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.


Regulation of Gene Expression by Fatty Acids for IBD

Regulation of Gene Expression by Fatty Acids for IBD

Dietary fat has several essential functions in the human body. First, it functions as a supply of energy and structural components for the cells and second, it functions as a regulator of gene expression, which influences lipid, carbohydrate, and protein metabolism, along with cell growth and differentiation. The effects of fatty acids on gene expression are cell-specific and influenced by structure and metabolism. Fatty acids interact with the genome. They regulate PPAR, and the activity or nuclear abundance like SREBP. Fatty acids bind directly with one another to regulate gene expression.

 

What’s the role of fatty acids towards disease pathogenesis?

 

Alternately, fatty acids behave on gene expression through their effects on specific enzyme-mediated pathways, such as cyclooxygenase, lipoxygenase, protein kinase C, or sphingomyelinase signal transduction pathways, or through pathways that require changes in tissue lipid to lipid raft composition which affect G-protein receptor or tyrosine kinase-linked receptor signaling. Additional definition of these fatty acid-regulated pathways can offer insight into the role dietary fat plays in human health as well as the beginning and growth of many chronic diseases, such as coronary artery disease and atherosclerosis, dyslipidemia and inflammation, obesity and diabetes, cancer, major depressive disorders, and schizophrenia. The effects of fatty acids on gene expression, however, have been widely described on inflammatory bowel disease, or IBD.

 

Fatty Acids and Gene Expression

 

The effect of fatty acids on gene expression was previously determined to result mainly from changes in tissue phospholipids or eicosanoid production. More recently, the discovery of nuclear receptors; such as peroxisome proliferator-activated receptors, or PPARs, and their regulation by fatty acids, has significantly altered this view. PPARs are ligand activated transcription factors that upon heterodimerization with the retinoic X receptor, or RXR, comprehend PPAR response elements in the promoter regions of different genes, that have an impact on gene transcription. PPARs bind various ligands, including nonsteroidal anti inflammatory medications, or NSAIDS, thiazolidinediones (antidiabetic agents) along with PUFAs and their metabolites. Several subtypes of the receptor are recognized (?,?,?) and are expressed in several different cells. PPAR? is extracted from the adrenal gland, with most of its numbers observed in the colon.

 

PPAR? has been implicated in the regulation of inflammation, and it has become a potential therapeutic goal in treating inflammatory diseases, such as IBD. It has been suggested that people with ulcerative colitis, or UC, have a mucosal deficit in PPAR? that could bring about the development of their own disease. Analysis of the mRNA and proteins within colonic biopsies demonstrated decreased levels of PPAR? in UC patients in comparison with Crohn’s patients or healthy subjects.

 

Using colon cancer lines, it has been demonstrated that PPAR ligands attenuate cytokine gene expression by inhibiting NF-?B via an I?B determined mechanism. Further research studies imply that PPAR activators inhibit COX2 by interruption with NF-?B. PPARs impair interactions with STAT and other signaling pathways as well as the AP-1 signaling pathway.

 

Animal studies support using PPAR for autoimmune inflammation. Inflammation decreased by ligands for PPAR. The direction of PPAR and RXR agonists synergistically reduced TNBS-induced colitis, together with improved macroscopic and histologic scores, reductions in TNF? and IL-1? mRNA, and diminished NF-?B DNA binding actions. Though clinical evidence is limited, the results of an open source research study with rosiglitazone, a PPAR? ligand as therapy for UC, demonstrated that 27 percent of patients achieved remission after 12 weeks of therapy. Thus, PPAR? ligands may represent a cure for UC, where double-blind, placebo-controlled, randomized trials have been warranted.

 

Of substantial curiosity, the capability to regulate PPAR nutritionally has been examined. Dietary PUFA demonstrated an impact during the regulation of transcription factors on gene expression. Fatty acid regulation of PPAR was originally detected by Gottlicher et al.. A choice of fatty acids, like eicosanoids, and metabolites are proven to activate PPAR. Both PPAR? and PPAR? bind mono- and polyunsaturated fatty acids. Thus, the anti inflammatory effects of n3 PUFA may entail PPAR and its interruption with NF?B, rather than only changes in eicosanoid synthesis.

 

Conclusion

 

Fatty acids regulate gene expression involved in lipid and energy metabolism. Polyunsaturated fatty acids, or PUFA, though not saturated or polyunsaturated FA, suppress the induction of lipogenic genes by inhibiting their expression and processing of SREBP-1c. This impact of PUFA suggests that SREBP-1c may regulate the synthesis of fatty acids to glycerolipids, among others. PPARalpha has a role in the adaptation to fasting by inducing ketogenesis in mitochondria. During fasting, fatty acids are considered as ligands of PPARalpha. Dietary PUFA, except for 18:2 n-6, are extremely prone to induce fatty acid oxidation enzymes through PPARalpha because of specific mechanisms. Signaling functions of PPARalpha pPARalpha is needed for controlling the synthesis of fatty acids. Further research is needed to conclude the full effects of fatty acids in relation to the regulation of transcription factors for gene expression in inflammatory bowel disease, or IBD.

 

Information referenced from the National Center for Biotechnology Information (NCBI) and the National University of Health Sciences. 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

 

WELLNESS TOPIC: EXTRA EXTRA: Managing Workplace Stress

 

 

Blank
References
1.�Liu Y, van Kruiningen HJ, West AB, Cartun RW, Cortot A, Colombel JF. Immunocytochemical evidence of Listeria, Escherichia coli, and Streptococcus antigens in Crohn’s disease.�Gastroenterology.�1995;108:1396�1404.�[PubMed]
2.�Sartor R.�Microbial factors in the pathogenesis of Crohn’s disease, ulcerative colitis and experimental intestinal inflammation.�Baltimore: Williams & Wilkins; 1995.
3.�Wakefield AJ, Ekbom A, Dhillon AP, Pittilo RM, Pounder RE. Crohn’s disease: pathogenesis and persistent measles virus infection.�Gastroenterology.�1995;108:911�916.�[PubMed]
4.�Sartor RB. Current concepts of the etiology and pathogenesis of ulcerative colitis and Crohn’s disease.�Gastroenterol Clin North Am.�1995;24:475�507.�[PubMed]
5.�Sartor RB. Pathogenesis and immune mechanisms of chronic inflammatory bowel diseases.�Am J Gastroenterol.�1997;92:5S�11S.�[PubMed]
6.�MacDermott RP. Alterations in the mucosal immune system in ulcerative colitis and Crohn’s disease.�Med Clin North Am.�1994;78:1207�1231.�[PubMed]
7.�Podolsky DK. Inflammatory bowel disease (1)�N Engl J Med.�1991;325:928�937.�[PubMed]
8.�Podolsky DK. Inflammatory bowel disease (2)�N Engl J Med.�1991;325:1008�1016.�[PubMed]
9.�Yang H, Rotter J.�The genetics of inflammatory disease.�Baltimore: Williams & Wilkins; 1994.
10.�Wurzelmann JI, Lyles CM, Sandler RS. Childhood infections and the risk of inflammatory bowel disease.�Dig Dis Sci.�1994;39:555�560.�[PubMed]
11.�Knoflach P, Park BH, Cunningham R, Weiser MM, Albini B. Serum antibodies to cow’s milk proteins in ulcerative colitis and Crohn’s disease.�Gastroenterology.�1987;92:479�485.�[PubMed]
12.�De Palma GD, Catanzano C. Removable self-expanding metal stents: a pilot study for treatment of achalasia of the esophagus.�Endoscopy.�1998;30:S95�S96.�[PubMed]
13.�Bernstein CN, Ament M, Artinian L, Ridgeway J, Shanahan F. Milk tolerance in adults with ulcerative colitis.�Am J Gastroenterol.�1994;89:872�877.�[PubMed]
14.�Matsui T, Iida M, Fujishima M, Imai K, Yao T. Increased sugar consumption in Japanese patients with Crohn’s disease.�Gastroenterol Jpn.�1990;25:271.�[PubMed]
15.�Kelly DG, Fleming CR. Nutritional considerations in inflammatory bowel diseases.�Gastroenterol Clin North Am.�1995;24:597�611.�[PubMed]
16.�Geerling BJ, Dagnelie PC, Badart-Smook A, Russel MG, Stockbr�gger RW, Brummer RJ. Diet as a risk factor for the development of ulcerative colitis.�Am J Gastroenterol.�2000;95:1008�1013.�[PubMed]
17.�Dudrick SJ, Latifi R, Schrager R. Nutritional management of inflammatory bowel disease.�Surg Clin North Am.�1991;71:609�623.�[PubMed]
18.�D’Odorico A, Bortolan S, Cardin R, D’Inca’ R, Martines D, Ferronato A, Sturniolo GC. Reduced plasma antioxidant concentrations and increased oxidative DNA damage in inflammatory bowel disease.�Scand J Gastroenterol.�2001;36:1289�1294.�[PubMed]
19.�Reimund JM, Hirth C, Koehl C, Baumann R, Duclos B. Antioxidant and immune status in active Crohn’s disease. A possible relationship.�Clin Nutr.�2000;19:43�48.�[PubMed]
20.�Romagnuolo J, Fedorak RN, Dias VC, Bamforth F, Teltscher M. Hyperhomocysteinemia and inflammatory bowel disease: prevalence and predictors in a cross-sectional study.�Am J Gastroenterol.�2001;96:2143�2149.�[PubMed]
21.�Lewis JD, Fisher RL. Nutrition support in inflammatory bowel disease.�Med Clin North Am.�1994;78:1443�1456.�[PubMed]
22.�Azcue M, Rashid M, Griffiths A, Pencharz PB. Energy expenditure and body composition in children with Crohn’s disease: effect of enteral nutrition and treatment with prednisolone.�Gut.�1997;41:203�208.[PMC free article][PubMed]
23.�Mingrone G, Capristo E, Greco AV, Benedetti G, De Gaetano A, Tataranni PA, Gasbarrini G. Elevated diet-induced thermogenesis and lipid oxidation rate in Crohn disease.�Am J Clin Nutr.�1999;69:325�330.[PubMed]
24.�Bjarnason I, Macpherson A, Mackintosh C, Buxton-Thomas M, Forgacs I, Moniz C. Reduced bone density in patients with inflammatory bowel disease.�Gut.�1997;40:228�233.�[PMC free article][PubMed]
25.�Griffiths AM, Nguyen P, Smith C, MacMillan JH, Sherman PM. Growth and clinical course of children with Crohn’s disease.�Gut.�1993;34:939�943.�[PMC free article][PubMed]
26.�Fischer JE, Foster GS, Abel RM, Abbott WM, Ryan JA. Hyperalimentation as primary therapy for inflammatory bowel disease.�Am J Surg.�1973;125:165�175.�[PubMed]
27.�Reilly J, Ryan JA, Strole W, Fischer JE. Hyperalimentation in inflammatory bowel disease.�Am J Surg.�1976;131:192�200.�[PubMed]
28.�Ganem D, Schneider RJ. Hepadnaviridae: The viruses and their replication. In: Knipe DM, Howley PM, editors.�Fields Virology. Volume 2.�Philadelphia: Lippincott, Williams & Wilkins; 2001. pp. 2923�2969.
29.�Jones VA, Dickinson RJ, Workman E, Wilson AJ, Freeman AH, Hunter JO. Crohn’s disease: maintenance of remission by diet.�Lancet.�1985;2:177�180.�[PubMed]
30.�Suzuki I, Kiyono H, Kitamura K, Green DR, McGhee JR. Abrogation of oral tolerance by contrasuppressor T cells suggests the presence of regulatory T-cell networks in the mucosal immune system.�Nature.�1986;320:451�454.�[PubMed]
31.�Ostro MJ, Greenberg GR, Jeejeebhoy KN. Total parenteral nutrition and complete bowel rest in the management of Crohn’s disease.�JPEN J Parenter Enteral Nutr.�1985;9:280�287.�[PubMed]
32.�Matuchansky C. Parenteral nutrition in inflammatory bowel disease.�Gut.�1986;27 Suppl 1:81�84.[PMC free article][PubMed]
33.�Payne-James JJ, Silk DB. Total parenteral nutrition as primary treatment in Crohn’s disease–RIP?�Gut.�1988;29:1304�1308.�[PMC free article][PubMed]
34.�Shiloni E, Coronado E, Freund HR. Role of total parenteral nutrition in the treatment of Crohn’s disease.�Am J Surg.�1989;157:180�185.�[PubMed]
35.�Dickinson RJ, Ashton MG, Axon AT, Smith RC, Yeung CK, Hill GL. Controlled trial of intravenous hyperalimentation and total bowel rest as an adjunct to the routine therapy of acute colitis.�Gastroenterology.�1980;79:1199�1204.�[PubMed]
36.�McIntyre PB, Powell-Tuck J, Wood SR, Lennard-Jones JE, Lerebours E, Hecketsweiler P, Galmiche JP, Colin R. Controlled trial of bowel rest in the treatment of severe acute colitis.�Gut.�1986;27:481�485.[PMC free article][PubMed]
37.�Greenberg GR, Fleming CR, Jeejeebhoy KN, Rosenberg IH, Sales D, Tremaine WJ. Controlled trial of bowel rest and nutritional support in the management of Crohn’s disease.�Gut.�1988;29:1309�1315.[PMC free article][PubMed]
38.�Hughes CA, Bates T, Dowling RH. Cholecystokinin and secretin prevent the intestinal mucosal hypoplasia of total parenteral nutrition in the dog.�Gastroenterology.�1978;75:34�41.�[PubMed]
39.�Stratton RJ, Smith TR. Role of enteral and parenteral nutrition in the patient with gastrointestinal and liver disease.�Best Pract Res Clin Gastroenterol.�2006;20:441�466.�[PubMed]
40.�O’Sullivan M, O’Morain C. Nutrition in inflammatory bowel disease.�Best Pract Res Clin Gastroenterol.�2006;20:561�573.�[PubMed]
41.�Gonz�lez-Huix F, Fern�ndez-Ba�ares F, Esteve-Comas M, Abad-Lacruz A, Cabr� E, Acero D, Figa M, Guilera M, Humbert P, de Le�n R. Enteral versus parenteral nutrition as adjunct therapy in acute ulcerative colitis.�Am J Gastroenterol.�1993;88:227�232.�[PubMed]
42.�Voitk AJ, Echave V, Feller JH, Brown RA, Gurd FN. Experience with elemental diet in the treatment of inflammatory bowel disease. Is this primary therapy?�Arch Surg.�1973;107:329�333.�[PubMed]
43.�Axelsson C, Jarnum S. Assessment of the therapeutic value of an elemental diet in chronic inflammatory bowel disease.�Scand J Gastroenterol.�1977;12:89�95.�[PubMed]
44.�Lochs H, Steinhardt HJ, Klaus-Wentz B, Zeitz M, Vogelsang H, Sommer H, Fleig WE, Bauer P, Schirrmeister J, Malchow H. Comparison of enteral nutrition and drug treatment in active Crohn’s disease. Results of the European Cooperative Crohn’s Disease Study. IV.�Gastroenterology.�1991;101:881�888.[PubMed]
45.�Malchow H, Steinhardt HJ, Lorenz-Meyer H, Strohm WD, Rasmussen S, Sommer H, Jarnum S, Brandes JW, Leonhardt H, Ewe K. Feasibility and effectiveness of a defined-formula diet regimen in treating active Crohn’s disease. European Cooperative Crohn’s Disease Study III.�Scand J Gastroenterol.�1990;25:235�244.�[PubMed]
46.�O’Brien CJ, Giaffer MH, Cann PA, Holdsworth CD. Elemental diet in steroid-dependent and steroid-refractory Crohn’s disease.�Am J Gastroenterol.�1991;86:1614�1618.�[PubMed]
47.�Okada M, Yao T, Yamamoto T, Takenaka K, Imamura K, Maeda K, Fujita K. Controlled trial comparing an elemental diet with prednisolone in the treatment of active Crohn’s disease.�Hepatogastroenterology.�1990;37:72�80.�[PubMed]
48.�O’Mor�in C, Segal AW, Levi AJ. Elemental diet as primary treatment of acute Crohn’s disease: a controlled trial.�Br Med J (Clin Res Ed)�1984;288:1859�1862.�[PMC free article][PubMed]
49.�Raouf AH, Hildrey V, Daniel J, Walker RJ, Krasner N, Elias E, Rhodes JM. Enteral feeding as sole treatment for Crohn’s disease: controlled trial of whole protein v amino acid based feed and a case study of dietary challenge.�Gut.�1991;32:702�707.�[PMC free article][PubMed]
50.�Rocchio MA, Cha CJ, Haas KF, Randall HT. Use of chemically defined diets in the management of patients with acute inflammatory bowel disease.�Am J Surg.�1974;127:469�475.�[PubMed]
51.�Saverymuttu S, Hodgson HJ, Chadwick VS. Controlled trial comparing prednisolone with an elemental diet plus non-absorbable antibiotics in active Crohn’s disease.�Gut.�1985;26:994�998.�[PMC free article][PubMed]
52.�Teahon K, Bjarnason I, Pearson M, Levi AJ. Ten years’ experience with an elemental diet in the management of Crohn’s disease.�Gut.�1990;31:1133�1137.�[PMC free article][PubMed]
53.�Teahon K, Smethurst P, Pearson M, Levi AJ, Bjarnason I. The effect of elemental diet on intestinal permeability and inflammation in Crohn’s disease.�Gastroenterology.�1991;101:84�89.�[PubMed]
54.�Heuschkel RB, Menache CC, Megerian JT, Baird AE. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children.�J Pediatr Gastroenterol Nutr.�2000;31:8�15.�[PubMed]
55.�Sanderson IR, Boulton P, Menzies I, Walker-Smith JA. Improvement of abnormal lactulose/rhamnose permeability in active Crohn’s disease of the small bowel by an elemental diet.�Gut.�1987;28:1073�1076.[PMC free article][PubMed]
56.�Sanderson IR, Udeen S, Davies PS, Savage MO, Walker-Smith JA. Remission induced by an elemental diet in small bowel Crohn’s disease.�Arch Dis Child.�1987;62:123�127.�[PMC free article][PubMed]
57.�Ruemmele FM, Roy CC, Levy E, Seidman EG. Nutrition as primary therapy in pediatric Crohn’s disease: fact or fantasy?�J Pediatr.�2000;136:285�291.�[PubMed]
58.�O’Morain C, O’Sullivan M. Nutritional support in Crohn’s disease: current status and future directions.�J Gastroenterol.�1995;30 Suppl 8:102�107.�[PubMed]
59.�Rigaud D, Cosnes J, Le Quintrec Y, Ren� E, Gendre JP, Mignon M. Controlled trial comparing two types of enteral nutrition in treatment of active Crohn’s disease: elemental versus polymeric diet.�Gut.�1991;32:1492�1497.�[PMC free article][PubMed]
60.�Royall D, Wolever TM, Jeejeebhoy KN. Evidence for colonic conservation of malabsorbed carbohydrate in short bowel syndrome.�Am J Gastroenterol.�1992;87:751�756.�[PubMed]
61.�Giaffer MH, North G, Holdsworth CD. Controlled trial of polymeric versus elemental diet in treatment of active Crohn’s disease.�Lancet.�1990;335:816�819.�[PubMed]
62.�Verma S, Kirkwood B, Brown S, Giaffer MH. Oral nutritional supplementation is effective in the maintenance of remission in Crohn’s disease.�Dig Liver Dis.�2000;32:769�774.�[PubMed]
63.�Levine GM, Deren JJ, Steiger E, Zinno R. Role of oral intake in maintenance of gut mass and disaccharide activity.�Gastroenterology.�1974;67:975�982.�[PubMed]
64.�Weser E, Heller R, Tawil T. Stimulation of mucosal growth in the rat ileum by bile and pancreatic secretions after jejunal resection.�Gastroenterology.�1977;73:524�529.�[PubMed]
65.�Fell JM, Paintin M, Arnaud-Battandier F, Beattie RM, Hollis A, Kitching P, Donnet-Hughes A, MacDonald TT, Walker-Smith JA. Mucosal healing and a fall in mucosal pro-inflammatory cytokine mRNA induced by a specific oral polymeric diet in paediatric Crohn’s disease.�Aliment Pharmacol Ther.�2000;14:281�289.�[PubMed]
66.�Souba WW, Smith RJ, Wilmore DW. Glutamine metabolism by the intestinal tract.�JPEN J Parenter Enteral Nutr.�1985;9:608�617.�[PubMed]
67.�Windmueller HG, Spaeth AE. Uptake and metabolism of plasma glutamine by the small intestine.�J Biol Chem.�1974;249:5070�5079.�[PubMed]
68.�Higashiguchi T, Hasselgren PO, Wagner K, Fischer JE. Effect of glutamine on protein synthesis in isolated intestinal epithelial cells.�JPEN J Parenter Enteral Nutr.�1993;17:307�314.�[PubMed]
69.�Burke DJ, Alverdy JC, Aoys E, Moss GS. Glutamine-supplemented total parenteral nutrition improves gut immune function.�Arch Surg.�1989;124:1396�1399.�[PubMed]
70.�Souba WW, Herskowitz K, Klimberg VS, Salloum RM, Plumley DA, Flynn TC, Copeland EM. The effects of sepsis and endotoxemia on gut glutamine metabolism.�Ann Surg.�1990;211:543�549; discussion 543-551;.�[PMC free article][PubMed]
71.�Den Hond E, Hiele M, Peeters M, Ghoos Y, Rutgeerts P. Effect of long-term oral glutamine supplements on small intestinal permeability in patients with Crohn’s disease.�JPEN J Parenter Enteral Nutr.�1999;23:7�11.�[PubMed]
72.�Akobeng AK, Miller V, Stanton J, Elbadri AM, Thomas AG. Double-blind randomized controlled trial of glutamine-enriched polymeric diet in the treatment of active Crohn’s disease.�J Pediatr Gastroenterol Nutr.�2000;30:78�84.�[PubMed]
73.�Jacobs LR, Lupton JR. Effect of dietary fibers on rat large bowel mucosal growth and cell proliferation.�Am J Physiol.�1984;246:G378�G385.�[PubMed]
74.�Spaeth G, Berg RD, Specian RD, Deitch EA. Food without fiber promotes bacterial translocation from the gut.�Surgery.�1990;108:240�246; discussion 246-247;.�[PubMed]
75.�Roediger WE, Moore A. Effect of short-chaim fatty acid on sodium absorption in isolated human colon perfused through the vascular bed.�Dig Dis Sci.�1981;26:100�106.�[PubMed]
76.�Sakata T. Stimulatory effect of short-chain fatty acids on epithelial cell proliferation in the rat intestine: a possible explanation for trophic effects of fermentable fibre, gut microbes and luminal trophic factors.�Br J Nutr.�1987;58:95�103.�[PubMed]
77.�Roediger WE. The colonic epithelium in ulcerative colitis: an energy-deficiency disease?�Lancet.�1980;2:712�715.�[PubMed]
78.�Chapman MA, Grahn MF, Boyle MA, Hutton M, Rogers J, Williams NS. Butyrate oxidation is impaired in the colonic mucosa of sufferers of quiescent ulcerative colitis.�Gut.�1994;35:73�76.[PMC free article][PubMed]
79.�Den Hond E, Hiele M, Evenepoel P, Peeters M, Ghoos Y, Rutgeerts P. In vivo butyrate metabolism and colonic permeability in extensive ulcerative colitis.�Gastroenterology.�1998;115:584�590.�[PubMed]
80.�Simpson EJ, Chapman MA, Dawson J, Berry D, Macdonald IA, Cole A. In vivo measurement of colonic butyrate metabolism in patients with quiescent ulcerative colitis.�Gut.�2000;46:73�77.[PMC free article][PubMed]
81.�Tappenden KA, Thomson AB, Wild GE, McBurney MI. Short-chain fatty acid-supplemented total parenteral nutrition enhances functional adaptation to intestinal resection in rats.�Gastroenterology.�1997;112:792�802.�[PubMed]
82.�Senagore AJ, MacKeigan JM, Scheider M, Ebrom JS. Short-chain fatty acid enemas: a cost-effective alternative in the treatment of nonspecific proctosigmoiditis.�Dis Colon Rectum.�1992;35:923�927.[PubMed]
83.�Segain JP, Raingeard de la Bl�ti�re D, Bourreille A, Leray V, Gervois N, Rosales C, Ferrier L, Bonnet C, Blotti�re HM, Galmiche JP. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease.�Gut.�2000;47:397�403.�[PMC free article][PubMed]
84.�Aslan A, Triadafilopoulos G. Fish oil fatty acid supplementation in active ulcerative colitis: a double-blind, placebo-controlled, crossover study.�Am J Gastroenterol.�1992;87:432�437.�[PubMed]
85.�Shoda R, Matsueda K, Yamato S, Umeda N. Epidemiologic analysis of Crohn disease in Japan: increased dietary intake of n-6 polyunsaturated fatty acids and animal protein relates to the increased incidence of Crohn disease in Japan.�Am J Clin Nutr.�1996;63:741�745.�[PubMed]
86.�Vilaseca J, Salas A, Guarner F, Rodr�guez R, Mart�nez M, Malagelada JR. Dietary fish oil reduces progression of chronic inflammatory lesions in a rat model of granulomatous colitis.�Gut.�1990;31:539�544.�[PMC free article][PubMed]
87.�Campos FG, Waitzberg DL, Habr-Gama A, Logullo AF, Noronha IL, Jancar S, Torrinhas RS, F�rst P. Impact of parenteral n-3 fatty acids on experimental acute colitis.�Br J Nutr.�2002;87 Suppl 1:S83�S88.[PubMed]
88.�Loeschke K, Ueberschaer B, Pietsch A, Gruber E, Ewe K, Wiebecke B, Heldwein W, Lorenz R. n-3 fatty acids only delay early relapse of ulcerative colitis in remission.�Dig Dis Sci.�1996;41:2087�2094.[PubMed]
89.�Belluzzi A, Brignola C, Campieri M, Pera A, Boschi S, Miglioli M. Effect of an enteric-coated fish-oil preparation on relapses in Crohn’s disease.�N Engl J Med.�1996;334:1557�1560.�[PubMed]
90.�Hawthorne AB, Daneshmend TK, Hawkey CJ, Belluzzi A, Everitt SJ, Holmes GK, Malkinson C, Shaheen MZ, Willars JE. Treatment of ulcerative colitis with fish oil supplementation: a prospective 12 month randomised controlled trial.�Gut.�1992;33:922�928.�[PMC free article][PubMed]
91.�Hillier K, Jewell R, Dorrell L, Smith CL. Incorporation of fatty acids from fish oil and olive oil into colonic mucosal lipids and effects upon eicosanoid synthesis in inflammatory bowel disease.�Gut.�1991;32:1151�1155.�[PMC free article][PubMed]
92.�Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma)�J Biol Chem.�1995;270:12953�12956.�[PubMed]
93.�Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs.�J Biol Chem.�1997;272:3406�3410.�[PubMed]
94.�Delerive P, Furman C, Teissier E, Fruchart J, Duriez P, Staels B. Oxidized phospholipids activate PPARalpha in a phospholipase A2-dependent manner.�FEBS Lett.�2000;471:34�38.�[PubMed]
95.�Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma.�Proc Natl Acad Sci USA.�1997;94:4318�4323.�[PMC free article][PubMed]
96.�Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta.�Proc Natl Acad Sci USA.�1997;94:4312�4317.�[PMC free article][PubMed]
97.�Mans�n A, Guardiola-Diaz H, Rafter J, Branting C, Gustafsson JA. Expression of the peroxisome proliferator-activated receptor (PPAR) in the mouse colonic mucosa.�Biochem Biophys Res Commun.�1996;222:844�851.�[PubMed]
98.�Desreumaux P, Ernst O, Geboes K, Gambiez L, Berrebi D, M�ller-Alouf H, Hafraoui S, Emilie D, Ectors N, Peuchmaur M, et al. Inflammatory alterations in mesenteric adipose tissue in Crohn’s disease.�Gastroenterology.�1999;117:73�81.�[PubMed]
99.�Su CG, Wen X, Bailey ST, Jiang W, Rangwala SM, Keilbaugh SA, Flanigan A, Murthy S, Lazar MA, Wu GD. A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response.�J Clin Invest.�1999;104:383�389.�[PMC free article][PubMed]
100.�Ricote M, Huang J, Fajas L, Li A, Welch J, Najib J, Witztum JL, Auwerx J, Palinski W, Glass CK. Expression of the peroxisome proliferator-activated receptor gamma (PPARgamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein.�Proc Natl Acad Sci USA.�1998;95:7614�7619.�[PMC free article][PubMed]
101.�Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, et al. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators.�Nature.�1998;393:790�793.�[PubMed]
102.�Marx N, Bourcier T, Sukhova GK, Libby P, Plutzky J. PPARgamma activation in human endothelial cells increases plasminogen activator inhibitor type-1 expression: PPARgamma as a potential mediator in vascular disease.�Arterioscler Thromb Vasc Biol.�1999;19:546�551.�[PubMed]
103.�Delerive P, Martin-Nizard F, Chinetti G, Trottein F, Fruchart JC, Najib J, Duriez P, Staels B. Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway.�Circ Res.�1999;85:394�402.�[PubMed]
104.�Sakai M, Matsushima-Hibiya Y, Nishizawa M, Nishi S. Suppression of rat glutathione transferase P expression by peroxisome proliferators: interaction between Jun and peroxisome proliferator-activated receptor alpha.�Cancer Res.�1995;55:5370�5376.�[PubMed]
105.�Zhou YC, Waxman DJ. STAT5b down-regulates peroxisome proliferator-activated receptor alpha transcription by inhibition of ligand-independent activation function region-1 trans-activation domain.�J Biol Chem.�1999;274:29874�29882.�[PubMed]
106.�Desreumaux P, Dubuquoy L, Nutten S, Peuchmaur M, Englaro W, Schoonjans K, Derijard B, Desvergne B, Wahli W, Chambon P, et al. Attenuation of colon inflammation through activators of the retinoid X receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma) heterodimer. A basis for new therapeutic strategies.�J Exp Med.�2001;193:827�838.�[PMC free article][PubMed]
107.�Lewis JD, Lichtenstein GR, Stein RB, Deren JJ, Judge TA, Fogt F, Furth EE, Demissie EJ, Hurd LB, Su CG, et al. An open-label trial of the PPAR-gamma ligand rosiglitazone for active ulcerative colitis.�Am J Gastroenterol.�2001;96:3323�3328.�[PubMed]
108.�G�ttlicher M, Widmark E, Li Q, Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor.�Proc Natl Acad Sci USA.�1992;89:4653�4657.[PMC free article][PubMed]
Close Accordion
Assessment and Treatment of Quadratus Lumborum

