurosci 2006, 7:41�53.
216. Bechter K, Reiber H, Herzog S, Fuchs D, Tumani H, Maxeiner HG:
Cerebrospinal fluid analysis in affective and schizophrenic spectrum
disorders: identification of subgroups with immune responses and
blood-CSF barrier dysfunction. J Psychiatr Res 2010, 44:321�330.
217. Harris LW, Wayland M, Lan M, Ryan M, Giger T, Lockstone H, Wuethrich I,
Mimmack M, Wang L, Kotter M, Craddock R, Bahn S: The cerebral
microvasculature in schizophrenia: a laser capture microdissection study.
PLoS One 2008, 3:e3964.
218. Lin JJ, Mula M, Hermann BP: Uncovering the neurobehavioural
comorbidities of epilepsy over the lifespan. Lancet 2012, 380:1180�1192.
219. Isingrini E, Belzung C, Freslon JL, Machet MC, Camus V: Fluoxetine effect on
aortic nitric oxide-dependent vasorelaxation in the unpredictable
chronic mild stress model of depression in mice. Psychosom Med 2012,
74:63�72.
220. Zhang XY, Zhou DF, Cao LY, Zhang PY, Wu GY, Shen YC: The effect of
risperidone treatment on superoxide dismutase in schizophrenia. J Clin
Psychopharmacol 2003, 23:128�131.
221. Lavoie KL, Pelletier R, Arsenault A, Dupuis J, Bacon SL: Association between
clinical depression and endothelial function measured by forearm
hyperemic reactivity. Psychosom Med 2010, 72:20�26.
222. Chrapko W, Jurasz P, Radomski MW, Archer SL, Newman SC, Baker G, Lara N,
Le Melledo JM: Alteration of decreased plasma NO metabolites and
platelet NO synthase activity by paroxetine in depressed patients.
Neuropsychopharmacology 2006, 31:1286�1293.
223. Chrapko WE, Jurasz P, Radomski MW, Lara N, Archer SL, Le Melledo JM:
Decreased platelet nitric oxide synthase activity and plasma nitric oxide
metabolites in major depressive disorder. Biol Psychiatry 2004, 56:129�134.
224. Stuehr DJ, Santolini J, Wang ZQ, Wei CC, Adak S: Update on mechanism
and catalytic regulation in the NO synthases. J Biol Chem 2004,
279:36167�36170.
225. Chen W, Druhan LJ, Chen CA, Hemann C, Chen YR, Berka V, Tsai AL, Zweier
JL: Peroxynitrite induces destruction of the tetrahydrobiopterin and
heme in endothelial nitric oxide synthase: transition from reversible to
irreversible enzyme inhibition. Biochemistry 2010, 49:3129�3137.
226. Chen CA, Wang TY, Varadharaj S, Reyes LA, Hemann C, Talukder MA, Chen
YR, Druhan LJ, Zweier JL: S-glutathionylation uncouples eNOS and
regulates its cellular and vascular function. Nature 2010, 468:1115�1118.
227. Szabo C, Ischiropoulos H, Radi R: Peroxynitrite: biochemistry,
pathophysiology and development of therapeutics. Nat Rev Drug Discov
2007, 6:662�680.
228. Papakostas GI, Shelton RC, Zajecka JM, Etemad B, Rickels K, Clain A, Baer L,
Dalton ED, Sacco GR, Schoenfeld D, Pencina M, Meisner A, Bottiglieri T,
Nelson E, Mischoulon D, Alpert JE, Barbee JG, Zisook S, Fava M: Lmethylfolate
as adjunctive therapy for SSRI-resistant major depression:
results of two randomized, double-blind, parallel-sequential trials. Am J
Psychiatry 2012, 169:1267�1274.
229. Antoniades C, Shirodaria C, Warrick N, Cai S, de Bono J, Lee J, Leeson P,
Neubauer S, Ratnatunga C, Pillai R, Refsum H, Channon KM: 5-
methyltetrahydrofolate rapidly improves endothelial function and
decreases superoxide production in human vessels: effects on vascular
tetrahydrobiopterin availability and endothelial nitric oxide synthase
coupling. Circulation 2006, 114:1193�1201.
230. Masano T, Kawashima S, Toh R, Satomi-Kobayashi S, Shinohara M, Takaya T,
Sasaki N, Takeda M, Tawa H, Yamashita T, Yokoyama M, Hirata K: Beneficial
effects of exogenous tetrahydrobiopterin on left ventricular remodeling
after myocardial infarction in rats: the possible role of oxidative stress
caused by uncoupled endothelial nitric oxide synthase. Circ J 2008,
72:1512�1519.
231. Alp NJ, Channon KM: Regulation of endothelial nitric oxide synthase by
tetrahydrobiopterin in vascular disease. Arterioscler Thromb Vasc Biol 2004,
24:413�420.
232. Szymanski S, Ashtari M, Zito J, Degreef G, Bogerts B, Lieberman J:
Gadolinium-DTPA enhanced gradient echo magnetic resonance scans in
first episode of psychosis and chronic schizophrenic patients.
Psychiatry Res 1991, 40:203�207.
233. Butler T, Weisholtz D, Isenberg N, Harding E, Epstein J, Stern E, Silbersweig
D: Neuroimaging of frontal-limbic dysfunction in schizophrenia and
epilepsy-related psychosis: toward a convergent neurobiology.
Epilepsy Behav 2012, 23:113�122.234. Butler T, Maoz A, Vallabhajosula S, Moeller J, Ichise M, Paresh K, Pervez F,
Friedman D, Goldsmith S, Najjar S, Osborne J, Solnes L, Wang X, French J,
Thesen T, Devinsky O, Kuzniecky R, Stern E, Silbersweig D: Imaging
inflammation in a patient with epilepsy associated with antibodies to
glutamic acid decarboxylase [abstract]. In Am Epilepsy Society Abstracts,
Volume 2. Baltimore: American Epilepsy Society; 2011:191.
235. van Berckel BN, Bossong MG, Boellaard R, Kloet R, Schuitemaker A, Caspers
E, Luurtsema G, Windhorst AD, Cahn W, Lammertsma AA, Kahn RS:
Microglia activation in recent-onset schizophrenia: a quantitative (R)-
[11C]PK11195 positron emission tomography study. Biol Psychiatry 2008,
64:820�822.
236. Doorduin J, de Vries EF, Willemsen AT, de Groot JC, Dierckx RA, Klein HC:
Neuroinflammation in schizophrenia-related psychosis: a PET study.
J Nucl Med 2009, 50:1801�1807.
237. Takano A, Arakawa R, Ito H, Tateno A, Takahashi H, Matsumoto R, Okubo Y,
Suhara T: Peripheral benzodiazepine receptors in patients with chronic
schizophrenia: a PET study with [11C]DAA1106. Int J
Neuropsychopharmacol 2010, 13:943�950.
238. M�ller N, Schwarz MJ, Dehning S, Douhe A, Cerovecki A, Goldstein-Muller B,
Spellmann I, Hetzel G, Maino K, Kleindienst N, M�ller HJ, Arolt V, Riedel M:
The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in
major depression: results of a double-blind, randomized, placebo
controlled, add-on pilot study to reboxetine. Mol Psychiatry 2006,
11:680�684.
239. Akhondzadeh S, Jafari S, Raisi F, Nasehi AA, Ghoreishi A, Salehi B, MohebbiRasa
S, Raznahan M, Kamalipour A: Clinical trial of adjunctive celecoxib
treatment in patients with major depression: a double blind and
placebo controlled trial. Depress Anxiety 2009, 26:607�611.
240. Mendlewicz J, Kriwin P, Oswald P, Souery D, Alboni S, Brunello N:
Shortened onset of action of antidepressants in major depression using
acetylsalicylic acid augmentation: a pilot open-label study. Int Clin
Psychopharmacol 2006, 21:227�231.
241. Uher R, Carver S, Power RA, Mors O, Maier W, Rietschel M, Hauser J,
Dernovsek MZ, Henigsberg N, Souery D, Placentino A, Farmer A, McGuffin P:
Non-steroidal anti-inflammatory drugs and efficacy of antidepressants in
major depressive disorder. Psychol Med 2012, 42:2027�2035.
242. M�ller N, Riedel M, Scheppach C, Brandstatter B, Sokullu S, Krampe K,
Ulmschneider M, Engel RR, Moller HJ, Schwarz MJ: Beneficial antipsychotic
effects of celecoxib add-on therapy compared to risperidone alone in
schizophrenia. Am J Psychiatry 2002, 159:1029�1034.
243. M�ller N, Riedel M, Schwarz MJ, Engel RR: Clinical effects of COX-2
inhibitors on cognition in schizophrenia. Eur Arch Psychiatry Clin Neurosci
2005, 255:149�151.
244. M�ller N, Krause D, Dehning S, Musil R, Schennach-Wolff R, Obermeier M,
Moller HJ, Klauss V, Schwarz MJ, Riedel M: Celecoxib treatment in an early
stage of schizophrenia: results of a randomized, double-blind, placebocontrolled
trial of celecoxib augmentation of amisulpride treatment.
Schizophr Res 2010, 121:118�124.
245. Sayyah M, Boostani H, Pakseresht S, Malayeri A: A preliminary randomized
double-blind clinical trial on the efficacy of celecoxib as an adjunct in
the treatment of obsessive-compulsive disorder. Psychiatry Res 2011,
189:403�406.
246. Sublette ME, Ellis SP, Geant AL, Mann JJ: Meta-analysis of the effects of
eicosapentaenoic acid (EPA) in clinical trials in depression. J Clin
Psychiatry 2011, 72:1577�1584.
247. Bloch MH, Hannestad J: Omega-3 fatty acids for the treatment of
depression: systematic review and meta-analysis. Mol Psychiatry 2012,
17:1272�1282.
248. Keller WR, Kum LM, Wehring HJ, Koola MM, Buchanan RW, Kelly DL: A
review of anti-inflammatory agents for symptoms of schizophrenia.
J Psychopharmacol.
249. Warner-Schmidt JL, Vanover KE, Chen EY, Marshall JJ, Greengard P:
Antidepressant effects of selective serotonin reuptake inhibitors (SSRIs)
are attenuated by antiinflammatory drugs in mice and humans. Proc Natl
Acad Sci USA 2011, 108:9262�9267.
250. Gallagher PJ, Castro V, Fava M, Weilburg JB, Murphy SN, Gainer VS, Churchill
SE, Kohane IS, Iosifescu DV, Smoller JW, Perlis RH: Antidepressant response
in patients with major depression exposed to NSAIDs: a
pharmacovigilance study. Am J Psychiatry 2012, 169:1065�1072.
251. Shelton RC: Does concomitant use of NSAIDs reduce the effectiveness of
antidepressants? Am J Psychiatry 2012, 169:1012�1015.
252. Martinez-Gras I, Perez-Nievas BG, Garcia-Bueno B, Madrigal JL, AndresEsteban
E, Rodriguez-Jimenez R, Hoenicka J, Palomo T, Rubio G, Leza JC:
The anti-inflammatory prostaglandin 15d-PGJ2 and its nuclear receptor
PPARgamma are decreased in schizophrenia. Schizophr Res 2011,
128:15�22.
253. Garcia-Bueno B, Perez-Nievas BG, Leza JC: Is there a role for the nuclear
receptor PPARgamma in neuropsychiatric diseases? Int J
Neuropsychopharmacol 2010, 13:1411�1429.
254. Meyer U: Anti-inflammatory signaling in schizophrenia. Brain Behav
Immun 2011, 25:1507�1518.
255. Ramer R, Heinemann K, Merkord J, Rohde H, Salamon A, Linnebacher M,
Hinz B: COX-2 and PPAR-gamma confer cannabidiol-induced apoptosis
of human lung cancer cells. Mol Cancer Ther 2013, 12:69�82.
256. Henry CJ, Huang Y, Wynne A, Hanke M, Himler J, Bailey MT, Sheridan JF,
Godbout JP: Minocycline attenuates lipopolysaccharide (LPS)-induced
neuroinflammation, sickness behavior, and anhedonia.
J Neuroinflammation 2008, 5:15.
257. Sarris J, Mischoulon D, Schweitzer I: Omega-3 for bipolar disorder: metaanalyses
of use in mania and bipolar depression. J Clin Psychiatry 2012,
73:81�86.
258. Amminger GP, Schafer MR, Papageorgiou K, Klier CM, Cotton SM, Harrigan
SM, Mackinnon A, McGorry PD, Berger GE: Long-chain omega-3 fatty acids
for indicated prevention of psychotic disorders: a randomized, placebocontrolled
trial. Arch Gen Psychiatry 2010, 67:146�154.
259. Fusar-Poli P, Berger G: Eicosapentaenoic acid interventions in
schizophrenia: meta-analysis of randomized, placebo-controlled studies.
J Clin Psychopharmacol 2012, 32:179�185.
260. Zorumski CF, Paul SM, Izumi Y, Covey DF, Mennerick S: Neurosteroids,
stress and depression: Potential therapeutic opportunities.
Neurosci Biobehav Rev 2013, 37:109�122.
261. Uhde TW, Singareddy R: Biological Research in Anxiety Disorders. In
Psychiatry as a Neuroscience. Edited by Juan Jose L-I, Wolfgang G, Mario M,
Norman S. Chichester: John Wiley & Sons, Ltd; 2002:237�286.
262. Gibson SA, Korado Z, Shelton RC: Oxidative stress and glutathione
response in tissue cultures from persons with major depression.
J Psychiatr Res 2012, 46:1326�1332.
263. Nery FG, Monkul ES, Hatch JP, Fonseca M, Zunta-Soares GB, Frey BN,
Bowden CL, Soares JC: Celecoxib as an adjunct in the treatment of
depressive or mixed episodes of bipolar disorder: a double-blind,
randomized, placebo-controlled study. 2008, 23:87�94.
264. Levine J, Cholestoy A, Zimmerman J: Possible antidepressant effect of
minocycline. 1996, 153:582.
265. Levkovitz Y, Mendlovich S, Riwkes S, Braw Y, Levkovitch-Verbin H, Gal G,
Fennig S, Treves I, Kron S: A double-blind, randomized study of
minocycline for the treatment of negative and cognitive symptoms in
early-phase schizohprenia. J Clin Psychiatry 2010, 71:138�149.
266. Miyaoka T, Yasukawa R, Yasuda H, Hayashida M, Inagaki T, Horiguchi J:
Possible antipsychotic effect of minocycline in patients with
schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2007, 31:304�307.
267. Miyaoka J, Yasukawa R, Yasuda H, Hayashida M, Inagaki T, Horiguchi J:
Minocycline as adjunctive therapy for schizophrenia: an open-label
study. 2008, 31:287�292.
268. Rodriguez CI, Bender J Jr, Marcus SM, Snape M, Rynn M, Simpson HB:
Minocycline augmentation of pharmacotherapy in obsessive-compulsive
disorder: an open-label trial. 2010, 71:1247�1249.
doi:10.1186/1742-2094-10-43

Cite this article as: Najjar et al.: Neuroinflammation and psychiatric
illness. Journal of Neuroinflammation 2013 10:43.

Close Accordion
Common Leading Causes Behind Hypertension | Wellness Clinic

Common Leading Causes Behind Hypertension | Wellness Clinic

Cardiovascular disease,�also called heart disease, involves numerous issues,�including diseased vessels, structural problems, and blood clots. High blood pressure, or hypertension, occurs when your blood pressure increases to unhealthy levels. Your blood pressure measurement takes into account how quickly blood is passing through your veins and the amount of resistance the blood meets while it’s pumping.

 

What are the leading causes of cardiovascular disease and hypertension?

 

Oxidative stress, inflammation and autoimmune dysfunction initiate and propagate hypertension and cardiovascular disease.�In a series of research studies correlated to cardiovascular disease and hypertension, Dr. Mark C. Houston, MD, discussed the role that oxidative stress, inflammation and autoimmune dysfunction plays in relation to treatment of hypertension and cardiovascular disease.

Oxidative Stress

 

Oxidative stress, with an imbalance between the defense mechanisms and RNS as well as ROS, contributes to the etiology of hypertension in humans and animals. Radical oxygen species and RNS are generated by numerous mobile sources, such as nicotinamide adenine dinucleotide phosphate hydrase (NADPH) oxidase, mitochondria, xanthine oxidase, uncoupled endothelium-derived nitric oxide (NO) synthase (U-eNOS), cyclo-oxygenase and lipo-oxygenase. Superoxide anion is your predominant ROS species produced with these tissues, which inhibits NO and also leads to downstream production of additional ROS (As seen in Figure 3).

 

Vascular Endothelium

 

Patients have impaired an oxidative stress reaction to several stimuli, an elevated plasma oxidative stress and exogenous and endogenous antioxidant defense mechanisms. Hypertensive subjects also have lower plasma ferric reducing ability of plasma vitamin C levels and increased plasma 8-isoprostanes, which correlate with both diastolic and systolic BP. Different single-nucleotide polymorphisms (SNP’s) in genes that codify for antioxidant enzymes are directly linked to hypertension. These include NADPH oxidase, xanthine oxidase, superoxide dismutase 3 (SOD 3), catalase, glutathione peroxidase 1 (GPx 1) and thioredoxin. Antioxidant deficiency and excess free radical production have been implicated in human hypertension in several epidemiologic, observational and interventional studies (Table 2).