Assessment and Treatment of Quadratus Lumborum

These assessment and treatment recommendations represent a synthesis of information derived from personal clinical experience and from the numerous sources which are cited, or are based on the work of researchers, clinicians and therapists who are named (Basmajian 1974, Cailliet 1962, Dvorak & Dvorak 1984, Fryette 1954, Greenman 1989, 1996, Janda 1983, Lewit 1992, 1999, Mennell 1964, Rolf 1977, Williams 1965).

 

Clinical Application of Neuromuscular Techniques: Quadratus Lumborum

 

Assessment of Shortness in Quadratus Lumborum (Figure 4.21)

 

Figure 4 21 Palpation Assessment for Quadratus Lumborum Overactivity Image 1

 

Figure 4.21 Palpation assessment for quadratus lumborum overactivity. The muscle is palpated, as is gluteus medius, during abduction of the leg. The correct firing sequence should be gluteus, followed at around 25� elevation by quadratus. If there is an immediate �grabbing� action by quadratus it indicates overactivity, and therefore stress, so shortness can be assumed (see details of similar functional assessments in Ch. 5).

 

Review Lewit�s functional palpation test described under the heading assessment and treatment of tensor fascia lata.

 

When the leg of the side-lying patient is abducted, and the practitioner�s palpating hand senses that quadratus becomes involved in this process before the leg has reached at least 25� of elevation, then it is clear that quadratus is overactive. If it has been overactive for any length of time then it is almost certainly hypertonic and short, and a need for MET can be assumed.

 

Quadratus lumborum test (a) (See also Fig. 5.11A, B.) The patient is side-lying and is asked to take the upper arm over the head to grasp the top edge of the table, �opening out� the lumbar area. The practitioner stands facing the back of the patient, and has easy access for palpation of quadratus lumborum�s lateral border � a major trigger point site (Travell & Simons 1992) � with the cephalad hand.

 

Activity of quadratus is tested (palpated for) with the cephalad hand as the leg is abducted, while also palpating gluteus medius with the caudad hand. If the muscles act simultaneously, or if quadratus fires first, then it is stressed, probably short, and will benefit from stretching.

 

Quadratus lumborum test (b) The patient stands, back towards crouching practitioner. Any leg length disparity (based on pelvic crest height) is equalised by using a book or pad under the short leg side heel. With the patient�s feet shoulder-width apart, a pure sidebending is requested, so that the patient runs a hand down the lateral thigh/calf. (Normal level of sidebending excursion allows the fingertips to reach to just below the knee.) (See Fig. 3.2A, B, C.)

 

The side to which the fingertips travel furthest is assessed. If sidebending to one side is limited then quadratus on the opposite side is probably short. Combined evidence from palpation (test a) and this sidebending test indicate whether or not it is necessary to treat quadratus.

 

Box 4.8 Notes on Quadratus Lumborum

 

  • Norris (2000) describes the divided roles in which quadratus is involved: The quadratus lumborum has been shown to be significant as a stabiliser in lumbar spine movements (McGill et al 1996) while tightening has also been described (Janda 1983). It seems likely that the muscle may act functionally differently in its medial and lateral portions, with the medial portion being more active as a stabiliser of the lumbar spine, and the lateral more active as a mobiliser [see stabiliser/mobiliser discussion Ch. 2]. Such subdivision is seen in a number of other muscles for example the gluteus medius where the posterior fibres are more posturally involved (Jull 1994) the internal oblique where the posterior fibres attaching to the lateral raphe are considered stabilisers (Bergmark 1989) the external oblique where the lateral fibres work during flexion in parallel with the rectus abdominis (Kendall et al 1993).
  • Janda (1983) observes that, when the patient is sidebending (as in method (b)) �when the lumbar spine appears straight, with compensatory motion occurring only from the thoracolumbar region upwards, tightness of quadratus lumborum may be suspected�. This �whole lumbar spine� involvement differs from a segmental restriction which would probably involve only a part of the lumbar spine.
  • Quadratus fibres merge with the diaphragm (as do those of psoas), which makes involvement in respiratory dysfunction a possibility since it plays a role in exhalation, both via this merging and by its attachment to the 12th rib.
  • Shortness of quadratus, or the presence of trigger points, can result in pain in the lower ribs and along the iliac crest if the lateral fibres are affected. Shortness of the medial fibres, or the presence of trigger points, can produce pain in the sacroiliac joint and the buttock.
  • Bilateral contraction produces extension and unilateral contraction produces extension and sidebending to the same side.
  • The important transition region, the lumbodorsal junction (LDJ), is the only one in the spine in which two mobile structures meet, and dysfunction results in alteration of the quality of motion between these structures (upper and lower trunk/dorsal and lumbar spines). In dysfunction there is often a degree of spasm or tightness in the muscles which stabilise the region, notably: psoas and erector spinae of the thoracolumbar region, as well as quadratus lumborum and rectus abdominis.
  • Symptomatic differential diagnosis of muscle involvement at the LDJ is possible as follows: psoas involvement usually triggers abdominal pain if severe and produces flexion of the hip and the typical antalgesic posture of lumbago; erector spinae involvement produces low back pain at its caudad end of attachment and interscapular pain at its thoracic attachment (as far up as the mid-thoracic level); quadratus lumborum involvement causes lumbar pain and pain at the attachment of the iliac crest and lower ribs; and rectus abdominis contraction may mimic abdominal pain and result in pain at the attachments at the pubic symphysis and the xiphoid process, as well as forwardbending of the trunk and restricted ability to extend the spine.

 

There is seldom pain at the site of the lesion in LDJ dysfunction. Lewit (1992) points out that even if a number of these muscles are implicated, it is seldom necessary, using PIR methods, to treat them all since, as the muscles most involved (discovered by tests for shortness, overactivity, sensitivity and direct palpation) are stretched and normalised, so will others begin automatically to normalise.

 

MET for Shortness in Quadratus Lumborum (�banana�)

 

Quadratus lumborum MET method (a) (Fig. 4.22) The patient lies supine with the feet crossed (the side to be treated crossed under the non-treated side leg) at the ankle. The patient is arranged in a light sidebend, away from the side to be treated, so that the pelvis is towards that side, and the feet and head away from that side (�banana shaped�). As this sidebend is being achieved the affected quadratus can be palpated for bind so that the barrier is correctly identified.

 

Figure 4 22 MET Treatment of Quadratus Lumborum Utilizing Banana Position Image 2

 

Figure 4.22 MET treatment of quadratus lumborum utilising �banana� position.

 

The patient�s heels are placed just off the side of the table, anchoring the lower extremities and pelvis. The patient places the arm of the side to be treated behind her neck as the practitioner, standing on the side opposite that to be treated, slides his cephalad hand under the patient�s shoulders to grasp the treated side axilla. The patient grasps the practitioner�s cephalad arm at the elbow, with the treated side hand, making the contact more secure.

 

The patient�s treated side elbow should, at this stage, be pointing superiorly. The practitioner�s caudad hand is placed firmly but carefully on the anterior superior iliac spine, on the side to be treated. The patient is instructed to very lightly sidebend towards the treated side. This should produce an isometric contraction in quadratus lumborum on the side to be treated.

 

After 7 seconds the patient is asked to relax completely, and then to sidebend towards the nontreated side, as the practitioner simultaneously transfers his bodyweight from the cephalad leg to the caudad leg and leans backwards slightly, in order to sidebend the patient. This effectively stretches quadratus lumborum. The stretch is held for 15�20 seconds, allowing a lengthening of shortened musculature in the region. Repeat as necessary.

 

Quadratus lumborum MET method (b) (Fig 4.23) The practitioner stands behind the side-lying patient, at waist level. The patient has the uppermost arm extended over the head to firmly grasp the top end of the table and, on an inhalation, abducts the uppermost leg until the practitioner palpates strong quadratus activity (elevation of around 30� usually).

 

Figure 4 23 MET Treatment of Quadratus Lumborum Image 3

 

Figure 4.23 MET treatment of quadratus lumborum. Note that it is important after the isometric contraction (sustained raised/abducted leg) that the muscle be eased into stretch, avoiding any defensive or protective resistance which sudden movement might produce. For this reason, body weight rather than arm strength should be used to apply traction.

 

The patient holds the leg (and, if appropriate, the breath, see Box 4.2) isometrically in this manner, allowing gravity to provide resistance. After the 10-second (or so) contraction, the patient allows the leg to hang slightly behind him over the back of the table. The practitioner straddles this and, cradling the pelvis with both hands (fingers interlocked over crest of pelvis), leans back to take out all slack and to �ease the pelvis away from the lower ribs� during an exhalation.

 

The stretch should be held for between 10 and 30 seconds. (The method will only be successful if the patient is grasping the top edge of the table, so providing a fixed point from which the practitioner can induce stretch.)

 

Contraction followed by stretch is repeated once or twice more with raised leg in front of, and once or twice with raised leg behind the trunk in order to activate different fibres. The direction of stretch should be varied so that it is always in the same direction as the long axis of the abducted leg. This calls for the practitioner changing from the back to the front of the table for the best results. When the leg hangs to the back of the trunk the long fibres of the muscle are mainly affected; and when the leg hangs forward of the body the diagonal fibres are mainly involved.

 

Quadratus lumborum MET method (c) Gravity-induced postisometric relaxation of quadratus lumborum � self-treatment (See Fig. 3.2A�C and captions) The patient stands, legs apart, bending sideways. The patient inhales and slightly raises the trunk (a few centimetres) at the same time as looking (with the eyes only) away from the side to which side-flexion is taking place. On exhalation, the sidebend is allowed to slowly go further to its elastic limit, while the patient looks towards the floor, in the direction of the side-flexion. (Care is needed that very little, if any, forward or backward bending is taking place at this time.) This sequence is repeated a number of times.

 

Eye positions influence the tendency to flex and sidebend (eyes look down) and extend (eyes look up) (Lewit 1999). Gravity-induced stretches of this sort require holding the stretch position for at least as long as the contraction, and ideally longer. More repetitions may be needed with a large muscle such as quadratus, and home stretches should be advised several times daily. Quadratus lumborum MET method (d) The side-lying treatment of latissimus dorsi described below also provides an effective quadratus stretch when the stabilising hand rests on the pelvic crest (see Fig. 4.29).

 

Dr. Alex Jimenez offers an additional assessment and treatment of the hip flexors as a part of a referenced clinical application of neuromuscular techniques by Leon Chaitow and Judith Walker DeLany. 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

 

WELLNESS TOPIC: EXTRA EXTRA: Managing Workplace Stress

 

 

Glutamine, Fiber & Fatty Acid Intake for IBD

Glutamine, Fiber & Fatty Acid Intake for IBD

Inflammatory bowel disease, or IBD, is a term used to describe inflammation of the gastrointestinal mucosa of unknown etiology. There are a selection of hypotheses associated to the development and perpetuation of IBD. Three main theories emerge from the literature. The first implicates a persistent intestinal infection; the second demonstrates that the upcoming signs of IBD are due to a defective mucosal barrier to luminal antigens; and the next suggests a dysregulated host immune response to ubiquitous antigens.

 

What are the nutritional components, if any, behind inflammatory bowel disease?

 

It is believed that IBD has both genetic and environmental components, therefore it’s immunologically mediated. Information gathered from IBD patients showing cytokine profiles, permeability defects, response to treatment and natural history of disease, may indicate a heterogeneous group of disorders that fall under the headings of ulcerative colitis, or UC, and Crohn’s disease, or CD. Previous epidemiological data on diet in UC and CD are conflicting, partly as a result of the heterogeneity of those diseases, making it difficult to get reliable statistics and publication bias, such as in the case of negative structures from breastfeeding.

 

Glutamine, Fiber and Fatty Acids

 

Diets high in glutamine, a significant source of energy for enterocytes, in addition to being the preferred fuel of the small intestine, are used with varying success. Glutamine is bekieved to exert its trophic effects on the small intestine by increasing protein synthesis and producing alanine for enteric gluconeogenesis. There is proof that glutamine protects the small intestinal mucosa during acute disease. However, oral glutamine supplements do not restore to normal the increased intestinal permeability discovered in patients with CD and these supplements do not beneficially affect the sufferers’ CDAI or C-reactive protein, also abbreviated as CRP, levels. Similarly, a randomized controlled trial demonstrated no benefit was connected to the usage of glutamine-enriched polymeric formulas in children with CD.