 

Table 2 Infographic

 

Radical oxygen species directly damage endothelial cells, degrade NO, influence eicosanoid metabolism, oxidize LDL, lipids, proteins, proteins, DNA and natural molecules, boost catecholamines, harm the genetic machinery, affect gene expression and transcription factors. The interrelations of systems, oxidative stress and cardiovascular disease are shown in Figures 6 and 7. The increased oxidative stress, inflammation and autoimmune vascular dysfunction in human hypertension results in a combination of an response to ROS and RNS increased generation of ROS and RNS and a decreased antioxidant reserve. Increased oxidative stress from the rostral ventrolateral medulla (RVLM) enhances glutamatergic excitatory inputs and attenuates GABA-ergic inhibitory inputs into the RVLM which contributes to increased sympathetic nervous system (SNS) activity from the paraventricular nucleus. Activation of this AT1R in the RVLM raises NADPH oxidase and increases oxidative stress and superoxide anion, increases SNS outflow causing an imbalance of SNS/PNS action with elevation of BP, increased heartbeat and alterations in heart rate variability and heart rate recovery time, which can be obstructed by AT1R blockers.

 

Endothelium-Dependent Responses

 

Neurohormonal and Oxidative Stress System Interaction

 

Inflammation

 

The link between hypertension and inflammation was suggested in both longitudinal and cross-sectional studies. Raised in high sensitivity C-reactive protein (HS-CRP) and other inflammatory cytokines such as interleukin-1B, (IL-1B), IL-6, tumor necrosis alpha (TNF-?) and chronic leukocytosis occur in hypertension and hypertensive-related TOD, such as increased carotid IMT. HS-CRP predicts CV events. Elevated HS-CRP is risk factor and a risk marker for hypertension and CVD. Increases in HS-CRP of over 3 ?g/mL can increase BP in only a couple of days that is directly proportional to the increase in HS-CRP. ENOS and nitric oxide are inhibited by HS-CRP. HS-CRP, down-regulates the AT2R, that counterbalances AT1R. Angiotensin II (A-II) upregulates many of those cytokines, notably IL-6, CAMs and chemokines by activating nuclear factor Kappa B (NF-?B) resulting in vasoconstriction. These events, as well as the increases in endothelin-1 and oxidative stress, elevate BP.

 

Autoimmune Dysfunction

 

Innate and adaptive immune responses are associated with hypertension and hypertension-induced CVD through at least three mechanisms: central nervous system stimulation cytokine generation and renal impairment. This includes salt-sensitive hypertension with dysregulation of both CD4+ and CD8+ lymphocytes increased inflammation because of T cell imbalance and chronic leukocytosis with increased neutrophils and lymphocytes that are decreased. Leukocytosis, especially neutrophils and decreased lymphocyte count raise BP in Blacks by. Macrophages and invade the wall, trigger TLRs, various subtypes govern BP and cause autoimmune vascular damage. Angiotensin II activates immune cells (T cells, macrophages and dendritic cells) and promotes cell infiltration into target organs. CD4+ T lymphocytes express AT1R and PPAR gamma receptors, and release TNF-?, interferon and interleukins inside the vascular wall when triggered (Figure 5). May play a role in the genesis of hypertension brought on by Angiotensin II. Patients have higher TLR 4 mRNA in monocytes in comparison to normal. Intensive decrease in BP to systolic BP (SBP) less than 130 mmHg vs SBP to just 140 mmHg reduces the TLR 4 longer. A-II activates the TLR expression resulting in inflammation and activation of the innate immune system. When TLR 4 is triggered there is downstream macrophage activation, increase metalloproteinase 9, migration, vascular remodeling, collagen accumulation in LVH the gut and cardiac fibrosis. The autonomic nervous system is essential in either increasing or decreasing inflammation and immune dysfunction. Efferent cholinergic pathways through the nerve innervate the spleen, nicotine acetylcholine receptor subunits and cytokine producing immune cells to BP and affect vasoconstriction. Nearby CNS inflammation or ischemia may mediate vascular hypertension and inflammation.

 

Aldosterone is correlated with increased adaptive immunity and autoimmune responses with CD4+ T cell activation and Th 17 polarization with improved IL 17, TGF-? and TNF-? which modulate over 30 inflammatory genes. Serum aldosterone is also an independent risk factor for CVD and CHD via non-hemodynamic effects as well as through increased BP. Blockade of receptors in brain, the heart, blood vessels and immune cells reduces CV danger even with the persistence of hypertension.

 

In conclusion, cardiovascular disease and hypertension have been associated with numerous issues, however, oxidative stress, inflammation and autoimmune dysfunction have been regarded as the most prevalent causes behind cardiovascular disease and hypertension.�Oxidative stress, defined as a disturbance in the balance between the production of reactive oxygen species (free radicals) and antioxidant defenses, inflammation and autoimmune dysfunction, occurs�when the body’s immune system attacks and destroys healthy body tissue by mistake. Cardiovascular disease and hypertension is an indication of cardiovascular issues which should be addressed by a healthcare professional.

 

The scope of our information is limited to chiropractic and spinal injuries and conditions. To discuss options on the subject matter, please feel free to ask Dr. Jimenez or contact us at 915-850-0900 .�Green-Call-Now-Button-24H-150x150-2.png

 

By Dr. Alex Jimenez

 

Additional Topics: Wellness

 

Overall health and wellness are essential towards maintaining the proper mental and physical balance in the body. From eating a balanced nutrition as well as exercising and participating in physical activities, to sleeping a healthy amount of time on a regular basis, following the best health and wellness tips can ultimately help maintain overall well-being. Eating plenty of fruits and vegetables can go a long way towards helping people become healthy.

 

blog picture of cartoon paperboy big news

 

TRENDING TOPIC: EXTRA EXTRA: About Chiropractic

 

 

Epidemiology & Pathophysiology of Cardiovascular Disease | Wellness Clinic

Epidemiology & Pathophysiology of Cardiovascular Disease | Wellness Clinic

Vascular biology, endothelial and vascular smooth muscle as well as cardiac dysfunction play a primary role in hypertension, cardio-vascular disease and target organ damage. Nutrient-gene interactions and epigenetics are predominant factors in promoting positive and negative effects in cardiovascular health and hypertension. In a series of research studies correlated to cardiovascular disease and hypertension, Dr. Mark C. Houston, MD, discussed the role that vascular biology and nutraceuticals play in relation to treatment of hypertension and cardiovascular disease.

 

What is the cause of cardiovascular disease and hypertension?

 

Vascular disease can appropriately be described as the balance between vascular injury and vascular repair (Figure 1). The endothelium is strategically found to be closely associated to the bloodstream as well as the vascular smooth muscle and it is also in charge of discharging a variety of substances in order to preserve vascular homeostasis and health (Figures 2 and 3). Numerous irritations which can provoke damage or harm to the endothelium, can lead to endothelial dysfunction, or ED, and can cause hypertension and other cardiovascular diseases.

 

Vascular Disease Balance

 

Hypertension might be a hemodynamic indication of a damaged or injuried endothelium and vascular smooth muscle which could be connected to definite inflammation responses, oxidative stress and immune dysfunction of the arteries leading to ED, vascular and cardiac smooth muscle disorder, loss of arterial elasticity together with reduced arterial compliance and increased systemic vascular resistance. Hypertension is a significant outcome of the interaction between environmental factors and genetics. Macronutrients and micronutrients are crucial in the regulation of blood pressure, or BP, following target organ damage, or TOD. Nutrient-gene interactions, subsequent gene expression, epigenetics, oxidative stress, inflammation and autoimmune vascular dysfunction have positive or negative influences on vascular biology in humans. Endothelial activation with endothelial dysfunction and vascular smooth muscle breakdown, or VSMD, can ultimately trigger and continue to stimulate the development and growth of hypertension.

 

Blood Vessel Structure

 

Vascular Endothelium

 

Macronutrient and micronutrient deficiencies are extremely common in the general population and may be even more common in individuals with hypertension and cardiovascular disease associated with genetics, environmental factors and even prescription drug and medication usage. These deficiencies have an enormous impact on cardiovascular health issues, such as hypertension, myocardial infarction, or MI, stroke and renal disease. The diagnosis and treatment of those deficiencies will reduce BP and improve ED, vascular biology and health, as well as cardiovascular function.