 

In animal research studies, dietary fiber has been implicated in keeping the integrity of the intestine, as well as in preventing bacterial translocation from the gut to the mesenteric lymph nodes. Short-chain fatty acids (SCFA, C1 to C6 natural fatty acids), are created by the fermentation of dietary polysaccharides in the common anaerobic bacteria in the colon. These SCFA are a source of energy for the colonocytes, which together improve sodium and water absorption, and promote blood circulation. Decreased quantities of SCFA, particularly butyrate, and a defect in the oxidation of butyrate from colonocytes, are indicated as a mechanism in the pathogenesis of inflammatory bowel disease. Evidence to support that concept requires the observation of the oxidation of C-labelled butyrate, demonstrated to decrease in patients with active UC in comparison with healthy controls. However, researchers have failed to reveal the differences between UC patients and controls in the oxidation of rectally administered C-labelled butyrate.

 

TPN supplemented with SCFA improved function adaptation to intestinal resection in rats. It remains to be discovered when patients with short bowel syndrome may make the most of SCFA.

 

Butyrate (C4 fatty acid) administered to UC patients contributed to remission levels like corticosteroids and mesalamine. In patients with CD, both intestinal biopsies and lamina propria cells packaged with butyrate had substantially decreased levels of inflammatory cytokines (TNF), possibly due to a reduction in NF?B stimulation and I?B degradation.

 

Eicosanoids are inflammatory mediators, which have also been implicated in the pathogenesis of chronic inflammatory damage in the intestine. Specimens from patients with IBD show enhanced eicosanoid formation. High dietary intake of omega-6 polyunsaturated fatty acids, abbreviated as PUFAs, which reduces omega-3 intake, and may contribute to IBD development. The benefits of fish oil, which contain n3 fatty acids, that were shown in certain inflammatory disorders, such as psoriasis and rheumatoid arthritis. Epidemiological observations of this very low prevalence of IBD in Japanese and Inuit populations consuming substantial n3 fatty acid fish provided a justification for utilizing n3 fatty acids in IBD. The n3 fatty acids are considered to compete with n6 fatty acids as precursors of eicosanoid synthesis. The n3 products reveal a series of 5 leukotrienes, which have considerably less physiological activity when compared with the arachidonate established series 4 counterparts. In addition, fish oil might have an anti inflammatory effect.

 

Rats fed with fish oil that had TNBS-induced inflammatory lesions in the intestine showed less prostaglandin- and leukotriene-mediated resistant response. Parenteral lipid emulsions enhanced with n3 fatty acids reduce diarrhea, weaken morphological changes and decreased colonic concentrations of inflammatory mediators in an animal model of acetic acid induced colitis.

 

Loeschke et al conducted a placebo-controlled trial of n3 fatty acids in preventing relapse in UC. Patients in remission who got n3 fatty acids experienced fewer relapses than did those receiving placebo. Unfortunately, the favorable results of this research study did not last throughout the total amount of the two year research, possibly due to diminished compliance punctually. In a multicenter placebo controlled relapse prevention trial, Belluzzi et al found a significant drop in the relapse rate in CD patients given an exceptional formula designed to allow postponed ileal release of n3 fatty acids. A fish oil diet has been shown to increase eicosapentanoic and docosahexanoic acids in the intestinal mucosal lipids of IBD sufferers, also demonstrating a reduction in arachadonic acid. A gain in the synthesis of leukotriene B5 along with a 53 percent decrease of leukotriene B4 was shown in UC patients, whereas the fish oil treatment revealed a nonsignificant trend to faster remission. Fish oil supplementation results in clinical improvement of active mild to moderate disease, but was not associated with a significant reduction in leukotriene B4 production. Consequently, fish oil supplementation of the diet may provide some short-term benefit to people with CD or UC. Using probiotics and prebiotics has received much attention; the interested reader is referred to recent reviews in this area.

 

Clinical Implications

 

It is widely known that nutritional deficiencies are common in people with CD and UC, and people have to be expected, diagnosed and treated. There are no special diets which may be recommended for all patients with IBD; dietary therapy needs to be individualized. TPN or TEN may be necessary to restore nutrient equilibrium in selected IBD patients with malnutrition, but in adults these interventions do not provide an essential decision to modify disease activity. The omega-3 PUFAs in fish oil may reduce disease activity in UC and CD when used at the short term together with regular medical therapy. Their mechanism of action is to enhance the activity of the amino acids PPAR, or peroxisome proliferator-activated receptors, in the intestine, inhibiting the AP-1 signaling pathway and NF-?B, weakening pro-inflammatory cytokine receptor expression. Future research will focus on the identification and use of certain dietary lipids to reduce intestinal inflammatory activity and also to maintain long-term disease remission.

 

Information referenced from the National Center for Biotechnology Information (NCBI) and the National University of Health Sciences. 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

 

WELLNESS TOPIC: EXTRA EXTRA: Managing Workplace Stress

 

 

Blank
References
1.�Liu Y, van Kruiningen HJ, West AB, Cartun RW, Cortot A, Colombel JF. Immunocytochemical evidence of Listeria, Escherichia coli, and Streptococcus antigens in Crohn’s disease.�Gastroenterology.�1995;108:1396�1404.�[PubMed]
2.�Sartor R.�Microbial factors in the pathogenesis of Crohn’s disease, ulcerative colitis and experimental intestinal inflammation.�Baltimore: Williams & Wilkins; 1995.
3.�Wakefield AJ, Ekbom A, Dhillon AP, Pittilo RM, Pounder RE. Crohn’s disease: pathogenesis and persistent measles virus infection.�Gastroenterology.�1995;108:911�916.�[PubMed]
4.�Sartor RB. Current concepts of the etiology and pathogenesis of ulcerative colitis and Crohn’s disease.�Gastroenterol Clin North Am.�1995;24:475�507.�[PubMed]
5.�Sartor RB. Pathogenesis and immune mechanisms of chronic inflammatory bowel diseases.�Am J Gastroenterol.�1997;92:5S�11S.�[PubMed]
6.�MacDermott RP. Alterations in the mucosal immune system in ulcerative colitis and Crohn’s disease.�Med Clin North Am.�1994;78:1207�1231.�[PubMed]
7.�Podolsky DK. Inflammatory bowel disease (1)�N Engl J Med.�1991;325:928�937.�[PubMed]
8.�Podolsky DK. Inflammatory bowel disease (2)�N Engl J Med.�1991;325:1008�1016.�[PubMed]
9.�Yang H, Rotter J.�The genetics of inflammatory disease.�Baltimore: Williams & Wilkins; 1994.
10.�Wurzelmann JI, Lyles CM, Sandler RS. Childhood infections and the risk of inflammatory bowel disease.�Dig Dis Sci.�1994;39:555�560.�[PubMed]
11.�Knoflach P, Park BH, Cunningham R, Weiser MM, Albini B. Serum antibodies to cow’s milk proteins in ulcerative colitis and Crohn’s disease.�Gastroenterology.�1987;92:479�485.�[PubMed]
12.�De Palma GD, Catanzano C. Removable self-expanding metal stents: a pilot study for treatment of achalasia of the esophagus.�Endoscopy.�1998;30:S95�S96.�[PubMed]
13.�Bernstein CN, Ament M, Artinian L, Ridgeway J, Shanahan F. Milk tolerance in adults with ulcerative colitis.�Am J Gastroenterol.�1994;89:872�877.�[PubMed]
14.�Matsui T, Iida M, Fujishima M, Imai K, Yao T. Increased sugar consumption in Japanese patients with Crohn’s disease.�Gastroenterol Jpn.�1990;25:271.�[PubMed]
15.�Kelly DG, Fleming CR. Nutritional considerations in inflammatory bowel diseases.�Gastroenterol Clin North Am.�1995;24:597�611.�[PubMed]
16.�Geerling BJ, Dagnelie PC, Badart-Smook A, Russel MG, Stockbr�gger RW, Brummer RJ. Diet as a risk factor for the development of ulcerative colitis.�Am J Gastroenterol.�2000;95:1008�1013.�[PubMed]
17.�Dudrick SJ, Latifi R, Schrager R. Nutritional management of inflammatory bowel disease.�Surg Clin North Am.�1991;71:609�623.�[PubMed]
18.�D’Odorico A, Bortolan S, Cardin R, D’Inca’ R, Martines D, Ferronato A, Sturniolo GC. Reduced plasma antioxidant concentrations and increased oxidative DNA damage in inflammatory bowel disease.�Scand J Gastroenterol.�2001;36:1289�1294.�[PubMed]
19.�Reimund JM, Hirth C, Koehl C, Baumann R, Duclos B. Antioxidant and immune status in active Crohn’s disease. A possible relationship.�Clin Nutr.�2000;19:43�48.�[PubMed]
20.�Romagnuolo J, Fedorak RN, Dias VC, Bamforth F, Teltscher M. Hyperhomocysteinemia and inflammatory bowel disease: prevalence and predictors in a cross-sectional study.�Am J Gastroenterol.�2001;96:2143�2149.�[PubMed]
21.�Lewis JD, Fisher RL. Nutrition support in inflammatory bowel disease.�Med Clin North Am.�1994;78:1443�1456.�[PubMed]
22.�Azcue M, Rashid M, Griffiths A, Pencharz PB. Energy expenditure and body composition in children with Crohn’s disease: effect of enteral nutrition and treatment with prednisolone.�Gut.�1997;41:203�208.[PMC free article][PubMed]
23.�Mingrone G, Capristo E, Greco AV, Benedetti G, De Gaetano A, Tataranni PA, Gasbarrini G. Elevated diet-induced thermogenesis and lipid oxidation rate in Crohn disease.�Am J Clin Nutr.�1999;69:325�330.[PubMed]
24.�Bjarnason I, Macpherson A, Mackintosh C, Buxton-Thomas M, Forgacs I, Moniz C. Reduced bone density in patients with inflammatory bowel disease.�Gut.�1997;40:228�233.�[PMC free article][PubMed]
25.�Griffiths AM, Nguyen P, Smith C, MacMillan JH, Sherman PM. Growth and clinical course of children with Crohn’s disease.�Gut.�1993;34:939�943.�[PMC free article][PubMed]
26.�Fischer JE, Foster GS, Abel RM, Abbott WM, Ryan JA. Hyperalimentation as primary therapy for inflammatory bowel disease.�Am J Surg.�1973;125:165�175.�[PubMed]
27.�Reilly J, Ryan JA, Strole W, Fischer JE. Hyperalimentation in inflammatory bowel disease.�Am J Surg.�1976;131:192�200.�[PubMed]
28.�Ganem D, Schneider RJ. Hepadnaviridae: The viruses and their replication. In: Knipe DM, Howley PM, editors.�Fields Virology. Volume 2.�Philadelphia: Lippincott, Williams & Wilkins; 2001. pp. 2923�2969.
29.�Jones VA, Dickinson RJ, Workman E, Wilson AJ, Freeman AH, Hunter JO. Crohn’s disease: maintenance of remission by diet.�Lancet.�1985;2:177�180.�[PubMed]
30.�Suzuki I, Kiyono H, Kitamura K, Green DR, McGhee JR. Abrogation of oral tolerance by contrasuppressor T cells suggests the presence of regulatory T-cell networks in the mucosal immune system.�Nature.�1986;320:451�454.�[PubMed]
31.�Ostro MJ, Greenberg GR, Jeejeebhoy KN. Total parenteral nutrition and complete bowel rest in the management of Crohn’s disease.�JPEN J Parenter Enteral Nutr.�1985;9:280�287.�[PubMed]
32.�Matuchansky C. Parenteral nutrition in inflammatory bowel disease.�Gut.�1986;27 Suppl 1:81�84.[PMC free article][PubMed]
33.�Payne-James JJ, Silk DB. Total parenteral nutrition as primary treatment in Crohn’s disease–RIP?�Gut.�1988;29:1304�1308.�[PMC free article][PubMed]
34.�Shiloni E, Coronado E, Freund HR. Role of total parenteral nutrition in the treatment of Crohn’s disease.�Am J Surg.�1989;157:180�185.�[PubMed]
35.�Dickinson RJ, Ashton MG, Axon AT, Smith RC, Yeung CK, Hill GL. Controlled trial of intravenous hyperalimentation and total bowel rest as an adjunct to the routine therapy of acute colitis.�Gastroenterology.�1980;79:1199�1204.�[PubMed]
36.�McIntyre PB, Powell-Tuck J, Wood SR, Lennard-Jones JE, Lerebours E, Hecketsweiler P, Galmiche JP, Colin R. Controlled trial of bowel rest in the treatment of severe acute colitis.�Gut.�1986;27:481�485.[PMC free article][PubMed]
37.�Greenberg GR, Fleming CR, Jeejeebhoy KN, Rosenberg IH, Sales D, Tremaine WJ. Controlled trial of bowel rest and nutritional support in the management of Crohn’s disease.�Gut.�1988;29:1309�1315.[PMC free article][PubMed]
38.�Hughes CA, Bates T, Dowling RH. Cholecystokinin and secretin prevent the intestinal mucosal hypoplasia of total parenteral nutrition in the dog.�Gastroenterology.�1978;75:34�41.�[PubMed]
39.�Stratton RJ, Smith TR. Role of enteral and parenteral nutrition in the patient with gastrointestinal and liver disease.�Best Pract Res Clin Gastroenterol.�2006;20:441�466.�[PubMed]
40.�O’Sullivan M, O’Morain C. Nutrition in inflammatory bowel disease.�Best Pract Res Clin Gastroenterol.�2006;20:561�573.�[PubMed]
41.�Gonz�lez-Huix F, Fern�ndez-Ba�ares F, Esteve-Comas M, Abad-Lacruz A, Cabr� E, Acero D, Figa M, Guilera M, Humbert P, de Le�n R. Enteral versus parenteral nutrition as adjunct therapy in acute ulcerative colitis.�Am J Gastroenterol.�1993;88:227�232.�[PubMed]
42.�Voitk AJ, Echave V, Feller JH, Brown RA, Gurd FN. Experience with elemental diet in the treatment of inflammatory bowel disease. Is this primary therapy?�Arch Surg.�1973;107:329�333.�[PubMed]
43.�Axelsson C, Jarnum S. Assessment of the therapeutic value of an elemental diet in chronic inflammatory bowel disease.�Scand J Gastroenterol.�1977;12:89�95.�[PubMed]
44.�Lochs H, Steinhardt HJ, Klaus-Wentz B, Zeitz M, Vogelsang H, Sommer H, Fleig WE, Bauer P, Schirrmeister J, Malchow H. Comparison of enteral nutrition and drug treatment in active Crohn’s disease. Results of the European Cooperative Crohn’s Disease Study. IV.�Gastroenterology.�1991;101:881�888.[PubMed]
45.�Malchow H, Steinhardt HJ, Lorenz-Meyer H, Strohm WD, Rasmussen S, Sommer H, Jarnum S, Brandes JW, Leonhardt H, Ewe K. Feasibility and effectiveness of a defined-formula diet regimen in treating active Crohn’s disease. European Cooperative Crohn’s Disease Study III.�Scand J Gastroenterol.�1990;25:235�244.�[PubMed]
46.�O’Brien CJ, Giaffer MH, Cann PA, Holdsworth CD. Elemental diet in steroid-dependent and steroid-refractory Crohn’s disease.�Am J Gastroenterol.�1991;86:1614�1618.�[PubMed]
47.�Okada M, Yao T, Yamamoto T, Takenaka K, Imamura K, Maeda K, Fujita K. Controlled trial comparing an elemental diet with prednisolone in the treatment of active Crohn’s disease.�Hepatogastroenterology.�1990;37:72�80.�[PubMed]
48.�O’Mor�in C, Segal AW, Levi AJ. Elemental diet as primary treatment of acute Crohn’s disease: a controlled trial.�Br Med J (Clin Res Ed)�1984;288:1859�1862.�[PMC free article][PubMed]
49.�Raouf AH, Hildrey V, Daniel J, Walker RJ, Krasner N, Elias E, Rhodes JM. Enteral feeding as sole treatment for Crohn’s disease: controlled trial of whole protein v amino acid based feed and a case study of dietary challenge.�Gut.�1991;32:702�707.�[PMC free article][PubMed]
50.�Rocchio MA, Cha CJ, Haas KF, Randall HT. Use of chemically defined diets in the management of patients with acute inflammatory bowel disease.�Am J Surg.�1974;127:469�475.�[PubMed]
51.�Saverymuttu S, Hodgson HJ, Chadwick VS. Controlled trial comparing prednisolone with an elemental diet plus non-absorbable antibiotics in active Crohn’s disease.�Gut.�1985;26:994�998.�[PMC free article][PubMed]
52.�Teahon K, Bjarnason I, Pearson M, Levi AJ. Ten years’ experience with an elemental diet in the management of Crohn’s disease.�Gut.�1990;31:1133�1137.�[PMC free article][PubMed]
53.�Teahon K, Smethurst P, Pearson M, Levi AJ, Bjarnason I. The effect of elemental diet on intestinal permeability and inflammation in Crohn’s disease.�Gastroenterology.�1991;101:84�89.�[PubMed]
54.�Heuschkel RB, Menache CC, Megerian JT, Baird AE. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children.�J Pediatr Gastroenterol Nutr.�2000;31:8�15.�[PubMed]
55.�Sanderson IR, Boulton P, Menzies I, Walker-Smith JA. Improvement of abnormal lactulose/rhamnose permeability in active Crohn’s disease of the small bowel by an elemental diet.�Gut.�1987;28:1073�1076.[PMC free article][PubMed]
56.�Sanderson IR, Udeen S, Davies PS, Savage MO, Walker-Smith JA. Remission induced by an elemental diet in small bowel Crohn’s disease.�Arch Dis Child.�1987;62:123�127.�[PMC free article][PubMed]
57.�Ruemmele FM, Roy CC, Levy E, Seidman EG. Nutrition as primary therapy in pediatric Crohn’s disease: fact or fantasy?�J Pediatr.�2000;136:285�291.�[PubMed]
58.�O’Morain C, O’Sullivan M. Nutritional support in Crohn’s disease: current status and future directions.�J Gastroenterol.�1995;30 Suppl 8:102�107.�[PubMed]
59.�Rigaud D, Cosnes J, Le Quintrec Y, Ren� E, Gendre JP, Mignon M. Controlled trial comparing two types of enteral nutrition in treatment of active Crohn’s disease: elemental versus polymeric diet.�Gut.�1991;32:1492�1497.�[PMC free article][PubMed]
60.�Royall D, Wolever TM, Jeejeebhoy KN. Evidence for colonic conservation of malabsorbed carbohydrate in short bowel syndrome.�Am J Gastroenterol.�1992;87:751�756.�[PubMed]
61.�Giaffer MH, North G, Holdsworth CD. Controlled trial of polymeric versus elemental diet in treatment of active Crohn’s disease.�Lancet.�1990;335:816�819.�[PubMed]
62.�Verma S, Kirkwood B, Brown S, Giaffer MH. Oral nutritional supplementation is effective in the maintenance of remission in Crohn’s disease.�Dig Liver Dis.�2000;32:769�774.�[PubMed]
63.�Levine GM, Deren JJ, Steiger E, Zinno R. Role of oral intake in maintenance of gut mass and disaccharide activity.�Gastroenterology.�1974;67:975�982.�[PubMed]
64.�Weser E, Heller R, Tawil T. Stimulation of mucosal growth in the rat ileum by bile and pancreatic secretions after jejunal resection.�Gastroenterology.�1977;73:524�529.�[PubMed]
65.�Fell JM, Paintin M, Arnaud-Battandier F, Beattie RM, Hollis A, Kitching P, Donnet-Hughes A, MacDonald TT, Walker-Smith JA. Mucosal healing and a fall in mucosal pro-inflammatory cytokine mRNA induced by a specific oral polymeric diet in paediatric Crohn’s disease.�Aliment Pharmacol Ther.�2000;14:281�289.�[PubMed]
66.�Souba WW, Smith RJ, Wilmore DW. Glutamine metabolism by the intestinal tract.�JPEN J Parenter Enteral Nutr.�1985;9:608�617.�[PubMed]
67.�Windmueller HG, Spaeth AE. Uptake and metabolism of plasma glutamine by the small intestine.�J Biol Chem.�1974;249:5070�5079.�[PubMed]
68.�Higashiguchi T, Hasselgren PO, Wagner K, Fischer JE. Effect of glutamine on protein synthesis in isolated intestinal epithelial cells.�JPEN J Parenter Enteral Nutr.�1993;17:307�314.�[PubMed]
69.�Burke DJ, Alverdy JC, Aoys E, Moss GS. Glutamine-supplemented total parenteral nutrition improves gut immune function.�Arch Surg.�1989;124:1396�1399.�[PubMed]
70.�Souba WW, Herskowitz K, Klimberg VS, Salloum RM, Plumley DA, Flynn TC, Copeland EM. The effects of sepsis and endotoxemia on gut glutamine metabolism.�Ann Surg.�1990;211:543�549; discussion 543-551;.�[PMC free article][PubMed]
71.�Den Hond E, Hiele M, Peeters M, Ghoos Y, Rutgeerts P. Effect of long-term oral glutamine supplements on small intestinal permeability in patients with Crohn’s disease.�JPEN J Parenter Enteral Nutr.�1999;23:7�11.�[PubMed]
72.�Akobeng AK, Miller V, Stanton J, Elbadri AM, Thomas AG. Double-blind randomized controlled trial of glutamine-enriched polymeric diet in the treatment of active Crohn’s disease.�J Pediatr Gastroenterol Nutr.�2000;30:78�84.�[PubMed]
73.�Jacobs LR, Lupton JR. Effect of dietary fibers on rat large bowel mucosal growth and cell proliferation.�Am J Physiol.�1984;246:G378�G385.�[PubMed]
74.�Spaeth G, Berg RD, Specian RD, Deitch EA. Food without fiber promotes bacterial translocation from the gut.�Surgery.�1990;108:240�246; discussion 246-247;.�[PubMed]
75.�Roediger WE, Moore A. Effect of short-chaim fatty acid on sodium absorption in isolated human colon perfused through the vascular bed.�Dig Dis Sci.�1981;26:100�106.�[PubMed]
76.�Sakata T. Stimulatory effect of short-chain fatty acids on epithelial cell proliferation in the rat intestine: a possible explanation for trophic effects of fermentable fibre, gut microbes and luminal trophic factors.�Br J Nutr.�1987;58:95�103.�[PubMed]
77.�Roediger WE. The colonic epithelium in ulcerative colitis: an energy-deficiency disease?�Lancet.�1980;2:712�715.�[PubMed]
78.�Chapman MA, Grahn MF, Boyle MA, Hutton M, Rogers J, Williams NS. Butyrate oxidation is impaired in the colonic mucosa of sufferers of quiescent ulcerative colitis.�Gut.�1994;35:73�76.[PMC free article][PubMed]
79.�Den Hond E, Hiele M, Evenepoel P, Peeters M, Ghoos Y, Rutgeerts P. In vivo butyrate metabolism and colonic permeability in extensive ulcerative colitis.�Gastroenterology.�1998;115:584�590.�[PubMed]
80.�Simpson EJ, Chapman MA, Dawson J, Berry D, Macdonald IA, Cole A. In vivo measurement of colonic butyrate metabolism in patients with quiescent ulcerative colitis.�Gut.�2000;46:73�77.[PMC free article][PubMed]
81.�Tappenden KA, Thomson AB, Wild GE, McBurney MI. Short-chain fatty acid-supplemented total parenteral nutrition enhances functional adaptation to intestinal resection in rats.�Gastroenterology.�1997;112:792�802.�[PubMed]
82.�Senagore AJ, MacKeigan JM, Scheider M, Ebrom JS. Short-chain fatty acid enemas: a cost-effective alternative in the treatment of nonspecific proctosigmoiditis.�Dis Colon Rectum.�1992;35:923�927.[PubMed]
83.�Segain JP, Raingeard de la Bl�ti�re D, Bourreille A, Leray V, Gervois N, Rosales C, Ferrier L, Bonnet C, Blotti�re HM, Galmiche JP. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease.�Gut.�2000;47:397�403.�[PMC free article][PubMed]
84.�Aslan A, Triadafilopoulos G. Fish oil fatty acid supplementation in active ulcerative colitis: a double-blind, placebo-controlled, crossover study.�Am J Gastroenterol.�1992;87:432�437.�[PubMed]
85.�Shoda R, Matsueda K, Yamato S, Umeda N. Epidemiologic analysis of Crohn disease in Japan: increased dietary intake of n-6 polyunsaturated fatty acids and animal protein relates to the increased incidence of Crohn disease in Japan.�Am J Clin Nutr.�1996;63:741�745.�[PubMed]
86.�Vilaseca J, Salas A, Guarner F, Rodr�guez R, Mart�nez M, Malagelada JR. Dietary fish oil reduces progression of chronic inflammatory lesions in a rat model of granulomatous colitis.�Gut.�1990;31:539�544.�[PMC free article][PubMed]
87.�Campos FG, Waitzberg DL, Habr-Gama A, Logullo AF, Noronha IL, Jancar S, Torrinhas RS, F�rst P. Impact of parenteral n-3 fatty acids on experimental acute colitis.�Br J Nutr.�2002;87 Suppl 1:S83�S88.[PubMed]
88.�Loeschke K, Ueberschaer B, Pietsch A, Gruber E, Ewe K, Wiebecke B, Heldwein W, Lorenz R. n-3 fatty acids only delay early relapse of ulcerative colitis in remission.�Dig Dis Sci.�1996;41:2087�2094.[PubMed]
89.�Belluzzi A, Brignola C, Campieri M, Pera A, Boschi S, Miglioli M. Effect of an enteric-coated fish-oil preparation on relapses in Crohn’s disease.�N Engl J Med.�1996;334:1557�1560.�[PubMed]
90.�Hawthorne AB, Daneshmend TK, Hawkey CJ, Belluzzi A, Everitt SJ, Holmes GK, Malkinson C, Shaheen MZ, Willars JE. Treatment of ulcerative colitis with fish oil supplementation: a prospective 12 month randomised controlled trial.�Gut.�1992;33:922�928.�[PMC free article][PubMed]
91.�Hillier K, Jewell R, Dorrell L, Smith CL. Incorporation of fatty acids from fish oil and olive oil into colonic mucosal lipids and effects upon eicosanoid synthesis in inflammatory bowel disease.�Gut.�1991;32:1151�1155.�[PMC free article][PubMed]
92.�Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma)�J Biol Chem.�1995;270:12953�12956.�[PubMed]
93.�Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs.�J Biol Chem.�1997;272:3406�3410.�[PubMed]
94.�Delerive P, Furman C, Teissier E, Fruchart J, Duriez P, Staels B. Oxidized phospholipids activate PPARalpha in a phospholipase A2-dependent manner.�FEBS Lett.�2000;471:34�38.�[PubMed]
95.�Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma.�Proc Natl Acad Sci USA.�1997;94:4318�4323.�[PMC free article][PubMed]
96.�Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta.�Proc Natl Acad Sci USA.�1997;94:4312�4317.�[PMC free article][PubMed]
97.�Mans�n A, Guardiola-Diaz H, Rafter J, Branting C, Gustafsson JA. Expression of the peroxisome proliferator-activated receptor (PPAR) in the mouse colonic mucosa.�Biochem Biophys Res Commun.�1996;222:844�851.�[PubMed]
98.�Desreumaux P, Ernst O, Geboes K, Gambiez L, Berrebi D, M�ller-Alouf H, Hafraoui S, Emilie D, Ectors N, Peuchmaur M, et al. Inflammatory alterations in mesenteric adipose tissue in Crohn’s disease.�Gastroenterology.�1999;117:73�81.�[PubMed]
99.�Su CG, Wen X, Bailey ST, Jiang W, Rangwala SM, Keilbaugh SA, Flanigan A, Murthy S, Lazar MA, Wu GD. A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response.�J Clin Invest.�1999;104:383�389.�[PMC free article][PubMed]
100.�Ricote M, Huang J, Fajas L, Li A, Welch J, Najib J, Witztum JL, Auwerx J, Palinski W, Glass CK. Expression of the peroxisome proliferator-activated receptor gamma (PPARgamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein.�Proc Natl Acad Sci USA.�1998;95:7614�7619.�[PMC free article][PubMed]
101.�Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, et al. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators.�Nature.�1998;393:790�793.�[PubMed]
102.�Marx N, Bourcier T, Sukhova GK, Libby P, Plutzky J. PPARgamma activation in human endothelial cells increases plasminogen activator inhibitor type-1 expression: PPARgamma as a potential mediator in vascular disease.�Arterioscler Thromb Vasc Biol.�1999;19:546�551.�[PubMed]
103.�Delerive P, Martin-Nizard F, Chinetti G, Trottein F, Fruchart JC, Najib J, Duriez P, Staels B. Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway.�Circ Res.�1999;85:394�402.�[PubMed]
104.�Sakai M, Matsushima-Hibiya Y, Nishizawa M, Nishi S. Suppression of rat glutathione transferase P expression by peroxisome proliferators: interaction between Jun and peroxisome proliferator-activated receptor alpha.�Cancer Res.�1995;55:5370�5376.�[PubMed]
105.�Zhou YC, Waxman DJ. STAT5b down-regulates peroxisome proliferator-activated receptor alpha transcription by inhibition of ligand-independent activation function region-1 trans-activation domain.�J Biol Chem.�1999;274:29874�29882.�[PubMed]
106.�Desreumaux P, Dubuquoy L, Nutten S, Peuchmaur M, Englaro W, Schoonjans K, Derijard B, Desvergne B, Wahli W, Chambon P, et al. Attenuation of colon inflammation through activators of the retinoid X receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma) heterodimer. A basis for new therapeutic strategies.�J Exp Med.�2001;193:827�838.�[PMC free article][PubMed]
107.�Lewis JD, Lichtenstein GR, Stein RB, Deren JJ, Judge TA, Fogt F, Furth EE, Demissie EJ, Hurd LB, Su CG, et al. An open-label trial of the PPAR-gamma ligand rosiglitazone for active ulcerative colitis.�Am J Gastroenterol.�2001;96:3323�3328.�[PubMed]
108.�G�ttlicher M, Widmark E, Li Q, Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor.�Proc Natl Acad Sci USA.�1992;89:4653�4657.[PMC free article][PubMed]
Close Accordion
Assessment and Treatment of Piriformis