 

Epidemiology

 

Epidemiology,�the branch of medicine that deals with the incidence, distribution, and possible control of diseases and other factors relating to health, emphasizes the role of diet and related nutritional intake when it comes to hypertension and cardiovascular disease. The transition from the Paleolithic diet to our modern diet has produced an outbreak of nutritionally-related diseases (Table 1). Hypertension, atherosclerosis, coronary heart disease, or CHD, MI, congestive heart failure, or CHF, cerebro-vascular accidents, or CVA, renal disease, type 2 diabetes mellitus, or T2DM, metabolic syndrome, or MS, and obesity are several examples of those diseases. Table 1 contrasts intake of nutrients included during the Paleolithic Era and modern time, involved in the regulation of blood pressure, or BP. An unnatural and unhealthy nutritional selection process has been established by evolution from a pre-agricultural, hunter-gatherer milieu into an agricultural, refrigeration society. In sum, diet has changed more than our genetics can adapt.

 

Dietary Intake of Nutrients Table

 

The human genetic makeup is approximately 99.9 percent that of our Paleolithic ancestors, however our nutritional, vitamin and mineral intakes have vastly changed. The macronutrient and micronutrient variations, oxidative stress from radical oxygen species, or ROS, and radical nitrogen species, or RNS, and inflammatory mediators, such as cell adhesion molecules, or CAMs, cytokines, signaling molecules and autoimmune vascular dysfunction of T cells and B cells, have contributed to the greatest prevalence of hypertension and other cardiovascular diseases through complex nutrient-gene interactions, epigenetic and nutrient-caveolae interactions and nutrient responses with pattern recognition receptors from the endothelium (Figure 4). A decrease in endothelin coupled with endothelial activation, increase in angiotensin II and nitric oxide bioavailability can cause coronary artery disease and vascular disease as well as hypertension. Poor nutrition, together with obesity and a sedentary lifestyle have led to an exponential increase in nutritionally-related ailments. In particular, the high Na+/K+ ratio of contemporary diets has contributed to hypertension, CVA, CHD, MI, CHF and renal disorder as have the relatively low intake of omega-3 PUFA, increase in omega-6 PUFA, saturated fat and trans fatty acids.

 

Infinite Insults

 

Pathophysiology

 

Vascular biology plays a major role in the initiation and perpetuation of hypertension. Oxidative stress (both ROS and RNS), inflammation and autoimmune vascular dysfunction (both T cells and B cells) are the primary pathophysiologic and functional mechanisms that cause cardiovascular disease (Figure 5). All three of them are closely interconnected and provide the absolute combination that leads to cardiovascular disease, or CVD, vascular smooth muscle and cardiac dysfunction, hypertension, vascular disease, atherosclerosis and endothelial dysfunction, or ED.

 

Autoimmune Vascular Dysfunction

 

Hypertension is not a disease but is the proper and continuous, unregulated reaction with an exaggerated outcome of the infinite irritations to the blood vessel following environmental-genetic expression patterns and downstream disturbances in which the vascular system is the innocent bystander. This really becomes a maladaptive vascular response that was initially meant to provide vascular defense to the endothelial insults (Figure 6). Hypertension is a vasculopathy, characterized by ED, structural remodeling, vascular inflammation, improved stiffness, decreased distensibility and loss of elasticity. These insults are biomechanical (BP, heartbeat, blood circulation, oscillatory flow, turbu-lence, enhancement, pulse wave velocity and reflected waves) and biohumoral or biochemical which includes all the non-mechanical causes like metabolic, endocrine, nutritional, toxic, infectious and other etiologies.

 

Endothelium-Dependent Responses

 

In addition to the connections for endocrine and nutritional causes of hypertension, infections and toxins can increase blood pressure as well. Various toxins, such as mercury, polychlorinated biphenyls, lead, cadmium, arsenic and iron, also increase BP and CVD. Numerous microbial organisms have also been implicated in hypertension and CHD. All of these irritations lead to the altered vascular structure and function that manifests clinically as hypertension. Patients with hypertension have abnormal microvasculature in the form of inward eutrophic remodeling of the tiny resistance arteries leading to impaired vasodilatory capacity, increased cardiovascular disease, increased media to lumen ratio, decreased maximal organ perfusion and decreased flow reserve, particularly in the heart with decreased coronary flow reserve. Significant functional structural microvascular impairment occurs even before the individual’s blood pressure begins to increase in normotensive offspring of the patients with hypertension, as evidenced by endothelial dysfunction, diminished vasodilation, forearm vascular resistance, diastolic dysfunction, increased left ventricular mass index, increased septal and posterior wall thickness and left untreated hypertrophy. Therefore, the processes underlying the circulatory issues are associated to a vascular phenotype of hypertension that may be determined by early life programming and imprinting which may be compounded by cerebral aging.

 

In conclusion, vascular biology, endothelial and vascular smooth muscle as well as cardiac dysfunction play a primary role in hypertension, cardio-vascular disease and target organ damage. Then, the epidemiology, or the incidence, distribution, and possible control of diseases and other factors relating to health, emphasizes the role of diet and related nutritional intake when it comes to hypertension and cardiovascular disease. And finally, oxidative stress, inflammation and autoimmune vascular dysfunction are the primary pathophysiologic and functional mechanisms that cause cardiovascular disease. Hypertension is an indication of cardiovascular issues which should be addressed by a healthcare professional.

 

The scope of our information is limited to chiropractic and spinal injuries and conditions. To discuss options on the subject matter, please feel free to ask Dr. Jimenez or contact us at 915-850-0900 .

Green-Call-Now-Button-24H-150x150-2.png

By Dr. Alex Jimenez

 

Additional Topics: Wellness

 

Overall health and wellness are essential towards maintaining the proper mental and physical balance in the body. From eating a balanced nutrition as well as exercising and participating in physical activities, to sleeping a healthy amount of time on a regular basis, following the best health and wellness tips can ultimately help maintain overall well-being. Eating plenty of fruits and vegetables can go a long way towards helping people become healthy.

 

blog picture of cartoon paperboy big news

 

TRENDING TOPIC: EXTRA EXTRA: About Chiropractic

 

 

Why Is The Thyroid So Prone To Autoimmune Disease?

Why Is The Thyroid So Prone To Autoimmune Disease?

Thyroid & Autoimmune Disease:

Key Words:

  • Autoimmune thyroid disease
  • Smoking
  • Environmental factors
  • Endogenous factors
  • Accelerator hypothesis
  • Selenium intake
  • Iodine intake

Abstract

The thyroid gland plays a major role in the human body; it produces the hormones necessary for appropriate energy levels and an active life. These hormones have a critical impact on early brain development and somatic growth. At the same time, the thyroid is highly vulnerable to autoimmune thyroid diseases (AITDs). They arise due to the complex inter- play of genetic, environmental, and endogenous factors, and the specific combination is required to initiate thyroid autoimmunity. When the thyroid cell becomes the target of autoimmunity, it interacts with the immune system and appears to affect disease progression. It can produce different growth factors, adhesion molecules, and a large array of cytokines. Preventable environmental factors, including high iodine intake, selenium deficiency, and pollutants such as tobacco smoke, as well as infectious diseases and certain drugs, have been implicated in the development of AITDs in genetically predisposed individuals. The susceptibility of the thyroid to AITDs may come from the complexity of hormonal synthesis, peculiar oligoelement requirements, and specific capabilities of the thyroid cell�s defense system. An improved understanding of this interplay could yield novel�treatment pathways, some of which might be as simple as identifying the need to avoid smoking or to control the in- take of some nutrients.

Introduction

The thyroid gland is important in the human body because of its ability to produce hormones necessary for appropriate energy levels and an active life. These molecules have pleiotropic effects, playing critical roles in early brain development, somatic growth, bone maturation, and the mRNA synthesis of more than 100 proteins that constantly regulate each and every bodily function.

At the same time, the thyroid is highly vulnerable to autoimmune diseases. The incidence of chronic autoimmune thyroiditis (CAT) and Graves� disease (GD) has in- creased dramatically over the past few decades, afflicting up to 5% of the general population. In children, CAT is the most common cause of acquired hypothyroidism in non-endemic goiter areas.