Assessment and Treatment of Piriformis

These assessment and treatment recommendations represent a synthesis of information derived from personal clinical experience and from the numerous sources which are cited, or are based on the work of researchers, clinicians and therapists who are named (Basmajian 1974, Cailliet 1962, Dvorak & Dvorak 1984, Fryette 1954, Greenman 1989, 1996, Janda 1983, Lewit 1992, 1999, Mennell 1964, Rolf 1977, Williams 1965).

 

Clinical Application of Neuromuscular Techniques: Piriformis

 

Assessment of Shortened Piriformis

 

Test (a) Stretch test. When short, piriformis will cause the affected side leg of the supine patient to appear to be short and externally rotated. With the patient supine, the tested leg is placed into flexion at the hip and knee so that the foot rests on the table lateral to the contralateral knee (the tested leg is crossed over the straight non-tested leg, in other words as shown in Fig. 4.17). The angle of hip flexion should not exceed 60� (see notes on piriformis in Box 4.6).

 

Figure 4 17 MET Treatment of Piriformis Muscle with Patient Supine Image 1

 

Figure 4.17 MET treatment of piriformis muscle with patient supine. The pelvis must be maintained in a stable position as the knee (right in this example) is adducted to stretch piriformis following an isometric contraction.

 

The non-tested side ASIS is stabilised to prevent pelvic motion during the test and the knee of the tested side is pushed into adduction to place a stretch on piriformis. If there is a short piriformis the degree of adduction will be limited and the patient will report discomfort behind the trochanter.

 

Test (b) Palpation test (Fig. 4.18) The patient is side-lying, tested side uppermost. The practitioner stands at the level of the pelvis in front of and facing the patient, and, in order to contact the insertion of piriformis, draws imaginary lines between:

 

  • ASIS and ischial tuberosity, and
  • PSIS and the most prominent point of trochanter.

 

Where these reference lines cross, just posterior to the trochanter, is the insertion of the muscle, and pressure here will produce marked discomfort if the structure is short or irritated.

 

Figure 4 18 Bony Landmarks Used as Coordinates in Piriformis Image 2

 

Figure 4.18 Using bony landmarks as coordinates the commonest tender areas are located in piriformis, in the belly and at the attachment of the muscle.

 

If the most common trigger point site in the belly of the muscle is sought, then the line from the ASIS should be taken to the tip of the coccyx rather than to the ischial tuberosity. Pressure where this line crosses the other will access the mid-point of the belly of piriformis where triggers are common. Light compression here which produces a painful response is indicative of a stressed muscle and possibly an active myofascial trigger point.

 

Piriformis Strength Test

 

The patient lies prone, both knees flexed to 90�, with practitioner at foot of table grasping lower legs at the limit of their separation (which internally rotates the hip and therefore allows comparison of range of movement permitted by shortened external rotators such as the piriformis).

 

The patient attempts to bring the ankles together as the practitioner assesses the relative strength of the two legs. Mitchell et al (1979) suggest that if there is relative shortness (as evidenced by the lower leg not being able to travel as far from the mid-line as its pair in this position), and if that same side also tests strong, then MET is called for. If there is shortness but also weakness then the reasons for the weakness need to be dealt with prior to stretching using MET.

 

Box 4.6 Notes on Piriformis

 

  • Piriformis paradox. The performance of external rotation of the hip by piriformis occurs when the angle of hip flexion is 60� or less. Once the angle of hip flexion is greater than 60� piriformis function changes, so that it becomes an internal rotator of the hip (Gluck & Liebenson 1997, Lehmkuhl & Smith 1983). The implications of this are illustrated in Figures 4.17 and 4.19.
  • This postural muscle, like all others which have a predominence of type l fibres, will shorten if stressed. In the case of piriformis, the effect of shortening is to increase its diameter and because of its location this allows for direct pressure to be exerted on the sciatic nerve, which passes under it in 80% of people. In the other 20% the nerve passes through the muscle so that contraction will produce veritable strangulation of the sciatic nerve.
  • In addition, the pudendal nerve and the blood vessels of the internal iliac artery, as well as common perineal nerves, posterior femoral cutaneous nerve and nerves of the hip rotators, can all be affected.
  • If there is sciatic pain associated with piriformis shortness, then on straight leg raising, which reproduces the pain, external rotation of the hip should relieve it, since this slackens piriformis. (This clue may, however, only apply to any degree if the individual is one of those in whom the nerve actually passes through the muscle.)
  • The effects can be circulatory, neurological and functional, inducing pain and paraesthesia of the affected limb as well as alterations to pelvic and lumbar function. Diagnosis usually hinges on the absence of spinal causative factors and the distributions of symptoms from the sacrum to the hip joint, over the gluteal region and down to the popliteal space. Palpation of the affected piriformis tendon, near the head of the trochanter, will elicit pain and the affected leg will probably be externally rotated.
  • The piriformis muscle syndrome is frequently characterised by such bizarre symptoms that they may seem unrelated. One characteristic complaint is a persistent, severe, radiating low back pain extending from the sacrum to the hip joint, over the gluteal region and the posterior portion of the upper leg, to the popliteal space. In the most severe cases the patient will be unable to lie or stand comfortably, and changes in position will not relieve the pain. Intense pain will occur when the patient sits or squats since this type of movement requires external rotation of the upper leg and flexion at the knee.
  • Compression of the pudendal nerve and blood vessels which pass through the greater sciatic foramen and re-enter the pelvis via the lesser sciatic foramen is possible because of piriformis contracture. Any compression would result in impaired circulation to the genitalia in both sexes. Since external rotation of the hips is required for coitus by women, pain noted during this act could relate to impaired circulation induced by piriformis dysfunction. This could also be a basis for impotency in men. (See also Box 4.7.)
  • Piriformis involvement often relates to a pattern of pain which includes: pain near the trochanter; pain in the inguinal area; local tenderness over the insertion behind trochanter; SI joint pain on the opposite side; externally rotated foot on the same side; pain unrelieved by most positions with standing and walking being the easiest; limitation of internal rotation of the leg which produces pain near the hip; and a short leg on the affected side.
  • The pain itself will be persistent and radiating, covering anywhere from the sacrum to the buttock, hip and leg including inguinal and perineal areas.
  • Bourdillon (1982) suggests that piriformis syndrome and SI joint dysfunction are intimately connected and that recurrent SI problems will not stabilise until hypertonic piriformis is corrected.
  • Janda (1996) points to the vast amount of pelvic organ dysfunction to which piriformis can contribute due to its relationship with circulation to the area.
  • Mitchell et al (1979) suggest that (as in psoas example above) piriformis shortness should only be treated if it is tested to be short and stronger than its pair. If it is short and weak (see p. 110 for strength test), then whatever is hypertonic and influencing it should be released and stretched first (Mitchell et al 1979). When it tests strong and short, piriformis should receive MET treatment.
  • Since piriformis is an external rotator of the hip it can be inhibited (made to test weak) if an internal rotator such as TFL is hypertonic or if its pair is hypertonic, since one piriformis will inhibit the other.

 

Box 4.7 Notes on Working and Resting Muscles

 

  • Richard (1978) reminds us that a working muscle will mobilise up to 10 times the quantity of blood mobilised by a resting muscle. He points out the link between pelvic circulation and lumbar, ischiatic and gluteal arteries and the chance this allows to engineer the involvement of 2400 square metres of capillaries by using repetitive pumping of these muscles (including piriformis).
  • The therapeutic use of this knowledge involves the patient being asked to repetitively contract both piriformis muscles against resistance. The patient is supine, knees bent, feet on the table; the practitioner resists their effort to abduct their flexed knees, using pulsed muscle energy approach (Ruddy�s method) in which two isometrically resisted pulsation/contractions per second are introduced for as long as possible (a minute seems a long time doing this).

 

Figure 4 19 MET Treatment of Piriformis with Hip Fully Flexed & Externally Rotated Image 3

 

Figure 4.19 MET treatment of piriformis with hip fully flexed and externally rotated (see Box 4.6, first bullet point).

 

Figure 4 20 A Combined Ischaemic Compression & MET Side Lying Treatment of Piriformis Image 4

 

Figure 4.20 A combined ischaemic compression (elbow pressure) and MET side-lying treatment of piriformis. The pressure is alternated with isometric contractions/stretching of the muscle until no further gain is achieved.

 

MET Treatment of Piriformis

 

Piriformis method (a) Side-lying The patient is side-lying, close to the edge of the table, affected side uppermost, both legs flexed at hip and knee. The practitioner stands facing the patient at hip level.

 

The practitioner places his cephalad elbow tip gently over the point behind trochanter, where piriformis inserts. The patient should be close enough to the edge of the table for the practitioner to stabilise the pelvis against his trunk (Fig. 4.20). At the same time, the practitioner�s caudad hand grasps the ankle and uses this to bring the upper leg/hip into internal rotation, taking out all the slack in piriformis.

 

A degree of inhibitory pressure (sufficient to cause discomfort but not pain) is applied via the elbow for 5�7 seconds while the muscle is kept at a reasonable but not excessive degree of stretch. The practitioner maintains contact on the point, but eases pressure, and asks the patient to introduce an isometric contraction (25% of strength for 5�7 seconds) to piriformis by bringing the lower leg towards the table against resistance. (The same acute and chronic rules as discussed previously are employed, together with cooperative breathing if appropriate, see Box 4.2.)

 

After the contraction ceases and the patient relaxes, the lower limb is taken to its new resistance barrier and elbow pressure is reapplied. This process is repeated until no further gain is achieved.

 

Piriformis method (b)1 This method is a variation on the method advocated by TePoorten (1960) which calls for longer and heavier compression, and no intermediate isometric contractions.

 

In the first stage of TePoorten�s method the patient lies on the non-affected side with knees flexed and hip joints flexed to 90�.The practitioner places his elbow on the piriformis musculotendinous junction and a steady pressure of 20�30 lb (9�13 kg) is applied. With his other hand he abducts the foot so that it will force an internal rotation of the upper leg.

 

The leg is held in this rotated position for periods of up to 2 minutes. This procedure is repeated two or three times. The patient is then placed in the supine position and the affected leg is tested for freedom of both external and internal rotation.

 

Piriformis method (b)2 The second stage of TePoorten�s treatment is performed with the patient supine with both legs extended. The foot of the affected leg is grasped and the leg is flexed at both the knee and the hip. As knee and hip flexion is performed the practitioner turns the foot inward, so inducing an external rotation of the upper leg. The practitioner then extends the knee, and simultaneously turns the foot outward, resulting in an internal rotation of the upper leg.

 

During these procedures the patient is instructed to partially resist the movements introduced by the practitioner (i.e. the procedure becomes an isokinetic activity). This treatment method, repeated two or three times, serves to relieve the contracture of the muscles of external and internal hip rotation.

 

Piriformis method (c) A series of MET isometric contractions and stretches can be applied with the patient prone and the affected side knee flexed. The hip is rotated internally by the practitioner using the foot as a lever to ease it laterally, so putting piriformis at stretch. Acute and chronic guidelines described earlier are used to determine the appropriate starting point for the contraction (at the barrier for acute and short of it for chronic).

 

The patient attempts to lightly bring the heel back towards the midline against resistance (avoiding strong contractions to avoid knee strain in this position) and this is held for 7�10 seconds. After release of the contraction the hip is rotated further to move piriformis to or through the barrier, as appropriate. Application of inhibitory pressure to the attachment or belly of piriformis is possible via thumb, if deemed necessary.

 

Piriformis method (d) A general approach which balances muscles of the region, as well as the pelvic diaphragm, is achieved by having the patient squat while the practitioner stands and stabilises both shoulders, preventing the patient from rising as this is attempted, while the breath is held. After 7�10 seconds the effort is released; a deeper squat is performed, and the procedure is repeated several times.

 

Piriformis method (e) This method is based on the test position (see Fig. 4.17) and is described by Lewit (1992). With the patient supine, the treated leg is placed into flexion at the hip and knee, so that the foot rests on the table lateral to the contralateral knee (the leg on the side to be treated is crossed over the other, straight, leg). The angle of hip flexion should not exceed 60� (see notes on piriformis, Box 4.6, for explanation).

 

The practitioner places one hand on the contralateral ASIS to prevent pelvic motion, while the other hand is placed against the lateral flexed knee as this is pushed into resisted abduction to contract piriformis for 7�10 seconds. Following the contraction the practitioner eases the treated side leg into adduction until a sense of resistance is noted; this is held for 10�30 seconds.

 

Piriformis method (f) Since contraction of one piriformis inhibits its pair, it is possible to self-treat an affected short piriformis by having the patient lie up against a wall with the non-affected side touching it, both knees flexed (modified from Retzlaff 1974). The patient monitors the affected side by palpating behind the trochanter, ensuring that no contraction takes place on that side.

 

After a contraction lasting 10 seconds or so of the non-affected side (the patient presses the knee against the wall), the patient moves away from the wall and the position described for piriformis test (see Fig. 4.17) above is adopted, and the patient pushes the affected side knee into adduction, stretching piriformis on that side. This is repeated several times.

 

Dr. Alex Jimenez offers an additional assessment and treatment of the hip flexors as a part of a referenced clinical application of neuromuscular techniques by Leon Chaitow and Judith Walker DeLany. 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

 

WELLNESS TOPIC: EXTRA EXTRA: Managing Workplace Stress

 

 

Psoriasis: Conventional And Alternative Treatment

Psoriasis: Conventional And Alternative Treatment

Psoriasis Abstract

Psoriasis is a common T-cell-mediated immune disorder characterized by circumscribed, red, thickened plaques with an overlying silver-white scale. It occurs worldwide, although the incidence is lower in warmer, sunnier climates. The primary cause of psoriasis is unknown. During an active disease state, an underlying inflammatory mechanism is frequently involved. Many conventional treatments focus on suppressing symptoms associated with psoriasis and have significant side effects. This article reviews several of the researched natural approaches to psoriasis treatment, while addressing its underlying cause. (Altern Med Rev 2007;12(4):319-330)

Introduction

Recent genetic and immunological advances have greatly increased understanding of the pathogenesis of psoriasis as a chronic, immune-mediated inflammatory disorder. The primary immune defect in psoriasis appears to be an increase in cell signaling via chemokines and cytokines that act on upregulated gene expression and cause hyper-proliferation of keratinocytes. A new understanding of this complex disease has catalyzed the development of targeted biological treatments. These revolutionary therapies are not without potential risk, however. A review of alternative natural therapies provides some options for increasing safety and efficacy in the management of psoriasis. Psoriasis � Pathophysiology, Conventional, and Alternative Approaches to Treatment Michael Traub, ND, and Keri Marshall MS, ND

Epidemiology

The prevalence of psoriasis varies widely depending on ethnicity. Psoriasis occurs most commonly in Caucasians, with an estimated occurrence of 60 cases per 100,000/year in this population. Its prevalence in the United States is 2-4 percent, although it is rare or absent in Native American and certain African-American populations. While common in Japan, it is much less common in China, with an estimated incidence of 0.3 percent. The prevalence in the general population of Northern Europe and Scandinavia is 1.5-3 percent. Women and men are equally affected by this condition. The observation that latitude affects prevalence is most likely related to the beneficial effect of sunlight on the disease.1 Although psoriasis can occur at any age, the mean age of onset for chronic plaque psoriasis is estimated at 33 years, with 75 percent of cases initiated before age 46.2 The age of onset appears to be slightly earlier in women than men. Longitudinal studies suggest spontaneous remission may occur in about one-third of patients with psoriasis.3

Pathophysiology

Until recently psoriasis was considered a disorder of epidermal keratinocytes; however, it is now recognized primarily as an immune-mediated disorder. In order to properly understand the immune dysfunction present in psoriasis, it is imperative to understand the normal immune response of skin. Skin is a primary lymphoid organ with an effective immunological surveillance system equipped with antigen presenting cells, cytokine synthesizing keratinocytes, epidermotropic T cells, dermal capillary endothelial cells, draining nodes, mast cells, tissue macrophages, granulocytes, fibroblasts, and non-Langerhans cells. Skin also has lymph nodes and circulating T lymphocytes. Together these cells communicate by means of cytokine secretion and respond accordingly via stimulation by bacteria, chemical, ultraviolet (UV) light, and other irritating factors. The primary cytokine released in response to antigen presentation is tumor necrosis factor-alpha (TNF-?). Generally, this is a controlled process unless the insult to the skin is prolonged, in which case imbalanced cytokine production leads to a pathological state such as psoriasis.

Debate continues whether psoriasis is an autoimmune disorder or a T-helper 1 (Th1) immune dysfunction. T-cell activation, TNF-?, and dendritic cells are pathogenic factors stimulated in response to a triggering factor, such as a physical injury, inflammation, bacteria, virus, or withdrawal of corticosteroid medication. Initially, immature dendritic cells in the epidermis stimulate T-cells from lymph nodes in response to as yet unidentified antigen stimulation. The lymphocytic infiltrate in psoriasis is predominately CD4 and CD8 T cells. Adhesion molecules that promote leukocyte adherence are highly expressed in psoriatic lesions.4 After T cells receive primary stimulation and activation, a resulting synthesis of mRNA for interleukin-2 (IL-2) occurs, resulting in a subsequent increase in IL-2 receptors. Psoriasis is considered a Th1-dominant disease due to the increase in cytokines of the Th1 pathway � interferon gamma (IFN-?), IL-2, and interleukin 12 (IL-12) � found in psoriatic plaques.

The increased IL-2 from activated T cells and IL-12 from Langerhans cells ultimately regulate genes that code for the transcription of cytokines such as IFN-?, TNF-?, and IL-2, responsible for differentiation, maturation, and proliferation of T cells into memory effector cells. Ultimately, T cells migrate to the skin, where they accumulate around dermal blood vessels. These are the first in a series of immunologic changes that result in the formation of acute psoriatic lesions. Because the above-described immune response is a somewhat normal response to antigen stimulation, it remains unclear why the T-cell activation that occurs, followed by subsequent migration of leukocytes into the epidermis and dermis, creates accelerated cellular proliferation. Upregulated gene regulation may be a causative factor. Vascular endothelial growth factor (VEGF) and interleukin-8 released from keratinocytes may contribute to the vascularization seen in psoriasis.5

Dendritic cells appear to be involved in the pathogenesis of psoriasis. One type of dendritic cell involved is the Langerhans cells, the outermost sentinel of the immune system that recognizes and captures antigens, migrates to local lymph nodes, and presents them to T cells. The activation of T lymphocytes releases pro-inflammatory cytokines such as TNF-? that lead to keratinocyte proliferation. This hyperproliferative response decreases epidermal transit time (the approximate time it takes for normal maturation of skin cells) from 28 days to 2-4 days and produces the typical erythematous scaly plaques of psoriasis. This understanding of pathogenic mechanisms has led to the development and therapeutic use of TNF-? blocking agents.

About 30 percent of individuals with psoriasis have a family history of the disease in a first- or seconddegree relative. At least nine chromosomal susceptibility loci have been elucidated (PSORS1-9). HLA-Cw6 is a major determinant of phenotypic expression. An association with the PSORS has been found with functional polymorphisms in modifier genes that mediate inflammation (e.g., TNF-?) and vascular growth (e.g., VEGF).6

It is known that psoriasis develops in bone marrow transplant recipients from donors with psoriasis, clears in recipients from donors without psoriasis, and that immunosuppressive drugs are effective in reducing psoriasis.7,8 Given the genetic predisposition to this disease, what can be done to reduce the genetic expression besides resorting to immunosuppressive therapies? A naturopathic approach consists of dietary modification,�therapeutic fasting, omega-3 supplementation, topical natural medicines, herbal medicine, and stress management.

Pizzorno and Murray propose the above-mentioned �unidentified antigens� result from incomplete protein digestion, increased intestinal permeability, and food allergies; bowel toxemia (endotoxins); impaired liver detoxification; bile acid deficiencies; alcohol consumption; excessive consumption of animal fats; nutrient deficiencies (vitamins A and E, zinc, and selenium); and stress.9 These hypotheses, although plausible, have not been adequately tested.

Co-Morbidities

Psoriasis is associated with several co-morbidities, including decreased quality of life, depression, increased cardiovascular risk, type 2 diabetes mellitus, metabolic syndrome, cancer, Crohn�s disease, and psoriatic arthritis. It remains unclear whether cancers, in particular skin cancer and lymphoma, are related to psoriasis or to its treatment. Phototherapy and immunosuppressive therapy can increase the risk of non-melanoma skin cancer, for example.10

Of particular concern is the observed link between psoriasis and cardiovascular disease. Evidence indicates psoriasis is an independent risk factor for cardiovascular disease.11 Dyslipidemia, coronary calcification, increased highly sensitive C-reactive protein (CRP), decreased folate, and hyperhomocysteinemia are found with significantly higher frequency in psoriasis patients.12 Inflammation is the common theme underlying both conditions, characterized by the presence of pro-inflammatory cytokines and endothelial activation.

The inflammatory processes underlying psoriasis also suggest the possibility of omega-3 fatty acid, folate, and vitamin B12 deficiencies, which are also often found in cardiovascular disease.13 High homocysteine and decreased folate levels correlate with Psoriasis Area and Severity Index (PASI). A rapid skin cell turnover rate in psoriasis may result in increased folate utilization and subsequent deficiency.14 The author of one study concludes: �Dietary supplementation of folic acid, B6, and B12 appears reasonable in psoriasis patients, particularly those with elevated homocysteine, low folate and additional cardiovascular risk factors.�15

Psoriatic arthritis is a clinical condition occurring in 25 percent of individuals afflicted with psoriasis.16 In approximately 10 percent of this population, the arthritic symptoms precede the skin lesions. Psoriatic arthritis often presents as seronegative inflammatory arthritis, with a classic presentation consisting of oligoarthritis, distal interphalangeal joint involvement, dactylitis (inflammation of the digits), and calcaneal inflammation.

Opinions conflict whether the skin condition and arthritis represent a differing manifestation of the same disease. Genetic evidence, immunological studies, and treatment response variability suggest they may be two different conditions, perhaps with similar underlying inflammation and immune irregularity.17,18

Although palmoplantar pustulosis (PP) is often described as a subtype of psoriasis, different demographics and genetic analysis suggests a different etiology than psoriasis. On appearance, PP has yellowbrown sterile pustules that appear on palms and soles. Only 25 percent of those affected report chronic plaque psoriasis. PP occurs more frequently in women (9:1/ female:male) and 95 percent of affected people have a current or previous history of smoking. As a result, PP may be considered a co-morbid condition rather than a distinct form of psoriasis.19

Diagnostic Criteria

Psoriasis is classified into several subtypes, with the chronic plaque (psoriasis vulgaris) form comprising approximately 90 percent of cases. Sharply demarcated erythematous silvery scaling plaques occur most commonly on the extensor surface of the elbows, knees, scalp, sacral, and groin regions. Other involved areas include the ears, glans penis, perianal region, and sites of repeated trauma. An active inflammatory case of psoriasis can demonstrate the Koebner phenomenon in which new lesions form at a site of trauma or pressure.

In the future, chronic plaque psoriasis might be found to consist of several related conditions with distinct phenotypical and genotypical characteristics, providing an explanation for its variable response to therapy, especially with biologic agents.

Inverse psoriasis occurs in intertriginous sites and skin folds and is red, shiny, and usually without scaling. Sebopsoriasis, which is often confused with seborrheic dermatitis, is characterized by greasy scales�in the eyebrows, nasolabial folds, and postauricular and presternal areas.

Acute guttate psoriasis occurs in children, adolescents, and young adults approximately two weeks after an acute beta-hemolytic streptococcal infection, such as tonsillitis or pharyngitis, or a viral infection. It manifests as an erythematous, papular eruption with lesions less than 1 cm in diameter on the trunk and extremities. Acute guttate psoriasis is usually self-limited, resolving within 3-4 months. One study indicated only one-third of individuals with guttate psoriasis develop classic plaque psoriasis.20

Pustular psoriasis (von Zumbusch) is also an acute psoriatic eruption. The patient presents with fever and small, monomorphic, painful, sterile pustules, often precipitated by an intercurrent infection or the abrupt withdrawal of systemic or superpotent topical steroids. It can be localized to the palms and soles (palmar-plantar psoriasis) or it can be generalized and potentially life-threatening.

Erythrodermic psoriasis, also life threatening, involves the entire body surface and can result in hypothermia, hypoalbuminemia, anemia, infection, and high-output cardiac failure.

Psoriatic nail disease occurs in approximately 50 percent of psoriasis patients and most commonly manifests as pitting. Other nail changes can include onycholysis, discoloration, thickening, and dystrophy.

Risk Factors

Development of psoriasis involves interaction of multiple genetic risk factors with environmental factors, such as beta-hemolytic streptococcal infection, HIV, stress, and medications (e.g., beta-blockers and lithium). As previously mentioned, folate and vitamin B12 deficiency can also predispose. In addition, there is evidence that alcoholism, cigarette smoking, obesity, type 2 diabetes mellitus, and metabolic syndrome increase risk for developing psoriasis.

With the exception of VEGF, no biomarkers have been found as reliable predictors of psoriasis activity. CRP, soluble adhesion molecules, and soluble cytokine receptors have been investigated but do not correlate with severity.21

Conventional Treatment

Conventional treatment of psoriasis is based on the degree of severity. Mild and limited psoriasis treatment includes topical corticosteroids, tars, anthralin, calcipotriene (a vitamin D3 analog), tazarotene (a retinoid), and phototherapy. Physicians can set realistic expectations for therapy, giving the patient control over the disease without expectation of complete cure. Scalp psoriasis usually responds to salicylic acid shampoos.

Narrow-band UVB is less effective but safer than psoralen plus ultraviolet A (PUVA), which carries with it an increased risk of skin cancer. Sun exposure is another form of phototherapy. UV exposure reduces antigen presenting and affects cell signaling, favoring development of T-helper 2 (Th2) immune responses. Antigen-presenting Langerhans cells are decreased in both number and function.22

A topical combination of calcipotriene and betamethasone (Taclonex�) has shown greater efficacy in severe psoriasis than monotherapy with either alone.23

Patient compliance must be considered when developing a treatment plan. The use of less messy topical solution and foam preparations of topical corticosteroids and calcipotriene (compared to ointments and creams) can improve compliance.

Systemic treatment of severe psoriasis usually involves the use of oral retinoids, methotrexate, cyclosporine, and biological agents that can significantly impact other bodily systems.

The oral retinoid acitretin is teratogenic and is converted to etretinate with concomitant alcohol ingestion. Etretinate has a longer half-life and is more teratogenic than acitretin. Female patients must use two forms of birth control and must not become pregnant for at least three years after treatment. Because of possible interaction with oral contraceptives, St. John�s wort (Hypericum perfoliatum) should be avoided. Other adverse effects include mucocutaneous effects, elevated triglycerides, alopecia, and hepatitis. Treatment with acitretin requires frequent monitoring of blood counts, comprehensive metabolic profiles, and urinalysis. Strategies to reduce acitretin toxicity include intermittent use, reduction of maintenance dose to every other day or every third day, combination treatment with PUVA or topical calcipotriene, low-fat diet, aerobic exercise, fish oil supplementation, and as stated above, alcohol avoidance.

Methotrexate (MTX) is the most commonly used systemic agent for psoriasis and, because it has been available for 35 years, most dermatologists are comfortable with its use. Methotrexate inhibits dihydrofolate reductase (resulting in a deficiency of active folic acid) and induces adenosine A1, a potent anti-inflammatory agonist. Its mechanism of action may be even more complex, evidenced by the fact that caffeine inhibits MTX�s anti-inflammatory effects in rheumatoid arthritis but not in psoriasis or psoriatic arthritis.24 The most common serious adverse effects of MTX are myelosuppression and liver fibrosis. While myelosuppression does not frequently occur, patients using MTX often report symptoms of headache, fatigue, and nausea. Folate supplementation reduces the incidence of megaloblastic anemia, hepatotoxicity, and gastrointestinal intolerance. Although folic acid and folinic acid appear to be equally effective, folic acid is more cost effective.25 However, a recent double-blind study of 22 psoriasis patients stable on long-term MTX therapy revealed folic acid reduced MTX�s efficacy in controlling psoriasis. Patients were randomly assigned to receive 5 mg/day folic acid or placebo for 12 weeks. The mean PASI increased (worsened) in the folic acid group, from 6.4 at baseline to 10.8 at 12 weeks. In the placebo group, the mean PASI fell from 9.8 at baseline to 9.2 at 12 weeks (p<0.05 for the difference in the change between groups).26

Cyclosporine, a potent and toxic drug, is sometimes considered for cases not controlled with acitretin, PUVA, or MTX, but is contraindicated in patients with abnormal renal function, poorly controlled hypertension, hepatic dysfunction, or immunosuppression. Prolonged use inevitably results in renal damage. Blood pressure and creatinine monitoring is essential.

Biological agents block T-cell activation and TNF-?. Alefacept (Amevive�) interferes with T-cell activation and reduces circulating CD 45 RO+ T cells. This drug is a fusion protein of the Fc receptor of human IgG1 and LFA3, a co-stimulatory ligand, which interacts with CD2 on the surface of T-cells. CD4 cells must be monitored weekly during treatment with this agent.

Efalizumab (Raptiva�) is a humanized antibody to CD11 that interferes with T-cell trafficking into inflamed tissues and prevents T-cell activation. Although it is rapidly effective, rebound may occur.

TNF-? blockers downregulate proinflammatory gene expression and reverse the psoriatic phenotype. Etanercept (Enbrel�) is a fusion protein directed against soluble TNF-?. Infliximab (Remicade�) is a mouse/human chimeric monoclonal antibody against soluble and cell-bound TNF-?, while adalimumab (Humira�) is a human monoclonal antibody against TNF-?. These TNF-? inhibitors are administered by injection and have been associated with the induction of various autoimmune phenomena. Like TNF-? itself, TNF-? inhibitors can have both proinflammatory and anti-inflammatory activities. Just because a particular agent blocks TNF-?, it does not necessarily benefit psoriasis. If a patient is genetically predisposed to overproducing TNF-?, blocking it may not be sufficient to produce benefit.27 Possible risks of TNF-? blockers include reactivation of latent tuberculosis, hepatotoxicity, lymphoma, and congestive heart failure.

Challenges that remain with biologics for psoriasis include: (1) understanding the predominant mechanism in psoriasis and psoriatic arthritis; (2) understanding different patient responses to therapy; (3) predicting clinical response before or early in therapy; (4) developing oral, inhaled, and topical formulations; and (5) determining whether treatment alters longterm outcome.

Fumaric acid is the primary psoriasis therapy in Germany. It decreases T-cell dependent cytokines, but is not as effective as other conventional treatments, and carries a high risk of toxicity and gastrointestinal intolerance.

Providing rotational and combination therapies increases efficacy and decreases toxicity of treatment. The future may bring stem-cell therapy and gene-based therapies, including �antisense� treatments that directly inhibit psoriasis-specific genes. However, the adverse effects and toxicity of conventional psoriasis treatments necessitate safer and effective natural treatments that can be used as alternatives or in an integrative fashion.

Natural Treatments For Psoriasis

Diet

An evidence-based approach suggests psoriasis, essentially an inflammatory disorder, should benefit from an anti-inflammatory diet, identification, elimination and/or rotation of allergenic foods, and therapeutic fasting.28-30 Although there is no published data on food allergy avoidance, many psoriasis patients show increased sensitivity to gluten and their psoriasis symptoms improve on a gluten-free diet.31 Measurement of antibodies to tissue transglutaminase and gliadin can help identify this subgroup. Evidence also suggests maintaining a healthy weight benefits psoriasis patients, since psoriasis positively correlates with increased body mass index.32

The balance between proinflammatory and anti-inflammatory eicosanoids is influenced in large part by the type of dietary fatty acids consumed. An antiinflammatory diet consists basically of an emphasis on �good fats� (cold water fish, nuts, seeds, olive oil, other high quality oils), whole grains, legumes, vegetables, and fruits and the avoidance of �bad fats� (saturated animal fats, trans fats, fried and processed foods, poor quality oils) and refined carbohydrates. In addition, an excessive amount of omega-6 fatty acids in the diet can contribute to an inflammatory response.33 The primary sources of dietary omega-6 oils are vegetable oils such as corn, soy, safflower, and sunflower, while the primary sources of arachidonic acid are meat, eggs, and dairy.

Prostaglandin E2 (PGE2) is a prominent eicosanoid derived from the omega-6 fatty acid arachidonic acid. A dominant action of PGE2 as a messenger molecule is to enhance sensitivity in pain neurons, increase swelling, and constrict blood vessels. Over-consumption of omega-6 oils provides excess substrate for the synthesis of PGE2, which drives an aggressive and sustained inflammatory response. Prostaglandin E3 (PGE3) is�derived from the omega-3 fatty acid, eicosapentaenoic acid (EPA). Higher levels of PGE3 reduce sensitivity to pain, relax blood vessels, increase blood flow, and support the body�s natural anti-inflammatory response (Figure 1).

psoriasis illustration

While both PGE2 and PGE3 are necessary for proper homeostasis, the relative amounts of these competing messenger molecules either contribute to or mitigate chronic inflammatory syndromes. EPA is thought to act by competing with arachidonic acid for binding sites on cyclooxygenase-2 (COX-2), producing a less potent inflammatory mediator, therefore reducing inflammation.34

Prior to the Industrial Revolution, there were no significant sources of omega-6 vegetable oils in the diet. Most cultures consumed diets low in these oils and high in fish or range-fed beef or bison higher in omega-3s, creating a ratio of omega-6:omega-3 that was approximately 3:1. The Industrial Revolution brought with it the knowledge and tools to refine vegetable oils, resulting in a rapid shift in dietary habits for most Western cultures. The ratio of omega-6:omega-3 was quickly pushed toward the current estimate of as high as 11:1 omega-6:omega-3.35 The human body has not been able to genetically adapt to this dramatic shift in fatty acid consumption.