Initial studies on the association between early fetal nutrition and the pathogenesis of autoimmune thyroid diseases (AITDs) resulted in controversial data. In twin studies, Phillips et al. [1] found that among monozygotic twins the smaller twin had higher levels of thyroid per- oxidase (TPO) antibodies. However, these data were not�confirmed in another twin study in which a larger cohort was analyzed [2]. The �accelerator hypothesis� and the influence of rapid childhood growth due to energy-dense food and adipokine imbalance have not been investigated in childhood AITDs. In both type 1 and type 2 diabetes, the accelerator hypothesis proposes a critical influence of obesity as an exogenous factor contributing to disease; even in a population of children with type 1 diabetes, the fattest presented with disease the earliest (evidence of true acceleration) [3]. With regard to AITDs, other accelerators in addition to obesity include low selenium (Se) and a high iodine intake. Obese children are hyperleptinemic, and leptin, with its numerous functions including the promotion of cell-mediated immune responses, is a good candidate for contributing to the pathogenesis of autoimmune diseases. Obese children have been found to have increased interferon (IFN)- -secreting T helper cells and altered thyroid structure and hormonal status [4�8].

Autoimmunity is generally considered to be only a cause of disease; nevertheless, human T cell repertoires naturally comprise autoimmune lymphocytes. Autoimmune T cells can help heal damaged tissues, indicating that natural autoimmunity can also contribute to health and benefit self-maintenance [9]. The immune system makes its decisions and acts by integrating multiple signals in an ongoing dialog with tissues. It is likely that the tissue itself provides signals that trigger the type of inflammation that is required for tissue self-maintenance and repair [9, 10].

thyroid Fig 1Autoimmune disorders result from a complex interplay of genetic, environmental, and endogenous factors (fig. 1), and a combination of these factors is required to initiate thyroid autoimmunity [11, 12]. Recent advances in genome-wide studies have made it possible to efficient- ly identify complex disease-associated genes. Using both the candidate gene approach and whole-genome linkage studies, 6 AITD susceptibility genes have been identified and confirmed; the first group includes the immunomodulatory gene products HLA-DR, CD40, cytotoxic T lymphocyte-associated factor (CTLA-4), and protein tyrosine phosphatase 22 (PTPN22), and the second group includes the thyroid-specific gene products thyroglobulin (Tg) and thyroid-stimulating hormone receptor (TSHR). Genetic factors predominate, accounting for approximately 80% of the likelihood of developing AITDs, whereas at least 20% is due to environmental factors (fig. 1). In recent years, a number of excellent reviews have been published on the genetic background of AITDs [13, 14].

An increased frequency of AITDs is reported in Turner syndrome (TS) and in other nondisjunctional chromosomal disorders such as Down and Klinefelter syndromes. The theory that maternal autoimmunity may lead to the preferential survival of a fetus with chromosomal aneuploidy is attractive but remains unproven [15]. The most prevalent autoimmune disorder in TS appears to be CAT, with a reported thyroid autoantibody incidence of 30� 50%. Hypothyroidism of autoimmune origin is so common in TS that almost every other TS woman will prob- ably develop hypothyroidism, and it increases with age [16, 17].

We know more about the minor details of AITDs, but the main question remains unanswered: why is the thyroid so prone to autoimmune disease? This review seeks to emphasize the role of the thyroid cell per se in AITDs and to focus attention on preventable exogenous factors.

Thyroid Cell Specificity

The thyroid cell produces a variety of immunologically active factors (table 1) and has complex nutrient requirements for hormonal synthesis and function (table 2), both of which influence susceptibility to AITDs. Thus, the thyroid cell is not just the innocent victim of an�unchecked and disordered immune system. It is increasingly obvious that the target cells interact with the immune system, often in ways that seem defensive and protective, yet they can go awry and exacerbate autoimmunity under particular circumstances [11].

thyroid Tabel 1

thyroid Table 2In most human autoimmune diseases, the events that trigger autoimmunity remain elusive. Most importantly, it is unclear whether autoimmunity results primarily from an immune defect, is secondary to target organ alterations, or both. The thyroid shows increased iodine uptake and oxidation prior to lymphocytic infiltration concomitant with decreased thyroid epithelial cell proliferation in vitro. Modifying thyroid function influences the development of thyroid autoimmunity [18]. The thyroid cell, unlike other epithelial cells in the endocrine system, is unique because it releases hormonal products on its basal surface instead of its apical surface, thus allowing for the trafficking of precious iodine twice across the cell.

Thyroid cells are capable of producing different factors (table 1), including IGF I, IGF II, and EGF, that can stimulate angiogenesis. The half-life of these molecules is short and they induce only local (non-systemic) effects. Stimulated thyroid follicular cells secrete several growth factors [19]. The expression of intercellular adhesion molecule-1 (ICAM-1) and lymphocyte function-associated antigen-3 (LFA-3) by thyroid cells is enhanced by IFN- , tumor necrosis factor (TNF), and interleukin (IL)-1. Thyroid cells express CD44, which acts as a homing receptor for hyaluronan, mediates leukocyte rolling (the first step in tissue homing), and may (like ICAM-1) induce lymphocyte activation under certain circumstances. Thyroid cells are now known to produce many cytokines (especially after stimulation with IL-1), including IL-1, IL-6, IL-8, IL-12, IL-13, and IL-15 [11]. Activated lymphocytes can produce TSH, which could have a variety of implications [20].

Low dose tolerance can easily be broken, and the thyroid is not well tolerated by the immune system. Auto- antigens in AITDs, as in other autoimmune endocrine diseases, include tissue-specific membrane receptors, enzymes, and secreted hormones. Mixed cellular and anti- body autoimmune responses are likely pathogenic to some degree. Circulating anti-Tg autoantibodies are also found in GD and CAT, as are autoantibodies to triiodothyronine (T3) and thyroxine (T4). The human (h) TSHR is the primary antigenic target in autoimmune hyperthyroidism [21]. The TPO autoantibody seems unlikely to have much pathogenic importance as it has limited access to TPO in vivo due to its location inside the cell. Further- more, anti-TPO autoantibodies do not inhibit the activity�of the enzyme. Thus, their clinical value is principally to document thyroid gland autoimmunity. However, TPO may act as a hidden antigen because it is not adjacent to the vasculature.

In humans, excess thyroid hormone results in the attenuation of natural killer (NK) cell activity, which in theory could lead to the continuation of an autoimmune disorder. Upon return to a euthyroid status and the resulting normalization of NK activity, a reversion to control of the abnormal immune reaction would occur with perpetuation of GD. Additionally, an anti-idiotype might function as an agonist for the original antigen. Thus, an antibody to an antibody (anti-idiotype) to TSH might bind to the TSHR and stimulate the thyroid [22]. A more likely hypothesis is that anti-idiotypic antibodies are rarely produced at a detectable level. Hodkinson et al. [23] recently found a positive association between thyroid hormone concentration and NK-like T cells in the elderly. This relationship has not been investigated in young patients.

Antigen Presentation By The Thyroid Cell

Bottazzo et al. [24] first suggested that antigen presentation by HLA-DR-expressing thyroid cells may be a critical aspect of thyroid autoimmune disease. It quickly became apparent that the only stimulus able to induce MHC class II expression on thyroid cells was the T cell cytokine IFN- . Normal cells respond exactly the same as AITD thyroid cells to IFN- , and in animal models of AITDs class II expression on thyroid cells is always followed by the appearance of lymphocytes in the gland. In addition to inducing MHC class II expression, IFN- increases MHC class I expression on thyroid cells, thus allowing potential for the recognition of thyroid cells by cytotoxic CD8+ T cells [11].

It is possible that direct antigen presentation by the thyroid cell itself may occur in individuals who inherit thyroid-reactive T cells; such a circumstance would effectively bypass the classical macrophage-processing mechanism. The HLA-DR antigen-expressing thyroid cell may be as effective as the macrophage at presenting thyroid- specific antigens to the immune system [25], but the thyroid cell is incapable of supplying the costimulatory signals that professional antigen-presenting cells (APCs) do [11]. Any stimulus that causes increased DR expression on thyrocytes, such as IFN- produced by T cells in response to infection, combined with increased TSH stimulation may allow thyrocytes to function as APCs. Although thyroid cells may perform this function poorly, they are numerous and localized in one area, therefore allowing for increased production of the already established normally occurring low levels of antibodies [12].

Environmental Factors

A number of environmental factors have been implicated in the development of AITD in genetically predisposed individuals, including high iodine intake, Se deficiency, pollutants such as tobacco smoke, infectious dis- eases, certain drugs, and physical and emotional stress [26�30]. Herein, we focus on these preventable triggers. Individual susceptibility suggests that, in addition to genetics, some endogenous factors are also important to the development of AITDs, such as growth spurts in childhood, puberty, pregnancy, menopause, aging, and gender (fig. 1, 2).

thyroid Fig 2Iodine

Dietary iodine plays an important role in the expression of AITDs. Epidemiological studies have suggested that AITDs are more common in areas of iodine sufficiency than in areas of iodine deficiency and that general increases in AITDs occur in parallel with increases in dietary iodine. CAT is less common in countries with a low iodine intake [27].

The thyroid requires the right amount of iodine. Either too much or too little causes problems. Too little io- dine brings all of the adaptive immune mechanisms of the thyroid into play, but despite these responses iodine deficiency disorders may still result. Too much iodine also affects the thyroid. Protective mechanisms include diminished trapping of iodide by the thyroid and de- creased iodide organification. In experimental thyroiditis several types of Tg epitopes have been found, including some containing iodine and/or hormones as well as some conformational epitopes. Experimentally increasing the iodination of Tg makes the protein more antigen- ic [28, 31]. Optimally, the iodine intake of a population should be kept within a relatively narrow interval that prevents iodine disorders, but not higher [29].

The mechanism of action of iodine in contributing to thyroid autoimmunity is not clear. Iodine may stimulate B lymphocytes to increase the production of immunoglobulin and thus induce AITDs by enhancing the activity of lymphocytes that have been primed by thyroid- specific antigens [30]. Iodine may enhance the antigen- presenting capabilities of macrophages, resulting in increased macrophage activity and enhanced lymphocyte stimulation. In addition, a high iodine intake in- creases the iodine content of the Tg molecule, which may increase its immunogenicity [31]. Lastly, iodine may provoke thyroid follicular cells to become APCs and thus potentiate AITDs by turning genetically predisposed normal thyrocytes into antigen-presenting thyrocytes.

Table 2 shows several minerals and trace elements that are essential for normal thyroid hormone metabolism. The role of these elements in childhood AITDs has not been well investigated.

Selenium

The second factor that has been strongly implicated in the development of autoimmune thyroiditis is the trace element Se. Se is a constituent of selenoproteins (SePs), in which it is incorporated as selenocysteine. Relevant actions of Se and SePs include antioxidant effects, appropriate functioning of the immune system, antiviral effects, influence on fertility, and a beneficial effect on mood [32]. Se deficiency is thought to be involved in the pathogenesis of autoimmune thyroiditis by lengthening the duration and exacerbating the severity of the disease; these effects may occur via reduced activity of the SeP glutathione peroxidase, which leads to an increased production of hydrogen peroxide. Another important class of SePs are the iodothyronine selenodeiodonases D1 and D2, which are responsible for producing biologically active T3 via 5 -deiodination in extrathyroidal tissues [33, 34].

Combined Se and iodine deficiencies lead to myxedematous cretinism. Adequate Se nutrition supports efficient thyroid hormone synthesis and metabolism and protects the thyroid gland from damage from excessive iodine exposure. In regions having severe combined deficiencies of iodine and Se, it is mandatory to normalize the Se supply before the initiation of iodine supplementation to prevent hypothyroidism [35].

In celiac disease, the inability to absorb Se may modulate SeP gene expression and promote intestinal mucosal damage, and this deficiency could additionally predispose to complications such as AITDs [34, 36].

Derumeaux et al. [37] discovered an inverse association between Se status and thyroid volume and echo- structure in French adults and concluded that Se may protect against AITDs. Duntas et al. [38] found beneficial effects when treating patients with autoimmune thyroiditis with selenomethionine for 6 months due to its ability to reduce anti-TPO antibodies. In the group treat- ed with LT4 combined with Se, these effects were very prominent in the first 3 months and were further sustained after 6 months of treatment. A striking majority of the patients reported an improvement in mood and well-being.

Environmental Pollutants

Various environmental toxins and pollutants have been implicated in the induction of AITDs.

Polyhalogenated biphenyls are commonly used com- pounds with a wide variety of industrial applications. Polybrominated biphenyls are flame retardant, and polychlorinated biphenyls (PCBs) are used as lubricants, adhesives, inks, and plasticizers. PCBs are known to accumulate in lakes and rivers and subsequently in the adipose tissue of fish and humans [27]. These compounds might trigger AITDs by interfering with iodide transport and inducing oxidative stress. There is evidence that peri- natal PCB exposure decreases thyroid hormone levels in rat pups. In adults, adolescents, and children from highly PCB-exposed areas, the concentration of PCBs in blood samples negatively correlated with levels of circulating thyroid hormones [39, 40]. Populations with long-term exposure to PCBs have increased prevalences of anti-TPO antibodies, which is probably related to the immunomodulatory effects of PCBs. Pollutants from car emissions and heavy industry as well as coal pollution and agricultural fungicides are also implicated in AITD development [26, 27].

Smoking is associated with an increased risk of developing GD and with a reduced remission rate after thionamide treatment. Even more striking is the effect of smoking on Graves� orbitopathy, which tends to be more severe in smokers [32, 41]. Smoking might contribute to the pathogenesis of GD by altering the structure of the thyrotropin receptor, making it more immunogenic and leading to the production of thyrotropin receptor-stimulating antibodies that react strongly with retroorbital tissue [41]. Smoking induces the polyclonal activation of B and T cells and increases presentation of antigens by damaged cells. Hypoxia may play a role in Graves� orbitopathy because retrobulbar fibroblasts show a significant increase in proliferation and glycosaminoglycan production when cultured under hypoxic conditions [42, 43]. The effects of parental smoking on thyroid function in fetuses or 1-year-old infants [44] provide additional insight into the interrelationship between smoking and thyroid dysfunction. The latter study found that infants whose mothers and fathers smoked had higher cord serum concentrations of Tg and thiocyanate than did infants whose parents did not smoke. The clinical picture observed in adolescents exposed to passive smoking could be due to direct stimulation of sympathetic nervous activity by nicotine in addition to the smoking-induced increase in thyroid hormone secretion [45].

The association of smoking with CAT is less well defined. Although a relationship with autoimmune hypothyroidism or postpartum thyroiditis has been reported, this finding was not supported by meta-analysis of the published papers [32, 45].

Infections

In some individuals, autoimmunity is the price that must be paid for the eradication of an infectious agent. Infections have been implicated in the pathogenesis of several autoimmune, endocrine, and non-endocrine diseases. Either viral or bacterial infections might represent a risk factor for the development of AITDs. Viruses have long been suspected as etiological agents in many auto- immune diseases, including AITDs; moreover, a viral cause of AITDs, infecting either the thyroid or immune cells, has been demonstrated in an avian model. Although viruses may be likely etiological agents in human AITDs, this possibility remains unproven [25, 27, 30].

An increased frequency of antibodies to the influenza B virus has been observed in a group of patients with thyrotoxicosis. In addition, virus-like particles have been found in the thyroids of chickens with autoimmune thyroiditis, with similar particles detected in the thyroids of humans. Serological evidence of prior staphylococcal and streptococcal illnesses has been described in a few patients with AITDs [27].

Some of the strongest evidence linking infectious agents to the induction of AITDs has been the association of Yersinia enterocolitica infection with thyroid disease. This Gram-negative coccobacillus commonly causes diarrhea along with a variety of abnormalities that suggest autoimmune disease, including arthralgias, arthritis, erythema nodosum, carditis, glomerulonephritis, and iritis. Weiss et al. [46] demonstrated that Y. enterocolitica had a saturable, hormone-specific binding site for the mammalian TSH that resembled the receptor for TSH in the human thyroid gland.

An immune response against a viral antigen that shares homology with the TSHR may be the inductive event that ultimately leads to TSHR autoimmunity [21]. A significant association between hepatitis C and AITDs has been found. Anti-TPO antibody titers have been shown to increase at the end of treatment with IFN- in patients with the hepatitis C virus, and these patients were more susceptible to AITDs than were hepatitis B patients. These patients should be screened for autoimmune thyroiditis before and after IFN treatment [47, 48].

Infection might induce an autoimmune response by various mechanisms, such as molecular mimicry, polyclonal T cell activation by microbial superantigens, and increased thyroid expression of human leukocyte anti- gens [49]. Inflammation induced by viral infections or by pollutants can modify cell signaling pathways and influence T cell activity and cytokine secretion profiles [26].

Drugs

Several drugs have been implicated in the pathogenesis of AITDs. Amiodarone is an iodine-containing drug with diverse effects on thyroid function. Serum titers of TPO antibodies are elevated in approximately half of the patients who develop amiodarone-induced hypothyroid- ism. Amiodarone has also been shown to affect T cell function [27]. Thyroid antibodies disappeared from the circulation 6 months after amiodarone discontinuation [32].

Lithium, a psychopharmaceutical and well-known goitrogen, has been shown to inhibit thyroid hormone release. Antithyroid antibodies are found more frequently in psychiatric patients on lithium therapy than in similar psychiatric patients treated with other drugs. Lithium-induced increases in serum TSH concentrations might enhance autoantigen expression on the surface of thyrocytes, thereby exacerbating autoimmune responses [32, 50].