Many modern cultures consume copious amounts of vegetable oils, mostly in processed foods. For example, soy oil production for food consumption increased 1,000-fold between 1909 and 1999.36 In addition, livestock, poultry, and farmed fish are being fed cornmeal and soy-based feed, which raises the omega-6 content of the meat and fish. When farm animals are raised on grass, worms, or other natural diets, the tissues are naturally higher in omega-3 fatty acids.37

The beef industry touts �marbling� in finished beef products, which is due to the corn and soy feed. Corn- and soy-fed cattle have a higher omega-6 fatty acid content compared to grass-fed cattle. While grassfed cattle can contain up to 4-percent omega-3 fatty acids, corn-fed cattle typically contains 0.5-percent omega-3s.37

The standard American diet supplies an average omega-6:omega-3 ratio of approximately 11:1. A vegetarian-based diet may put an individual at risk for�eating high amounts of vegetable oils and soy products, and low amounts of fish, which can tip the balance toward a pro-inflammatory state. Reducing dietary vegetable oils and increasing the omega-3 fats EPA and docosahexaenoic acid (DHA) by consuming fatty fish such as cod, salmon, mackerel, and sardines can benefit individuals experiencing chronic inflammatory conditions.33

Several herbs used as seasonings, including turmeric, red pepper, cloves, ginger, cumin, anise, fennel, basil, rosemary, garlic, and pomegranate, can block nuclear factor-kappaB (NF?B) activation of inflammatory cytokines.38

Dietary approaches that modify fatty acid intake can influence the eicosanoid profile in such a way that inflammatory processes such as arachidonic acid production and T-cell activation are dampened, while cytokines such as interleukin-4 (the primary cytokine responsible for stimulating a Th2 immune response) are upregulated.34

Nutritional Supplementation

Essential Fatty Acids

Essential fatty acids (EFAs) influence the pathophysiology of psoriasis in three ways: first, EFAs impact the kinetics of cell membranes; second, EFAs impact dermal and epidermal blood flow via improved endothelial function; and third, EFAs act as an immunomodulating agent through their impact on eicosanoids. EFAs are used as basic substrates in the development of the phospholipid bi-layer in virtually every cell in the human body, including the dermis and epidermis. They create structural integrity that regulates fluidity, which impacts cell transport, messenger binding, and cell communication. Omega-3 fatty acids can act both directly and indirectly on endothelial function by reducing mononuclear cell cytokines such as IL-1 and TNF?, 39 decreasing formation of chemo-attractant protein platelet-derived growth factor (PDGF), increasing bioavailability of nitric oxide, and reducing expression of adhesion molecules. The cumulative effect modulating these bioactive mediators is to prevent vascularization, or new blood vessel growth within the psoriatic plaque, while simultaneously allowing improved perfusion of dermal tissue.

Components of both natural and acquired immunity, including the production of key immune modulators, can be affected by omega-3 and -6 fatty acid intake, as discussed above. Immunomodulatory effects of omega-3 fatty acids include suppression of lymphoproliferation, CD4+ cells, antigen presentation, adhesion molecule presentation, Th1 and Th2 responses, and pro-inflammatory cytokine production.34

Several studies have demonstrated the benefit of intravenous or oral supplementation of fish oil for psoriasis.40-42 In a study by Mayser et al, intravenous infusions of omega-3 fatty acids led to an increase in the anti-inflammatory leukotriene B5 (LTB5) within 4-7 days of starting treatment, when compared to control patients.43 In this trial, patients received either an omega-3 or omega-6 preparation twice daily for 10 days. No side effects were noted.

EPA competes with arachidonic acid (AA) for 5-lipoxygenase and produces LTB5, which is only one-tenth as potent as the inflammatory mediator leukotriene B4 (LTB4). Levels of LTB4 have been shown to be elevated in psoriatic plaques and demonstrate chemotactic properties necessary for infiltration of leukocyte and keratinocyte proliferation.43

Ziboh�s review article on omega-3s and psoriasis references six studies conducted using oral fish oil supplementation with mixed results. Unfortunately, original references cannot be found. Two studies were double-blind and placebo-controlled, using 1.8 g EPA and DHA over courses of eight weeks and 12 weeks. The eight-week study demonstrated benefit in itching, scaling, and erythema, while the 12-week study showed no benefit.44

Three open studies were conducted, providing 10-18 g EPA and DHA daily for eight weeks. All studies showed improvement, with two studies demonstrating mild-to-moderate and one study demonstrating moderate-to-excellent improvement in scaling, itching, and lesion thickness. One open study combined with a low-fat diet showed a significant reduction in psoriatic symptoms.44,45

Several studies have explored the use of topical fish oil at varying EPA concentrations. Some studies reported benefits, including a reduction in plaque thickness and scaling.46,47 In one study by Puglia et al, fish oil extracts and ketoprofen were applied topically to�psoriatic lesions, with an observed reduction in erythema.48 The most significant drawback to topical fish oil application is compliance due to the odor.

Fish oil has also proven to be beneficial in autoimmune joint conditions such as rheumatoid arthritis (RA).49 While fish oil supplementation has not been used in clinical trials for the treatment of psoriatic arthritis, it may be beneficial in treating this condition, which has many similarities to RA, including a common underlying inflammatory mechanism and immune dysfunction.

Folate

Methotrexate therapy results in a folate deficiency. As mentioned above, in patients receiving MTX for psoriasis, folate supplementation reduced the incidence of hepatotoxicity and gastrointestinal intolerance but might impair the efficacy of MTX.24 When supplementing with folic acid or the active forms, folinic acid or 5-methyltetrahydrofolate, the recommended dose is 1-5 mg/day.

Bioactive Whey Protein Isolate

XP-828L is a novel dietary supplement made of a protein extract derived from bovine whey that has recently been shown to be beneficial in psoriasis.50,51 The bioactive profile of XP-828L is likely due to the presence of growth factors, immunoglobulins, and active peptides found in this specific whey extract. An in vitro study demonstrated XP-828L has immune-regulating effects, including inhibiting the production of Th1 cytokines such as IFN-g and IL-2, which may make it effective in treating T-helper 1-related disorders, such as psoriasis.52

An open-label study was conducted on 11 adult patients with chronic, stable plaque psoriasis on two percent or more of total body surface area. Study participants received 5 g twice daily of XP-828L for 56 days. Evaluations using PASI and Physician�s Global Assessment (PGA) scores were made on the initial screening day and again on days 1, 28, and 56. At the conclusion of the study, seven of the 11 subjects had a reduced PASI score that ranged from 9.5 percent to 81.3 percent.50 The results of a larger double-blind,�placebo-controlled study of 84 individuals with mildto-moderate psoriasis showed XP-828L (5 g/day for 56 days) significantly reduced the PGA score compared to placebo (p<0.05). No adverse affects were noted from any study participants in either study.50,51

Vitamin D

It has been established that patients with disseminated psoriasis have significantly decreased serum levels of the biologically active form of vitamin D, 1-alpha,25-dihydroxyvitamin D3 (1-?,25(OH)2D3; calcitriol) compared to age- and sex-matched controls and also compared to patients with moderate psoriasis.53 Whether this is a contributing factor to psoriasis or a result of the disorder has not been elucidated.

Keratinocytes in the epidermis convert 7-dehydrocholesterol to vitamin D3 in the presence of UVB. Sunlight, UVB phototherapy, oral calcitriol, and topical vitamin D analogs are effective therapy for psoriasis due to vitamin D�s anti-proliferative and pro-differentiating actions on keratinocytes.54-56

Calcitriol-binding to vitamin D receptors (VDR) in the skin modulates the expression of a large number of genes including cell cycle regulators, growth factors, and their receptors. Polymorphisms of the VDR gene are associated with psoriasis and may predispose to the development of psoriasis and resistance to calcipotriol therapy, as well as contribute to liver dysfunction in patients with psoriasis.57

Given vitamin D�s importance in psoriasis, cancer, inflammatory diseases, and other conditions, it has been suggested by some investigators that recommendations for sun protection and skin cancer prevention may need to be re-evaluated to allow for sufficient vitamin D status. A recent study showed abundant sun exposure in a sample of adults in Hawaii did not necessarily ensure vitamin D adequacy, which points to the need for vitamin D supplementation to achieve optimal blood levels.58

Studies have demonstrated that oral vitamin D can be safely taken in daily doses of up to 5,000 IU, with some experts recommending up to 10,000 IU daily to correct a deficiency.59-61 Oral and topical vitamin D, sunlight, and UVB phototherapy have shown considerable efficacy in the treatment of psoriasis.56

Topical Treatments Of Psoriasis

Several topical treatments for psoriasis may provide benefit, including calcipotriene (Dovonex�; a synthetic vitamin D3 analogue), Berberis aquifolium cream (10%)62 (Psoriaflora�; Relieva�), curcumin gel (1%), Aloe vera, and a flavonoid-rich salve (Flavsalve�).

Curcumin gel yielded 90-percent resolution of plaques in 50 percent of patients within 2-6 weeks; the remainder of the study subjects showed 50- to 85-percent improvement. Curcumin was found to be twice as effective as calcipotriol cream (which generally takes three months to exert its full effect). The mechanism of curcumin is as a selective phosphorylase kinase inhibitor, thereby reducing inflammation through inhibition of NF?B.63

A controlled trial of Aloe vera extract cream (0.5%) in 60 patients for 4-12 months demonstrated a significant clearing of psoriatic plaques (82.8%) compared to placebo (7.7%) (p<0.001). In addition, the PASI decreased to a mean of 2.2.64

The scaliness of psoriasis benefits from the use of emollients. Intercellular lipids such as ceramides (lipid molecules composed of fatty acids and sphingosine) play an important role in the regulation of skin-water barrier homeostasis and water-holding capacity. It has been shown that ceramides are decreased in the psoriatic epidermis. Newer ceramide-containing emollients (e.g., CeraVe�, Mimyx�, Aveeno Eczema Care) have shown benefit in psoriasis and may improve skin barrier function and decrease water loss.65

Botanical Influences

A Chinese herbal formula (Herose� Psoria Capsule) has demonstrated safety and efficacy in the treatment of severe plaque psoriasis.66 Herose consists of rhizoma Zingiberis, radix Salviae miltiorrhizae, radix Astragali, ramulus Cinnamomi, radix Paeoniae alba, radix Codonopsis pilosula, and semen Coicis. In an openlabel trial, 15 subjects took four Herose capsules (450 mg each) three times daily for 10 months. The investigator evaluated the PASI and therapeutic response to Herose for each patient. The formula is intended for warming the yang and promoting blood circulation.

Lifestyle Interventions

Lifestyle factors such as cigarette smoking and alcohol consumption are associated with severity of psoriasis.67 Physical activity and outdoor activities (taking precautions not to sunburn) are beneficial.68 Bathing and sunbathing at the Dead Sea for four weeks resulted in a decrease of PASI of 81.5 percent, a 78-percent decrease in keratinocyte hyperplasia, and almost total elimination of T lymphocytes from the epidermis, with a low number remaining in the dermis.69

Stress management can benefit individuals with psoriasis. Subjects who listened to a guided meditation tape while undergoing phototherapy cleared four times faster than those who received phototherapy only, as judged by two independent dermatologists. Psoriasis status was assessed in three ways: direct inspection by clinic nurses; direct inspection by physicians blinded to the patient�s study condition (tape or no-tape); and blinded physician evaluation of photographs of psoriasis lesions. Four sequential indicators of skin status were monitored during the study: a First Response Point, a Turning Point, a Halfway Point, and a Clearing Point. Subjects in the tape groups reached the Halfway Point (p= 0.013) and the Clearing Point (p=0.033) significantly more rapidly than those in the no-tape condition, for both UVB and PUVA treatments.70 Finally, psychotherapy can be an essential adjunct for individuals with persistent unresolved psychological issues such as anxiety, depression, and the psychosocial stress of this chronic skin disease.

Discussion

Psoriasis is characterized by T-cell activation that releases pro-inflammatory cytokines such as TNF-?, leading to keratinocyte proliferation and the typical skin lesions of psoriasis.

The conventional approach to psoriasis consists of utilizing topical and/or oral corticosteroids, other immunosuppressant drugs, oral retinoids, UV light, and several (not necessarily novel, having been used previously for Crohn�s and RA) biological agents. Although these treatments can be highly effective at controlling the disease, none are universally safe and effective, and each carries a considerable risk profile.

There is some evidence for the use of dietary modification and fish oil to decrease inflammation in psoriasis. More research is warranted to clarify the use�of these and various topical botanical therapies and lifestyle interventions for improving clinical symptoms, decreasing the phenotypic expression of psoriasis, and providing safe and effective treatments.

 

blank
References:

1. Griffiths CEM, Camp RDR, Barker JNWN.
Psoriasis. In: Burns DA, Breathnach SM, Cox N,
Griffiths CE, eds. Rook�s Textbook of Dermatology. 7th
ed. Oxford: Blackwell; 2005:35.1-35.69.
2. Nevitt GJ, Hutchinson PE. Psoriasis in the
community; prevalence, severity and patients belief
and attitudes towards the disease. Br J Dermatol
1996;135:533-537.
3. Farber EM, Nall ML. The natural history of psoriasis
in 5600 patients. Dermatologica 1974;148:1-18.
4. Robert C, Kupper TS. Inflammatory skin diseases,
T cells and immune surveillance. N Engl J Med
1999;341:1817-1828.
5. Simonetti O, Lucarini G, Goteri G, et al. VEGF is
likely a key factor in the link between inflammation
and angiogenesis in psoriasis: results of an
immunohistochemical study. Int J Immunopathol
Pharmacol 2006;19:751-760.
6. Capon F, Munro M, Barker J, Trembath R. Searching
for the major histocompatibility complex psoriasis
susceptibility gene. J Invest Dermatol 2002;118:745-
751.
7. Wahie S, Alexandroff A, Reynolds NJ, Meggit SJ.
Psoriasis occurring after myeloablative therapy and
autologous stem cell transplantation. Br J Dermatol
2006;154:194-195.
8. Eedy DJ, Burrows D, Bridges JM, Jones FG.
Clearance of severe psoriasis after allogenic bone
marrow transplantation. BMJ 1990;300:908.
9. Pizzorno JE, Murray MT. Textbook of Natural
Medicine. 3rd ed. St. Louis, MO: Churchill
Livingstone; 2006:2080.
10. Lindelof B, Eklund G, Liden S, Stern RS. The
prevalence of malignant tumors in patients with
psoriasis. J Am Acad Dermatol 1990;22:1056-1060.
11. Mrowietz U, Elder JT, Barker J. The importance of
disease associations and concomitant therapy for the
long-term management of psoriasis patients. Arch
Dermatol Res 2006;298:309-319.
12. Rocha-Pereira P, Santos-Silva A, Rebelo I, et al.
Dyslipidemia and oxidative stress in mild and in
severe psoriasis as a risk for cardiovascular disease.
Clin Chim Acta 2001;303:33-39.
13. Ludwig RJ, Herzog C, Rostock A, et al. Psoriasis:
a possible risk factor for development of coronary
artery calcification. Br J Dermatol 2007;156:271-276.

14. Vanizor Kural B, Orem A, Cimsit G, et al.
Plasma homocysteine and its relationships with
atherothrombotic markers in psoriatic patients. Clin
Chim Acta 2003;332:23-30.
15. Malerba M, Gisondi P, Radaeli A, et al. Plasma
homocysteine and folate levels in patients
with chronic plaque psoriasis. Br J Dermatol
2006;155:1165-1169.
16. Zachariae H. Prevalence of joint disease in patients
with psoriasis: implications for therapy. Am J Clin
Dermatol 2003;4:441-447.
17. Ho P, Bruce IN, Silman A, et al. Evidence for
common genetic control in pathways of inflammation
for Crohn�s disease and psoriatic arthritis. Arthritis
Rheum 2005;52:3596-3602.
18. Pitzalis C, Cauli A, Pipitone N, et al. Cutaneous
lymphocyte antigen-positive T lymphocytes
preferentially migrate to the skin but not to the joint
in psoriatic arthritis. Arthritis Rheum 1996;39:137-
145.
19. Asumalahti K, Ameen M, Suomela S, et al. Genetic
analysis of PSORS1 distinguishes guttate psoriasis
and palmoplantar pustulosis. J Invest Dermatol
2003;120:627-632.
20. Martin BA, Chalmers RJ, Telfer NR. How great
is the risk of further psoriasis following a single
episode of acute guttate psoriasis? Arch Dermatol
1996:132:717-718.
21. Creamer D, Allen MH, Groves RW, Barker JN.
Circulating vascular permeability factor/vascular
endothelial growth factor in erythroderma. Lancet
1996;348:1101.
22. Zanolli MD, Camisa C, Feldman S, et al. Psoriasis:
the high notes on current treatment. Program of the
American Academy of Dermatology, Academy 2000;
August 5, 2000; Nashville, TN.
23. Kaufmann R, Bibby AJ, Bissonnette R, et al. A new
calcipotriol/betamethasone dipropionate formulation
(Daivobet) is an effective once-daily treatment for
psoriasis vulgaris. Dermatology 2002;205:389-393.
24. Swanson DL, Barnes SA, Mengden Koon SJ, elAzhary
RA. Caffeine consumption and methotrexate
dosing requirement in psoriasis and psoriatic arthritis.
Int J Dermatol 2007;46:157-159.
25. Strober BE, Menon K. Folate supplementation during
methotrexate therapy for patients with psoriasis. J
Am Acad Dermatol 2005;53:652-659.
26. Salim A, Tan E, Ilchyshyn A, Berth-Jones J. Folic acid
supplementation during treatment of psoriasis with
methotrexate: a randomized, double-blind, placebocontrolled
trial. Br J Dermatol 2006;154:1169-1174.
27. Fiorentino D. The yin and yang of TNF-(alpha)
inhibition. Arch Dermatol 2007;143:233-236.
28. Wolters M. Diet and psoriasis: experimental data and
clinical evidence. Br J Dermatol 2005;153:706-714.
29. Brown AC, Hairfield M, Richards DG, et al. Medical
nutrition therapy as a potential complementary
treatment for psoriasis � five case reports. Altern Med
Rev 2004;9:297-307.
30. Lithell H, Bruce A, Gustafsson IB, et al. A fasting
and vegetarian diet treatment trial on chronic
inflammatory disorders. Acta Derm Venereol
1983;63:397-403.
31. Chalmers RJ, Kirby B. Gluten and psoriasis. Br J
Dermatol 2000;142:5-7.
32. Naldi L, Parazzini F, Peli L, et al. Dietary factors and
the risk of psoriasis. Results of an Italian case-control
study. Br J Dermatol 1996;134:101-106.
33. Adam O, Beringer C, Kless T, et al. Antiinflammatory
effects of a low arachidonic acid diet
and fish oil in patients with rheumatoid arthritis.
Rheumatol Int 2003;23:27-36.
34. Calder PC. n-3 Polyunsaturated fatty acids,
inflammation, and inflammatory diseases. Am J Clin
Nutr 2006;83:1505S-1519S.
35. Yehuda S.Omega-6/omega-3 ratio and brain-related
functions. World Rev Nutr Diet 2003;92:37-56.
36. Sirtori CR. Risks and benefits of soy phytoestrogens
in cardiovascular diseases, cancer, climacteric
symptoms and osteoporosis. Drug Saf 2001;24:665-
682.
37. Marchello MJ, Driskell JA. Nutrient composition of
grass- and grain-finished bison. Great Plains Research
2001;11:65-82.
38. Aggarwal BB, Shishodia S. Suppression of the
nuclear factor-kappaB activation pathway by spicederived
phytochemicals: reasoning for seasoning. Ann
N Y Acad Sci 2004;1030:434-441.
39. Yaqoob P. Fatty acids as gatekeepers of immune cell
regulation. Trends Immunol 2003;24:639-645.
40. Bittiner SB, Tucker WF, Cartwright I, Bleehen SS. A
double-blind, randomised, placebo-controlled trial of
fish oil in psoriasis. Lancet 1988;1:378-380.
41. Gupta AK, Ellis CN, Tellner DC, et al. Double-blind,
placebo-controlled study to evaluate the efficacy
of fish oil and low-dose UVB in the treatment of
psoriasis. Br J Dermatol 1989;120:801-807.
42. Mayser P, Mrowietz U, Arenberger P, et al. Omega-3
fatty acid-based lipid infusion in patients with
chronic plaque psoriasis: results of a double-blind,
randomized, placebo-controlled, multicenter trial. J
Am Acad Dermatol 1998;38:539-547.
43. Mayser P, Grimm H, Grimminger F. n-3 fatty acids in
psoriasis. Br J Nutr 2002;87:S77-S82.
44. Ziboh VA. The role of n-3 fatty acids in psoriasis. In:
Kremer J, ed. Medicinal Fatty Acids in Inflammation.
Basel, Switzerland: Birkhauser Verlag; 1998:45-53.