Other agents involved in thyroid autoimmunity are IL-2 (thyroid autoimmune phenomena with or without hypothyroidism), IFN- (thyroid dysfunction, hypothyroidism, and occurrence of thyroid autoantibodies), highly active antiretroviral therapy (HAART; possible occurrence of thyroid autoimmune phenomena and dysfunction), and Campath-1H, a humanized monoclonal antibody targeting the CD52 antigen on lymphocytes and monocytes that is used after transplantation (occurrence of GD) [32].

Stress

Although numerous anecdotal reports have associated the onset of AITDs, and particularly GD, with stressful events, objective evidence has been difficult to obtain. Both psychological stress, such as bereavement, and physical stress, such as trauma or major illness, have been implicated [27].

Neuroendocrine immune mechanisms responsible for the putative effects of stress on the onset and course of GD are poorly defined, but they might include activation of the HPA axis (although this should cause immunosuppression) and a shift from a Th1 (cell-mediated) immune response to a Th2 (humoral) immune response [32, 51].

Additionally, heat shock proteins (HSPs), which are well-known stress proteins, could share epitopes with the TSHR. Heufelder et al. [52] found that high levels of HSP- 72 expression in AITDs may reflect a state of chronic cellular stress, but this finding could also indicate an immunomodulatory function of HSP-72 in AITDs. HSPs are ubiquitous, highly conserved proteins that are expressed in response to a wide variety of physiological and environmental insults. They allow cells to survive otherwise lethal conditions. HSPs have been postulated to be critical antigens in both autoimmune diseases and experimental models of autoimmunity [53, 54].

Improving stress by the prolonged use of bromazepam has been shown to increase the remission rate of hyper- thyroidism after a thionamide course [55]. The relation- ship between stress and CAT is less evident. Graves� patients might be stressed because of hyperthyroidism and not hyperthyroid because of stress, whereas CAT patients are not stressed because they are euthyroid or hypothyroid [32]. Whatever the mechanism of action, stress may cause decompensation in a genetically susceptible individual and lead to the induction or exacerbation of an AITD.

Pregnancy And Postpartum

AITDs tend to be more frequent in women. The reason for this gender-related difference is not clear and is not explained by the additional X chromosome in females [42]. The possibility that genes responsible for immune responses are located on the X chromosome has been considered but not confirmed. Sex steroids could modify immune responses by acting directly on immune cells. Estrogens are well-known stimulators of TSH secretion, which could enhance HLA-DR expression. Parity per se does not seem to play a significant role [32, 56].

The accumulation of fetal cells in the maternal thyroid gland during pregnancy (painless postpartum thyroiditis) may induce autoimmune thyroiditis [57]. Pregnancy is accompanied by a suppression of the immune system with a shift in the Th1/Th2 balance towards Th2 immunity, a process that is aimed at protecting the fetus. A possible link between pregnancy and the postpartum occurrence of AITDs might be represented by fetal microchimerism. Fetal cells pass into the maternal circulation and may persist in the maternal blood. Microchimerism of presumed fetal origin has been shown in thyroid tissue specimens of women with previous pregnancies, particularly in those with AITDs. The persistence of activated�intrathyroidal fetal cells might influence thyroid autoimmunity in genetically susceptible women by modulating or even initiating maternal immune responses in a graft- versus-host reaction upon termination of pregnancy-re- lated immune suppression. It cannot presently be ruled out, however, that intrathyroidal fetal cells are only innocent bystanders and do not participate in triggering or exacerbating thyroid autoimmune responses [32, 54, 58]. Mothers who have given birth to sons have thyroidal Y chromosome-positive cells more frequently if they are affected by either CAT or GD than if they have thyroid adenomas [59].

The presence of elevated TPO antibodies in about 10% of pregnant women is associated with an increased risk of miscarriage, gestational thyroid dysfunction, and postpartum thyroiditis [48]. Maternal-to-fetal transfer of TSHR antibodies with polyclonal activity and a different half-life can lead to a transient perinatal thyroid dysfunction, opposite to a maternal one [60].

Conclusion

A rapidly growing body of evidence on the interplay between genetic, environmental, and endogenous factors has expanded our knowledge of the complex etiopathogenesis of AITDs. Autoimmune thyroid disorders are examples of common diseases in which immunogenetic factors play an important role.

The thyroid cell itself appears to play a major role in disease progression by interacting with the immune system. The complexity of hormonal synthesis, unique oligoelement requirements, and the specific capabilities of the thyroid cell defense system probably make the thyroid prone to AITDs. The initial insult to the human thyroid gland that activates the onset of AITDs remains un- known and seems to be strongly individual. Understand- ing more about the interaction between genes and the environment could yield entirely novel pathways, some of which might be as simple as identifying the need to avoid smoking or to control the intake of particular nutrients. Evidence for many causal agents is, however, scarce, and more data are certainly required. We believe that it is particularly important to draw attention to this problem in pediatric patients. Lessons learned from the enigmatic questions raised in AITD studies could clarify the pathogenesis of other organ-specific autoimmune disorders.

L. Saranac S. Zivanovic B. Bjelakovic H. Stamenkovic M. Novak B. Kamenov Pediatric Clinic, University Clinical Center, Nis, Serbia

Blank
References:

1 Phillips DI, Osmond C, Baird J, Huckle A,
Rees-Smith B: Is birthweight associated with
thyroid autoimmunity? A study in twins.
Thyroid 2002;12:377�380.
2 Brix TH, Hansen PS, Rudbeck AB, Hansen
JB, Skythe A, Kyvik KO, Hegedus L: Low
birth weight is not associated with thyroid
autoimmunity: a population-based twin
study. J Clin Endocrinol Metab 2006;91:
3499�3502.
3 Wilkin TJ: The great weight gain experiment,
accelerators and their implications for
autoantibodies in diabetes. Arch Dis Child
2006;91:456�458.
4 Matarese G, La Cava A, Sanna V, Lord MG,
Lechler RI, Fontana S, Zappacosta S: Balancing
susceptibility to infection and autoimmunity:
a role of leptin? Trends Immunol
2002;23:182�187.
5 Radetti G, Kleon W, Buzi F, Crivellero C,
Pappalardo L, Di Lorgi N, Maghnie M: Thyroid
structure and function are affected in
childhood obesity. J Clin Endocrinol Metab
2008;93:4749�4754.
6 Pacifico L, Di Renzo L, Anania C, Osborn JF,
Ippoliti F, Schiavo E, Chiesa C: Increased Thelper
interferon-gamma-secreting cells in
obese children. Eur J Endocrinol 2006;154:
691�697.
7 Marras V, Casini MR, Pilia S, Carta D, Civolani
P, Porcu M, Uccheddu AP, Loche S: Thyroid
function in obese children and adolescents.
Horm Res Paediatr 2010;73:193�197.
8 Saranac L, Zivanovic S, Novak M: High fT3
(free triiodothyronine), new syndrome or innocent
bystander. Endocr Abstracts Eur
Congr Endocrinol, Prague, 2010, p 771.
9 Schwartz M, Cohen IR: Autoimmunity can
benefit self-maintenance. Immunol Today
2000;21:265�268.
10 Cohen IR, Schwartz M: Autoimmune maintenance
and neuroprotection of the central
nervous system. J Neuroimmunol 1999;100:
111�114.
11 Weetman AP: Autoimmune thyroid disease:
propagation and progression. Eur J Endocrinol
2003;148:1�9.
12 Weetman AP: New aspects of thyroid autoimmunity.
Horm Res 1997;48(suppl 4):51�
54.
13 Jacobson EM, Tomer Y: The CD40, CTLA-4,
thyroglobulin, TSH receptor, and PTPN22
gene quintet and its contribution to thyroid
autoimmunity: back to the future. J Autoimmun
2007;28:85�98.
14 Tomer Y, Huber A: The etiology of autoimmune
thyroid disease: a story of genes and
environment. J Autoimmun 2009;32:231�
239.
15 Saenger P: Turner syndrome; in Sperling MA
(ed): Pediatric Endocrinology, ed 3. Philadelphia,
Saunders Elsevier, 2008, pp 610�661.
16 El-Mansoury M, Bryman I, Berntorp K,
Hanson C, Wilhelmsen L, Landin-Wilhelmsen
K: Hypothyroidism is common in Turner
syndrome: results of a five-year follow up.
J Clin Endocrinol Metab 2005;90:2131�2135.
17 Mortensen KH, Cleemann L, Hjerrild BE,
Nexo E, Locht H, Jeppesen EM, Gravholt
CH: Increased prevalence of autoimmunity
in Turner syndrome � influence of age. Clin
Experim Immunol 2009;156:205�210.
18 Homo-Delarche F, Boitard C: Autoimmune
diabetes: the role of the islets of Langerhans.
Immunol Today 1996;17:456�460.
19 Denef JF, Ovaert C, Many MC: Experimental
goitrogenesis (in French). Ann Endocrinol
(Paris) 1989;50:1�15.
20 Fabry Z, Raine CS, Hart MN: Nervous tissue
as an immune compartment: the dialect of
the immune response in the CNS. Immunol
Today 1994;15:218�224.
21 Song YH, Li Y, Maclaren NK: The nature of
autoantigens targeted in autoimmune endocrine
diseases. Immunol Today 1996;17:232�
238.
22 Zakarija M, McKenzie JM: The spectrum
and significance of autoantibodies reacting
with the thyrotropin receptor. Endocrinol
Metab Clin North Am 1987;16:343�364.
23 Hodkinson CF, Simpson EEA, Beattie JH,
O�Conor JM, Campbell DJ, Strain JJ, Wallace
JM: Preliminary evidence of immune function
modulation by thyroid hormones in
healthy men and women aged 55�70 years. J
Endocrinol 2009;202:55�63.
24 Botazzo GF, Pujol-Borrell R, Hanafusa T,
Feldmann M: Role of aberrant HLA-DR expression
and antigen presentation in induction
of endocrine autoimmunity. Lancet
1983;2:1115�1119.
25 Davies TF, Piccini LA: Intrathyroidal MHC
class II antigen expression and thyroid autoimmunity.
Endocrinol Metab Clin North
Am 1987;16:247�268.
26 Duntas LH: Environmental factors and autoimmune
thyroiditis. Nat Clin Pract Endocrinol
Metab 2008;4:454�460.
27 Safran M, Paul TL, Roti E, Braverman LE:
Environmental factors affecting autoimmune
thyroid disease. Endocrinol Metab
Clin North Am 1987;6:327�342.
28 Dunn JT: What is happening with our iodine?
J Clin Endocrinol Metab 1998;3398�
3400.
29 Laurberg P, Cerqueira C, Ovesen L, Rasmusen
LB, Perrild H, Andersen S, Pedersen IB,
Carle A: Iodine intake as a determinant of
thyroid disorders in population. Best Pract
Res Clin Endocrinol Metab 2010;24:13�27.
30 Weetman AP, McGregor AM: Autoimmune
thyroid disease: developments in our understanding.
Endocr Rev 1984;5:309�355.
31 Carayanniotis G, Rao VP: Searching for
pathogenic epitopes in thyroglobulin: parameters
and caveats. Immunol Today 1997;
18:83�88.
32 Bartalena L, Tanda ML, Piantanida E, Lai A,
Compri E, Lombardi V: Environment and
thyroid autoimmunity; in Wiersinga WM,
Drexhage HA, Weetman AP, et al (eds): The
Thyroid and Autoimmunity: Merck European
Thyroid Symposium Noordwijk 2006,
June 15�18. Stuttgart, Thieme, 2007 pp 60�
73.
33 Berry MJ, Bany L, Larsen PR: Type I iodothyronine
deiodinase is a selenocysteine-containing
enzyme. Nature 1991;349:438�440.
34 Duntas LH: Selenium and inflammation:
underlying anti-inflammatory mechanisms.
Horm Metab Res 2009;41:443�447.
35 Zimmerman MB, Kohrle J: The impact of
iron and selenium deficiencies on iodine and
thyroid metabolism: biochemistry and relevance
to public health. Thyroid 2002;12:
867�878.
36 Duntas LH: Does celiac disease trigger autoimmune
thyroiditis. Nat Rev Endocrinol
2009;5:190�191.
37 Derumeaux E, Valeix P, Castetbon K, Bensimon
M, Boutron-Ruault MC, Arnaud JH,
Hercberg S: Association of selenium with
thyroid volume and echostructure in 35- to
60-year-old French adults. Eur J Endocrinol
2003;148:309�315.
38 Duntas LH, Mantzou E, Koutras DA: Effects
of a six month treatment with selenomethionine
in patients with autoimmune thyroiditis.
Eur J Endocrinol 2003;148:389�393.
39 Meerts IA, Assink Y, Cenijn PH, Van Den
Berg JH, Weijers BM, Bergman A, Koeman
JH, Brouwer A: Placental transfer of a hydroxylated
polychlorinated biphenyl and effects
on fetal and maternal thyroid hormone
homeostasis in the rat. Toxicol Sci 2002;68:
361�372.
40 Boas M, Feldt-Rasmussen U, Skakkebaek
NE, Main KM: Environmental chemicals
and thyroid function. Eur J Endocrinol 2006;
154:599�611.
41 Utiger RD: Effects of smoking on thyroid
function. Eur J Endocrinol 1998;138:368�
369.
42 Prummel MF, Strieder T, Wiersinga WM:
The environment and autoimmune diseases.
Eur J Endocrinol 2004;150:605�618.
43 Pontikides N, Krassas GE: Influence of cigarette
smoking on thyroid function, goiter
formation and autoimmune thyroid disorders.
Hormones (Athens) 2002;1:91�98.
44 Gasparoni A, Autelli M, Ravagni-Probizer
MF, Bartoli A, Regazzi-Bonora M, Chirico
G, Rondini G: Effect of passive smoking on
thyroid function in infants. Eur J Endocrinol
1998;138:379�382.
45 Vestergaard P: Smoking and thyroid disorders
� a meta-analysis. Eur J Endocrinol
2002;146:153�161.
46 Weiss M, Ingbar SH, Winblad S, Kasper DL:
Demonstration of a saturable binding site for
thyrotropin in Yersinia enterocolitica . Science
1983;219:1331�1333.
47 Fernandez-Soto L, Gonzales A, Escobar-Jimenez
F, Vazquez R, Ocete E, Olea N, Salmeron
J: Increased risk of autoimmune thyroid
disease in hepatitis C vs B before, during and
after discontinuing interferon therapy. Arch
Intern Med 1998;158:1445�1448.
48 Testa A, Castaldi P, Fanti V, Fiore GF, Grieco
V, De Rosa A, Pazardjklian MG, De Rosa G:
Prevalence of HCV antibodies in autoimmune
thyroid disease. Eur Rev Med Pharmacol
Sci 2006;10:183�186.
49 Davies TF: Infection and autoimmune thyroid
disease. J Clin Enocrinol Metab 2008;93:
674�676.
50 Lazarus JH, John R, Bennie EH, Chalmers
RJ, Crockett G: Lithium therapy and thyroid
function: a long-term study. Psychol Med
1981;11:85�92.
51 Dayan CM: Stressful life events and Graves�
disease revisited. Clin Endocrinol (Oxf)
2001;55:13�14.
52 Heufelder AE, Goellner JR, Wenzel BE,
Bahn RS: Immunohistochemical detection
and localization of a 72-kilodalton heat
shock protein in autoimmune thyroid disease.
J Clin Endocrinol Metab 1992;74:724�
731.
53 Parcellier A, Gurbuxani S, Schmitt E, Solary
E, Garrido C: Heat shock proteins, cellular
chaperones that modulates mitochondrial
cell death pathways. Biochem Biophys Res
Commun 2003;304:505�512.
54 Gaston JS: Are heat shock proteins involved
in autoimmunity? Int Clin Lab Res 1992;22:
90�94.
55 Benvenga S: Benzodiazepine and remission
of Graves� disease. Thyroid 1996;6:659�660.
56 Adams D: How the immune system works
and why it causes autoimmune diseases. Immunol
Today 1998;17:300�303.
57 Pierce EN, Farwel AP, Braverman LE: Thyroiditis.
N Engl J Med 2003;348:2646�2655.
58 Badenhoop K: Microchimerism and the
model of postpartum thyroiditis; in Wiersinga
WM, Drexhage HA, Weetman AP, et
al (eds): The Thyroid and Autoimmunity:
Merck European Thyroid Symposium
Noordwijk 2006, June 15�18. Stuttgart,
Thieme, 2007, pp 99�103.
59 Szabolcs I: Clinical relevance of thyroid peroxidase
autoantibodies in euthyroid individuals;
in Wiersinga WM, Drexhage HA,
Weetman AP, et al (eds): The Thyroid and
Autoimmunity: Merck European Thyroid
Symposium Noordwijk 2006, June 15�18.
Stuttgart, Thieme, 2007, pp 133�142.
60 Saranac L, Miljkovic M, Stamenkovic H, Mileusnic-Milenovic
R, Petrovic G, Kamenov
B: Late onset transient thyroid dysfunction
in children born to mothers with autoimmune
thyroid disease. Facta Univ Ser Med
Biol 2003;10:52�56.

Close Accordion