45. Calder PC. n-3 Polyunsaturated fatty acids,
inflammation and immunity: pouring oil on troubled
waters or another fishy tale? Nutr Res 2001;21:309-
341.
46. Zulfakar MH, Edwards M, Heard CM. Is there a role
for topically delivered eicosapentaenoic acid in the
treatment of psoriasis? Eur J Dermatol 2007;17:284-
291.
47. Richards H, Thomas CP, Bowen JL, Heard CM.
In vitro transcutaneous delivery of ketoprofen and
polyunsaturated fatty acids from a pluronic lecithin
organogel vehicle containing fish oil. J Pharm
Pharmacol 2006;58:903-908.
48. Puglia C, Tropea S, Rizza L, et al. In vitro
percutaneous absorption studies and in vivo
evaluation of anti-inflammatory activity of essential
fatty acids (EFA) from fish oil extracts. Int J Pharm
2005;299:41-48.
49. Cleland LG, James MJ. Fish oil and rheumatoid
arthritis: antiinflammatory and collateral health
benefits. J Rheumatol 2000;27:2305-2307.
50. Poulin Y, Pouliot Y, Lamiot E, et al. Safety and
efficacy of a milk-derived extract in the treatment of
plaque psoriasis: an open-label study. J Cutan Med
Surg 2005;9:271-275.
51. Poulin Y, Bissonnette R, Juneau C, et al. XP-828L
in the treatment of mild to moderate psoriasis:
randomized, double-blind, placebo-controlled study. J
Cutan Med Surg 2006;10:241-248.
52. Aattouri N, Gauthier SF, Santure M, et al.
Immunosuppressive effect of a milk-derived extract.
12th International Congress of Immunology and 4th
Annual Conference of FOCIS. Montreal, Canada;
July 18-23, 2004.
53. Staberg B, Oxholm A, Klemp P, Christiansen C.
Abnormal vitamin D metabolism in patients with
psoriasis. Acta Derm Venereol 1987;67:65-68.
54. Reichrath J. Vitamin D and the skin: an ancient
friend, revisited. Exp Dermatol 2007;16:618-625.
55. Osmancevic A, Landin-Wilhelmsen K, Larko O,
et al. UVB therapy increases 25(OH) vitamin D
syntheses in postmenopausal women with psoriasis.
Photodermatol Photoimmunol Photomed 2007;23:172-
178.
56. Perez A, Raab R, Chen TC, et al. Safety and efficacy
of oral calcitriol (1,25-dihydroxyvitamin D3) for the
treatment of psoriasis. Br J Dermatol 1996;134:1070-
1078.
57. Okita H, Ohtsuka T, Yamakage A, Yamazaki
S. Polymorphism of the vitamin D(3) receptor
in patients with psoriasis. Arch Dermatol Res
2002;294:159-162.
58. Binkley N, Novotny R, Krueger D, et al. Low vitamin
D status despite abundant sun exposure. J Clin
Endocrinol Metab 2007;92:2130-2135.
59. Grant WB, Holick MF. Benefits and requirements of
vitamin D for optimal health: a review. Altern Med
Rev 2005;10:94-111.
60. Hollis BW. Circulating 25-hydroxyvitamin
D levels indicative of vitamin D sufficiency:
implications for establishing a new effective dietary
intake recommendation for vitamin D. J Nutr
2005;135:317-322.
61. Vieth R, Bischoff-Ferrari H, Boucher BJ, et al. The
urgent need to recommend an intake of vitamin D
that is effective. Am J Clin Nutr 2007;85:649-650.
62. Gulliver WP, Donsky HJ. A report on three recent
clinical trials using Mahonia aquifolium 10% topical
cream and a review of the worldwide clinical
experience with Mahonia aquifolium for the treatment
of plaque psoriasis. Am J Ther 2005;12:398-406.
63. Heng MC, Song MK, Harker J, Heng MK. Druginduced
suppression of phosphorylase kinase activity
correlates with resolution of psoriasis as assessed
by clinical, histological and immunohistochemical
parameters. Br J Dermatol 2000;143:937-949.
64. Syed TA, Ahmad SA, Holt AH, et al. Management
of psoriasis with Aloe vera extract in a hydrophilic
cream: a placebo-controlled, double-blind study. Trop
Med Int Health 1996;1:505-509.
65. Lew BL, Cho Y, Kim J, et al. Ceramides and cell
signaling molecules in psoriatic epidermis: reduced
levels of ceramides, PKC-alpha, and JNK. J Korean
Med Sci 2006;21:95-99.
66. Yuqi TT. Review of a treatment for psoriasis using
Herose, a botanical formula. J Dermatol 2005;32:940-
945.
67. Chodorowska G, Kwiatek J. Psoriasis and cigarette
smoking. Ann Univ Mariae Curie Sklodowska [Med]
2004;59:535-538.
68. Schiener R, Brockow T, Franke A, et al. Bath PUVA
and saltwater baths followed by UV-B phototherapy
as treatments for psoriasis: a randomized controlled
trial. Arch Dermatol 2007;143:586-596.
69. Hodak E, Gottlieb AB, Segal T, et al. Climatotherapy
at the Dead Sea is a remittive therapy for psoriasis:
combined effects on epidermal and immunologic
activation. J Am Acad Dermatol 2003;49:451-457.
70. Kabat-Zinn J, Wheeler E, Light T, et al. Influence
of a mindfulness meditation-based stress reduction
intervention on rates of skin clearing in patients
with moderate to severe psoriasis undergoing
phototherapy (UVB) and photochemotherapy
(PUVA). Psychosom Medicine 1998;60:625-632.

Close Accordion
Nutritional Regulation for Inflammatory Bowel Disease

Nutritional Regulation for Inflammatory Bowel Disease

Inflammatory bowel disease is an umbrella term used to describe a group of gastrointestinal diseases characterized by chronic, ongoing inflammation of all or part of the gastrointestinal tract, or GI tract, such as Crohn’s disease, or CD, and ulcerative colitis, UC. While many factors have been determined to cause inflammatory bowel disease, research studies have concluded that nutrition can increase the risk of gastrointestinal diseases, including inflammatory bowel disease.

 

How does nutrition affect inflammatory bowel disease?

 

Nutrient deficiencies are common among individuals with inflammatory bowel disease, or IBD. Both complete parenteral and enteral nutrition can provide significant supportive treatment for patients with IBD, however, in adults those alone may not be helpful as a form of primary treatment. Clinical intervention using omega-3 polyunsaturated fatty acids found in fish oil could be beneficial for the nutritional regulation of IBD patients and recent research studies have emphasized the function of PPAR on NF?B action towards its possible beneficial impact on dietary lipids for overall intestinal functioning.

 

Nutrition in Inflammatory Bowel Disease

 

Specific antibody isotypes of essential milk proteins are located in both UC and CD patients. In CD, the antibodies are associated with disease. Although cultural origin, rather than the IBD disease condition, seems to be the primary cause of lactose intolerance, the avoidance of milk products by IBD patients is extensive. Lack of breast-feeding during infancy was associated with CD but not UC. Additionally, higher carbohydrate intake was recorded in CD. Others have suggested a deficiency of dietary fiber as a predisposing factor for IBD. The growth of UC has also been associated with higher intakes of polyunsaturated fatty acids (MUFA), n6 polyunsaturated fatty acids (n6 PUFA), sulphur-containing diets and vitamin B6.

 

Deficiencies

 

Inflammatory bowel disease is related to several nutritional deficiencies, such as anemia, hypoalbuminemia, hypomagnesia, hypocalcemia and hypophosphatemia, including deficiencies in folic acid, niacin, vitamins A, B12, C, and D, in addition to deficiencies of iron, magnesium and zinc. Further research studies are needed to determine if reduced levels of micronutrients are of some significance to the result of gastrointestinal diseases. Plasma antioxidant concentrations are lower in IBD patients, especially those who have an active form of the disease. Antioxidant action, evaluated by measuring selenium levels and erythrocyte glutathione peroxidase activity, is inversely associated with inflammatory biomarkers, such as TNF?. Hyperhomocysteinemia is more prevalent in patients with IBD, and is characterized with low serum as well as reduced concentrations of vitamin B12, folate and B6.

 

Several mechanisms are responsible for the malnutrition observed in IBD patients. Primarily, there’s a decline in the oral consumption of nutrients due to abdominal pain and anorexia. Second, the mucosal inflammation and related diarrhea reduces blood, protein, minerals, electrolytes and trace components. Paradoxically, multiple resections or bacterial vaginosis might have an adverse nutrient impact; and finally, herbal remedies may also cause malnutrition. By way of instance, sulfasalazine reduces nitric acid absorption, and corticosteroids reduce calcium absorption in addition to negatively impacting protein metabolism. Alterations in energy metabolism may result in increased resting energy expenditure and lipid oxidation in patients with inflammatory bowel disease. There are many effects of malnutrition and each can decrease bone mineral density, in addition to growth retardation and delayed sexual maturity in children. Osteoporosis may also be involved as a consequence of pro-inflammatory cytokine profiles.

 

Nutritional treatment may take on a range of forms including Total Parenteral Nutrition (TPN) and Complete Enteral Nutrition (TEN). The diets used are elemental, polymeric, and exception diets. Elemental diets contain nutrients reduced to their fundamental elements: amino acids, such as proteins, sugar for carbs, and short-chain triglycerides, such as fats. Polymeric formulas contain entire proteins, such as nitrogen, glucose polymers for carbs and long-chain triglycerides for fat or starch.

 

Total Parenteral Nutrition (TPN)

 

Using TPN for the nutritional regulation of IBD is based on specific theoretical benefits, including how: gut rest may be beneficial since it reduces motor and transportation function in the diseased intestine; a drop in antigenic stimulation can remove the immunologic reactions to food, particularly in the presence of diminished intestinal permeability; TPN promotes protein synthesis in the gut which provides cell renewal, recovery, and alteration of impaired immunocompetence.

 

Researchers demonstrated remission rates of 63 percent to 89 percent with TPN in a large retrospective collection of CD patients which were difficult in standard medical management. But, Matuchansky et al highlighted that there have been high relapse rates (40%-62%) after two decades. It’s been implied that TPN be utilized exclusively in a nutritionally supportive function. In UC, there’s absolutely no evidence for much better results with TPN. Though remission rates of 9 percent to 80 percent are reported, TPN provided to patients with acute colitis seems to be beneficial as perioperative nutritional support. In patients with moderate disease, TPN is significantly more successful but isn’t better than steroid treatment, and so the invasiveness and price of TPN are unjustified. Any advantages related to TPN might be due to the nutritional regulation, rather than gut rest, as gut rest alone has no impact on disease activity. Accordingly, though TPN has a function in patients using a non-functioning gut or the brief gut syndrome because of excess resections, TPN is of limited use as a primary treatment in IBD. This isn’t designed to be an extensive breakdown of TPN, but it needs to be cautioned that in specialist centers, TPN is associated with complications like sepsis and cholestatic liver disease.

 

Total enteral nutrition (TEN), Elemental & Defined Formula Diets

 

TEN prevents possible toxic dietary variables and antigenic exposure, because there are only amino acids, sugar or oligosaccharides and very low lipid content. TEN isn’t associated with cholestasis, biliary sludge or gallstone formation, as can be observed with TPN. Atrophy of the small intestinal mucosa was discovered in animal models receiving long-term TPN, yet this atrophy is prevented with TEN. Additionally, a 6-wk TPN therapy in dogs led to marked decrease in pancreatic fat, a reduction in small intestinal mass as well as a decline in intestinal disaccharidase activity in puppies. Because of this, TEN is more preferable than TPN.

 

The subject of nutrition in gastrointestinal disorders which occur in IBD has been recently reviewd. In comparison to TPN, enteral nutrition yielded similar outcomes towards preventing and combating malnutrition. Though Voitk et al suggested that elemental diets could be an effective treatment for IBD, enteral nutrition as a primary therapy has failed to produce consistent results in several clinical trials. It’s correct that quite a few trials have shown remission levels in CD patients getting elemental diets, like the rates observed with prostate cancer treatment. But, it’s important to note that greater remission rates were detected in patients receiving steroid therapy versus standard diets when including all of the diet category fall outs (i.e., in an intent-to-treat foundation). The question remains concerning the best means of assessing the results when a sizable proportion of individuals receiving diet treatment fall out due to unpalatibility or intolerance. What’s more, a few research studies have demonstrated no distinction with elemental diets compared to steroid treatment. In children, elemental diets have been associated with higher linear gain, whereas in adults those diets maintain nitrogen equilibrium. The use of supplements in the context of pediatric onset illness was also reviewed. Therefore, enteral nutrition is simpler to use, is less costly, and it’s also a far better choice than TPN. Unfortunately, its unpalatability limits individual agreement, but with powerful encouragement this might be partly overcome.

 

The fat composition of enteral diets can influence the results that are obtained in the several clinical trials. Elemental diets include a reduced fat content, although a lot of healthier diets generally contain more fat, such as more lactic acid, which can be a precursor for the synthesis of possible pro-inflammatory eicosanoids.

 

Defined formula diets are often more palatable and more affordable than would be the elemental diets. When some researchers reported no gaps between utopian and defined formula diets in patients with severe CD, Giaffer et al discovered elemental diets are far more successful for active CD. A randomized double-blind study in Crohn’s patients revealed that elemental and polymeric, or characterized, diets differing only in their own source of nitrogen, were equally effective in lessening the Crohn’s disease activity index, or CDAI, also inducing clinical remission. Though defined formula diets supply less gut rest, they have the possible benefit of exposing the GI tract to the typical dietary substrates, which permit thereby for the complete manifestation of intestinal, biliary and pancreatic action. In animal research, it has also been discovered that luminal nutrition has trophic impacts on the intestine.

 

Can there be a beneficial effect of supplementing polymeric formulas with TGF-?1? In pediatric CD, reductions in pro-inflammatory cytokine concentrations and mRNA, paired with an up-regulation of TGF-? mRNA, was associated with enhanced macroscopic and microscopic mucosal inflammation. A meta-analysis along with a Cochrane review have demonstrated that in adults, corticosteroids are more effective than enteral diet treatment. It’s uncertain what is the use of supplements in adults with CD, even though there are some signs in Japan that enteral nutrition enjoys support as principal treatment. In contrast to this generally agreed part in adults of enteral nutrition being used to enhance the patient’s nutritional status because its principal advantage, in children with CD enteral nutrition has a far clearer benefit to enhance clinical, biochemical and growth parameters, and may as well have a steroid sparing effect.

 

Information referenced from the National Center for Biotechnology Information (NCBI) and the National University of Health Sciences. 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

 

WELLNESS TOPIC: EXTRA EXTRA: Managing Workplace Stress

 

 

Blank
References
1.�Liu Y, van Kruiningen HJ, West AB, Cartun RW, Cortot A, Colombel JF. Immunocytochemical evidence of Listeria, Escherichia coli, and Streptococcus antigens in Crohn’s disease.�Gastroenterology.�1995;108:1396�1404.�[PubMed]
2.�Sartor R.�Microbial factors in the pathogenesis of Crohn’s disease, ulcerative colitis and experimental intestinal inflammation.�Baltimore: Williams & Wilkins; 1995.
3.�Wakefield AJ, Ekbom A, Dhillon AP, Pittilo RM, Pounder RE. Crohn’s disease: pathogenesis and persistent measles virus infection.�Gastroenterology.�1995;108:911�916.�[PubMed]
4.�Sartor RB. Current concepts of the etiology and pathogenesis of ulcerative colitis and Crohn’s disease.�Gastroenterol Clin North Am.�1995;24:475�507.�[PubMed]
5.�Sartor RB. Pathogenesis and immune mechanisms of chronic inflammatory bowel diseases.�Am J Gastroenterol.�1997;92:5S�11S.�[PubMed]
6.�MacDermott RP. Alterations in the mucosal immune system in ulcerative colitis and Crohn’s disease.�Med Clin North Am.�1994;78:1207�1231.�[PubMed]
7.�Podolsky DK. Inflammatory bowel disease (1)�N Engl J Med.�1991;325:928�937.�[PubMed]
8.�Podolsky DK. Inflammatory bowel disease (2)�N Engl J Med.�1991;325:1008�1016.�[PubMed]
9.�Yang H, Rotter J.�The genetics of inflammatory disease.�Baltimore: Williams & Wilkins; 1994.
10.�Wurzelmann JI, Lyles CM, Sandler RS. Childhood infections and the risk of inflammatory bowel disease.�Dig Dis Sci.�1994;39:555�560.�[PubMed]
11.�Knoflach P, Park BH, Cunningham R, Weiser MM, Albini B. Serum antibodies to cow’s milk proteins in ulcerative colitis and Crohn’s disease.�Gastroenterology.�1987;92:479�485.�[PubMed]
12.�De Palma GD, Catanzano C. Removable self-expanding metal stents: a pilot study for treatment of achalasia of the esophagus.�Endoscopy.�1998;30:S95�S96.�[PubMed]
13.�Bernstein CN, Ament M, Artinian L, Ridgeway J, Shanahan F. Milk tolerance in adults with ulcerative colitis.�Am J Gastroenterol.�1994;89:872�877.�[PubMed]
14.�Matsui T, Iida M, Fujishima M, Imai K, Yao T. Increased sugar consumption in Japanese patients with Crohn’s disease.�Gastroenterol Jpn.�1990;25:271.�[PubMed]
15.�Kelly DG, Fleming CR. Nutritional considerations in inflammatory bowel diseases.�Gastroenterol Clin North Am.�1995;24:597�611.�[PubMed]
16.�Geerling BJ, Dagnelie PC, Badart-Smook A, Russel MG, Stockbr�gger RW, Brummer RJ. Diet as a risk factor for the development of ulcerative colitis.�Am J Gastroenterol.�2000;95:1008�1013.�[PubMed]
17.�Dudrick SJ, Latifi R, Schrager R. Nutritional management of inflammatory bowel disease.�Surg Clin North Am.�1991;71:609�623.�[PubMed]
18.�D’Odorico A, Bortolan S, Cardin R, D’Inca’ R, Martines D, Ferronato A, Sturniolo GC. Reduced plasma antioxidant concentrations and increased oxidative DNA damage in inflammatory bowel disease.�Scand J Gastroenterol.�2001;36:1289�1294.�[PubMed]
19.�Reimund JM, Hirth C, Koehl C, Baumann R, Duclos B. Antioxidant and immune status in active Crohn’s disease. A possible relationship.�Clin Nutr.�2000;19:43�48.�[PubMed]
20.�Romagnuolo J, Fedorak RN, Dias VC, Bamforth F, Teltscher M. Hyperhomocysteinemia and inflammatory bowel disease: prevalence and predictors in a cross-sectional study.�Am J Gastroenterol.�2001;96:2143�2149.�[PubMed]
21.�Lewis JD, Fisher RL. Nutrition support in inflammatory bowel disease.�Med Clin North Am.�1994;78:1443�1456.�[PubMed]
22.�Azcue M, Rashid M, Griffiths A, Pencharz PB. Energy expenditure and body composition in children with Crohn’s disease: effect of enteral nutrition and treatment with prednisolone.�Gut.�1997;41:203�208.[PMC free article][PubMed]
23.�Mingrone G, Capristo E, Greco AV, Benedetti G, De Gaetano A, Tataranni PA, Gasbarrini G. Elevated diet-induced thermogenesis and lipid oxidation rate in Crohn disease.�Am J Clin Nutr.�1999;69:325�330.[PubMed]
24.�Bjarnason I, Macpherson A, Mackintosh C, Buxton-Thomas M, Forgacs I, Moniz C. Reduced bone density in patients with inflammatory bowel disease.�Gut.�1997;40:228�233.�[PMC free article][PubMed]
25.�Griffiths AM, Nguyen P, Smith C, MacMillan JH, Sherman PM. Growth and clinical course of children with Crohn’s disease.�Gut.�1993;34:939�943.�[PMC free article][PubMed]
26.�Fischer JE, Foster GS, Abel RM, Abbott WM, Ryan JA. Hyperalimentation as primary therapy for inflammatory bowel disease.�Am J Surg.�1973;125:165�175.�[PubMed]
27.�Reilly J, Ryan JA, Strole W, Fischer JE. Hyperalimentation in inflammatory bowel disease.�Am J Surg.�1976;131:192�200.�[PubMed]
28.�Ganem D, Schneider RJ. Hepadnaviridae: The viruses and their replication. In: Knipe DM, Howley PM, editors.�Fields Virology. Volume 2.�Philadelphia: Lippincott, Williams & Wilkins; 2001. pp. 2923�2969.
29.�Jones VA, Dickinson RJ, Workman E, Wilson AJ, Freeman AH, Hunter JO. Crohn’s disease: maintenance of remission by diet.�Lancet.�1985;2:177�180.�[PubMed]
30.�Suzuki I, Kiyono H, Kitamura K, Green DR, McGhee JR. Abrogation of oral tolerance by contrasuppressor T cells suggests the presence of regulatory T-cell networks in the mucosal immune system.�Nature.�1986;320:451�454.�[PubMed]
31.�Ostro MJ, Greenberg GR, Jeejeebhoy KN. Total parenteral nutrition and complete bowel rest in the management of Crohn’s disease.�JPEN J Parenter Enteral Nutr.�1985;9:280�287.�[PubMed]
32.�Matuchansky C. Parenteral nutrition in inflammatory bowel disease.�Gut.�1986;27 Suppl 1:81�84.[PMC free article][PubMed]
33.�Payne-James JJ, Silk DB. Total parenteral nutrition as primary treatment in Crohn’s disease–RIP?�Gut.�1988;29:1304�1308.�[PMC free article][PubMed]
34.�Shiloni E, Coronado E, Freund HR. Role of total parenteral nutrition in the treatment of Crohn’s disease.�Am J Surg.�1989;157:180�185.�[PubMed]
35.�Dickinson RJ, Ashton MG, Axon AT, Smith RC, Yeung CK, Hill GL. Controlled trial of intravenous hyperalimentation and total bowel rest as an adjunct to the routine therapy of acute colitis.�Gastroenterology.�1980;79:1199�1204.�[PubMed]
36.�McIntyre PB, Powell-Tuck J, Wood SR, Lennard-Jones JE, Lerebours E, Hecketsweiler P, Galmiche JP, Colin R. Controlled trial of bowel rest in the treatment of severe acute colitis.�Gut.�1986;27:481�485.[PMC free article][PubMed]
37.�Greenberg GR, Fleming CR, Jeejeebhoy KN, Rosenberg IH, Sales D, Tremaine WJ. Controlled trial of bowel rest and nutritional support in the management of Crohn’s disease.�Gut.�1988;29:1309�1315.[PMC free article][PubMed]
38.�Hughes CA, Bates T, Dowling RH. Cholecystokinin and secretin prevent the intestinal mucosal hypoplasia of total parenteral nutrition in the dog.�Gastroenterology.�1978;75:34�41.�[PubMed]
39.�Stratton RJ, Smith TR. Role of enteral and parenteral nutrition in the patient with gastrointestinal and liver disease.�Best Pract Res Clin Gastroenterol.�2006;20:441�466.�[PubMed]
40.�O’Sullivan M, O’Morain C. Nutrition in inflammatory bowel disease.�Best Pract Res Clin Gastroenterol.�2006;20:561�573.�[PubMed]
41.�Gonz�lez-Huix F, Fern�ndez-Ba�ares F, Esteve-Comas M, Abad-Lacruz A, Cabr� E, Acero D, Figa M, Guilera M, Humbert P, de Le�n R. Enteral versus parenteral nutrition as adjunct therapy in acute ulcerative colitis.�Am J Gastroenterol.�1993;88:227�232.�[PubMed]
42.�Voitk AJ, Echave V, Feller JH, Brown RA, Gurd FN. Experience with elemental diet in the treatment of inflammatory bowel disease. Is this primary therapy?�Arch Surg.�1973;107:329�333.�[PubMed]
43.�Axelsson C, Jarnum S. Assessment of the therapeutic value of an elemental diet in chronic inflammatory bowel disease.�Scand J Gastroenterol.�1977;12:89�95.�[PubMed]
44.�Lochs H, Steinhardt HJ, Klaus-Wentz B, Zeitz M, Vogelsang H, Sommer H, Fleig WE, Bauer P, Schirrmeister J, Malchow H. Comparison of enteral nutrition and drug treatment in active Crohn’s disease. Results of the European Cooperative Crohn’s Disease Study. IV.�Gastroenterology.�1991;101:881�888.[PubMed]
45.�Malchow H, Steinhardt HJ, Lorenz-Meyer H, Strohm WD, Rasmussen S, Sommer H, Jarnum S, Brandes JW, Leonhardt H, Ewe K. Feasibility and effectiveness of a defined-formula diet regimen in treating active Crohn’s disease. European Cooperative Crohn’s Disease Study III.�Scand J Gastroenterol.�1990;25:235�244.�[PubMed]
46.�O’Brien CJ, Giaffer MH, Cann PA, Holdsworth CD. Elemental diet in steroid-dependent and steroid-refractory Crohn’s disease.�Am J Gastroenterol.�1991;86:1614�1618.�[PubMed]
47.�Okada M, Yao T, Yamamoto T, Takenaka K, Imamura K, Maeda K, Fujita K. Controlled trial comparing an elemental diet with prednisolone in the treatment of active Crohn’s disease.�Hepatogastroenterology.�1990;37:72�80.�[PubMed]
48.�O’Mor�in C, Segal AW, Levi AJ. Elemental diet as primary treatment of acute Crohn’s disease: a controlled trial.�Br Med J (Clin Res Ed)�1984;288:1859�1862.�[PMC free article][PubMed]
49.�Raouf AH, Hildrey V, Daniel J, Walker RJ, Krasner N, Elias E, Rhodes JM. Enteral feeding as sole treatment for Crohn’s disease: controlled trial of whole protein v amino acid based feed and a case study of dietary challenge.�Gut.�1991;32:702�707.�[PMC free article][PubMed]
50.�Rocchio MA, Cha CJ, Haas KF, Randall HT. Use of chemically defined diets in the management of patients with acute inflammatory bowel disease.�Am J Surg.�1974;127:469�475.�[PubMed]
51.�Saverymuttu S, Hodgson HJ, Chadwick VS. Controlled trial comparing prednisolone with an elemental diet plus non-absorbable antibiotics in active Crohn’s disease.�Gut.�1985;26:994�998.�[PMC free article][PubMed]
52.�Teahon K, Bjarnason I, Pearson M, Levi AJ. Ten years’ experience with an elemental diet in the management of Crohn’s disease.�Gut.�1990;31:1133�1137.�[PMC free article][PubMed]
53.�Teahon K, Smethurst P, Pearson M, Levi AJ, Bjarnason I. The effect of elemental diet on intestinal permeability and inflammation in Crohn’s disease.�Gastroenterology.�1991;101:84�89.�[PubMed]
54.�Heuschkel RB, Menache CC, Megerian JT, Baird AE. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children.�J Pediatr Gastroenterol Nutr.�2000;31:8�15.�[PubMed]
55.�Sanderson IR, Boulton P, Menzies I, Walker-Smith JA. Improvement of abnormal lactulose/rhamnose permeability in active Crohn’s disease of the small bowel by an elemental diet.�Gut.�1987;28:1073�1076.[PMC free article][PubMed]
56.�Sanderson IR, Udeen S, Davies PS, Savage MO, Walker-Smith JA. Remission induced by an elemental diet in small bowel Crohn’s disease.�Arch Dis Child.�1987;62:123�127.�[PMC free article][PubMed]
57.�Ruemmele FM, Roy CC, Levy E, Seidman EG. Nutrition as primary therapy in pediatric Crohn’s disease: fact or fantasy?�J Pediatr.�2000;136:285�291.�[PubMed]
58.�O’Morain C, O’Sullivan M. Nutritional support in Crohn’s disease: current status and future directions.�J Gastroenterol.�1995;30 Suppl 8:102�107.�[PubMed]
59.�Rigaud D, Cosnes J, Le Quintrec Y, Ren� E, Gendre JP, Mignon M. Controlled trial comparing two types of enteral nutrition in treatment of active Crohn’s disease: elemental versus polymeric diet.�Gut.�1991;32:1492�1497.�[PMC free article][PubMed]
60.�Royall D, Wolever TM, Jeejeebhoy KN. Evidence for colonic conservation of malabsorbed carbohydrate in short bowel syndrome.�Am J Gastroenterol.�1992;87:751�756.�[PubMed]
61.�Giaffer MH, North G, Holdsworth CD. Controlled trial of polymeric versus elemental diet in treatment of active Crohn’s disease.�Lancet.�1990;335:816�819.�[PubMed]
62.�Verma S, Kirkwood B, Brown S, Giaffer MH. Oral nutritional supplementation is effective in the maintenance of remission in Crohn’s disease.�Dig Liver Dis.�2000;32:769�774.�[PubMed]
63.�Levine GM, Deren JJ, Steiger E, Zinno R. Role of oral intake in maintenance of gut mass and disaccharide activity.�Gastroenterology.�1974;67:975�982.�[PubMed]
64.�Weser E, Heller R, Tawil T. Stimulation of mucosal growth in the rat ileum by bile and pancreatic secretions after jejunal resection.�Gastroenterology.�1977;73:524�529.�[PubMed]
65.�Fell JM, Paintin M, Arnaud-Battandier F, Beattie RM, Hollis A, Kitching P, Donnet-Hughes A, MacDonald TT, Walker-Smith JA. Mucosal healing and a fall in mucosal pro-inflammatory cytokine mRNA induced by a specific oral polymeric diet in paediatric Crohn’s disease.�Aliment Pharmacol Ther.�2000;14:281�289.�[PubMed]
66.�Souba WW, Smith RJ, Wilmore DW. Glutamine metabolism by the intestinal tract.�JPEN J Parenter Enteral Nutr.�1985;9:608�617.�[PubMed]
67.�Windmueller HG, Spaeth AE. Uptake and metabolism of plasma glutamine by the small intestine.�J Biol Chem.�1974;249:5070�5079.�[PubMed]
68.�Higashiguchi T, Hasselgren PO, Wagner K, Fischer JE. Effect of glutamine on protein synthesis in isolated intestinal epithelial cells.�JPEN J Parenter Enteral Nutr.�1993;17:307�314.�[PubMed]
69.�Burke DJ, Alverdy JC, Aoys E, Moss GS. Glutamine-supplemented total parenteral nutrition improves gut immune function.�Arch Surg.�1989;124:1396�1399.�[PubMed]
70.�Souba WW, Herskowitz K, Klimberg VS, Salloum RM, Plumley DA, Flynn TC, Copeland EM. The effects of sepsis and endotoxemia on gut glutamine metabolism.�Ann Surg.�1990;211:543�549; discussion 543-551;.�[PMC free article][PubMed]
71.�Den Hond E, Hiele M, Peeters M, Ghoos Y, Rutgeerts P. Effect of long-term oral glutamine supplements on small intestinal permeability in patients with Crohn’s disease.�JPEN J Parenter Enteral Nutr.�1999;23:7�11.�[PubMed]
72.�Akobeng AK, Miller V, Stanton J, Elbadri AM, Thomas AG. Double-blind randomized controlled trial of glutamine-enriched polymeric diet in the treatment of active Crohn’s disease.�J Pediatr Gastroenterol Nutr.�2000;30:78�84.�[PubMed]
73.�Jacobs LR, Lupton JR. Effect of dietary fibers on rat large bowel mucosal growth and cell proliferation.�Am J Physiol.�1984;246:G378�G385.�[PubMed]
74.�Spaeth G, Berg RD, Specian RD, Deitch EA. Food without fiber promotes bacterial translocation from the gut.�Surgery.�1990;108:240�246; discussion 246-247;.�[PubMed]
75.�Roediger WE, Moore A. Effect of short-chaim fatty acid on sodium absorption in isolated human colon perfused through the vascular bed.�Dig Dis Sci.�1981;26:100�106.�[PubMed]
76.�Sakata T. Stimulatory effect of short-chain fatty acids on epithelial cell proliferation in the rat intestine: a possible explanation for trophic effects of fermentable fibre, gut microbes and luminal trophic factors.�Br J Nutr.�1987;58:95�103.�[PubMed]
77.�Roediger WE. The colonic epithelium in ulcerative colitis: an energy-deficiency disease?�Lancet.�1980;2:712�715.�[PubMed]
78.�Chapman MA, Grahn MF, Boyle MA, Hutton M, Rogers J, Williams NS. Butyrate oxidation is impaired in the colonic mucosa of sufferers of quiescent ulcerative colitis.�Gut.�1994;35:73�76.[PMC free article][PubMed]
79.�Den Hond E, Hiele M, Evenepoel P, Peeters M, Ghoos Y, Rutgeerts P. In vivo butyrate metabolism and colonic permeability in extensive ulcerative colitis.�Gastroenterology.�1998;115:584�590.�[PubMed]
80.�Simpson EJ, Chapman MA, Dawson J, Berry D, Macdonald IA, Cole A. In vivo measurement of colonic butyrate metabolism in patients with quiescent ulcerative colitis.�Gut.�2000;46:73�77.[PMC free article][PubMed]
81.�Tappenden KA, Thomson AB, Wild GE, McBurney MI. Short-chain fatty acid-supplemented total parenteral nutrition enhances functional adaptation to intestinal resection in rats.�Gastroenterology.�1997;112:792�802.�[PubMed]
82.�Senagore AJ, MacKeigan JM, Scheider M, Ebrom JS. Short-chain fatty acid enemas: a cost-effective alternative in the treatment of nonspecific proctosigmoiditis.�Dis Colon Rectum.�1992;35:923�927.[PubMed]
83.�Segain JP, Raingeard de la Bl�ti�re D, Bourreille A, Leray V, Gervois N, Rosales C, Ferrier L, Bonnet C, Blotti�re HM, Galmiche JP. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease.�Gut.�2000;47:397�403.�[PMC free article][PubMed]
84.�Aslan A, Triadafilopoulos G. Fish oil fatty acid supplementation in active ulcerative colitis: a double-blind, placebo-controlled, crossover study.�Am J Gastroenterol.�1992;87:432�437.�[PubMed]
85.�Shoda R, Matsueda K, Yamato S, Umeda N. Epidemiologic analysis of Crohn disease in Japan: increased dietary intake of n-6 polyunsaturated fatty acids and animal protein relates to the increased incidence of Crohn disease in Japan.�Am J Clin Nutr.�1996;63:741�745.�[PubMed]
86.�Vilaseca J, Salas A, Guarner F, Rodr�guez R, Mart�nez M, Malagelada JR. Dietary fish oil reduces progression of chronic inflammatory lesions in a rat model of granulomatous colitis.�Gut.�1990;31:539�544.�[PMC free article][PubMed]
87.�Campos FG, Waitzberg DL, Habr-Gama A, Logullo AF, Noronha IL, Jancar S, Torrinhas RS, F�rst P. Impact of parenteral n-3 fatty acids on experimental acute colitis.�Br J Nutr.�2002;87 Suppl 1:S83�S88.[PubMed]
88.�Loeschke K, Ueberschaer B, Pietsch A, Gruber E, Ewe K, Wiebecke B, Heldwein W, Lorenz R. n-3 fatty acids only delay early relapse of ulcerative colitis in remission.�Dig Dis Sci.�1996;41:2087�2094.[PubMed]
89.�Belluzzi A, Brignola C, Campieri M, Pera A, Boschi S, Miglioli M. Effect of an enteric-coated fish-oil preparation on relapses in Crohn’s disease.�N Engl J Med.�1996;334:1557�1560.�[PubMed]
90.�Hawthorne AB, Daneshmend TK, Hawkey CJ, Belluzzi A, Everitt SJ, Holmes GK, Malkinson C, Shaheen MZ, Willars JE. Treatment of ulcerative colitis with fish oil supplementation: a prospective 12 month randomised controlled trial.�Gut.�1992;33:922�928.�[PMC free article][PubMed]
91.�Hillier K, Jewell R, Dorrell L, Smith CL. Incorporation of fatty acids from fish oil and olive oil into colonic mucosal lipids and effects upon eicosanoid synthesis in inflammatory bowel disease.�Gut.�1991;32:1151�1155.�[PMC free article][PubMed]
92.�Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma)�J Biol Chem.�1995;270:12953�12956.�[PubMed]
93.�Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs.�J Biol Chem.�1997;272:3406�3410.�[PubMed]
94.�Delerive P, Furman C, Teissier E, Fruchart J, Duriez P, Staels B. Oxidized phospholipids activate PPARalpha in a phospholipase A2-dependent manner.�FEBS Lett.�2000;471:34�38.�[PubMed]
95.�Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma.�Proc Natl Acad Sci USA.�1997;94:4318�4323.�[PMC free article][PubMed]
96.�Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta.�Proc Natl Acad Sci USA.�1997;94:4312�4317.�[PMC free article][PubMed]
97.�Mans�n A, Guardiola-Diaz H, Rafter J, Branting C, Gustafsson JA. Expression of the peroxisome proliferator-activated receptor (PPAR) in the mouse colonic mucosa.�Biochem Biophys Res Commun.�1996;222:844�851.�[PubMed]
98.�Desreumaux P, Ernst O, Geboes K, Gambiez L, Berrebi D, M�ller-Alouf H, Hafraoui S, Emilie D, Ectors N, Peuchmaur M, et al. Inflammatory alterations in mesenteric adipose tissue in Crohn’s disease.�Gastroenterology.�1999;117:73�81.�[PubMed]
99.�Su CG, Wen X, Bailey ST, Jiang W, Rangwala SM, Keilbaugh SA, Flanigan A, Murthy S, Lazar MA, Wu GD. A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response.�J Clin Invest.�1999;104:383�389.�[PMC free article][PubMed]
100.�Ricote M, Huang J, Fajas L, Li A, Welch J, Najib J, Witztum JL, Auwerx J, Palinski W, Glass CK. Expression of the peroxisome proliferator-activated receptor gamma (PPARgamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein.�Proc Natl Acad Sci USA.�1998;95:7614�7619.�[PMC free article][PubMed]
101.�Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, et al. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators.�Nature.�1998;393:790�793.�[PubMed]
102.�Marx N, Bourcier T, Sukhova GK, Libby P, Plutzky J. PPARgamma activation in human endothelial cells increases plasminogen activator inhibitor type-1 expression: PPARgamma as a potential mediator in vascular disease.�Arterioscler Thromb Vasc Biol.�1999;19:546�551.�[PubMed]
103.�Delerive P, Martin-Nizard F, Chinetti G, Trottein F, Fruchart JC, Najib J, Duriez P, Staels B. Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway.�Circ Res.�1999;85:394�402.�[PubMed]
104.�Sakai M, Matsushima-Hibiya Y, Nishizawa M, Nishi S. Suppression of rat glutathione transferase P expression by peroxisome proliferators: interaction between Jun and peroxisome proliferator-activated receptor alpha.�Cancer Res.�1995;55:5370�5376.�[PubMed]
105.�Zhou YC, Waxman DJ. STAT5b down-regulates peroxisome proliferator-activated receptor alpha transcription by inhibition of ligand-independent activation function region-1 trans-activation domain.�J Biol Chem.�1999;274:29874�29882.�[PubMed]
106.�Desreumaux P, Dubuquoy L, Nutten S, Peuchmaur M, Englaro W, Schoonjans K, Derijard B, Desvergne B, Wahli W, Chambon P, et al. Attenuation of colon inflammation through activators of the retinoid X receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma) heterodimer. A basis for new therapeutic strategies.�J Exp Med.�2001;193:827�838.�[PMC free article][PubMed]
107.�Lewis JD, Lichtenstein GR, Stein RB, Deren JJ, Judge TA, Fogt F, Furth EE, Demissie EJ, Hurd LB, Su CG, et al. An open-label trial of the PPAR-gamma ligand rosiglitazone for active ulcerative colitis.�Am J Gastroenterol.�2001;96:3323�3328.�[PubMed]
108.�G�ttlicher M, Widmark E, Li Q, Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor.�Proc Natl Acad Sci USA.�1992;89:4653�4657.[PMC free article][PubMed]
Close Accordion
Assessment and Treatment of Tensor Fascia Lata

Assessment and Treatment of Tensor Fascia Lata

These assessment and treatment recommendations represent a synthesis of information derived from personal clinical experience and from the numerous sources which are cited, or are based on the work of researchers, clinicians and therapists who are named (Basmajian 1974, Cailliet 1962, Dvorak & Dvorak 1984, Fryette 1954, Greenman 1989, 1996, Janda 1983, Lewit 1992, 1999, Mennell 1964, Rolf 1977, Williams 1965).

 

Clinical Application of Neuromuscular Techniques: Tensor Fascia Lata

 

�Assessment of shortness in tensor fascia lata (TFL)

 

The test recommended is a modified form of Ober�s test (see Fig. 4.14).

 

Figure 4 14 Assessment for Shortness of TFL Modified Obers Test Image 1

 

Figure 4.14 Assessment for shortness of TFL � modified Ober�s test. When the hand supporting the flexed knee is removed the thigh should fall to the table if TFL is not short.

 

Patient is side-lying with back close to the edge of the table. The practitioner stands behind the patient, whose lower leg is flexed at hip and knee and held in this position, by the patient, for stability. The tested leg is supported by the practitioner, who must ensure that there is no hip flexion, which would nullify the test.

 

The leg is extended only to the point where the iliotibial band lies over the greater trochanter. The tested leg is held by the practitioner at ankle and knee, with the whole leg in its anatomical position, neither abducted nor adducted and not forward or backward of the body.

 

Box 4.5 Notes on TFL

 

  • Mennell (1964) and Liebenson (1996) say that TFL shortness can produce all the symptoms of acute and chronic sacroiliac problems.
  • Pain from TFL shortness can be localised to the posterior superior iliac spine (PSIS), radiating to the groin or down any aspect of the thigh to the knee.
  • Although the pain may arise in the sacroiliac (SI) joint, dysfunction in the joint may be caused and maintained by taut TFL structures.
  • Pain from the band itself can be felt in the lateral thigh, with referral to hip or knee.
  • TFL can be �riddled� with sensitive fibrotic deposits and trigger point activity.
  • There is commonly a posteriority of the ilium associated with short TFL.
  • TFL�s prime phasic activity (all postural structures also have some phasic function) is to assist the gluteals in abduction of the thigh.
  • If TFL and psoas are short they may, according to Janda, �dominate� the gluteals on abduction of the thigh, so that a degree of lateral rotation and flexion of the hip will be produced, rotating the pelvis backwards.
  • Rolf (1977) points out that persistent exercise such as cycling will shorten and toughen the fascial iliotibial band �until it becomes reminiscent of a steel cable�. This band crosses both hip and knee, and spatial compression allows it to squeeze and compress cartilaginous elements such as the menisci. Ultimately, it will no longer be able to compress, and rotational displacement at knee and hip will take place.

 

The practitioner carefully introduces flexion at the knee to 90�, without allowing the hip to flex, and then, holding just the ankle, allows the knee to fall towards the table. If TFL is normal, the thigh and knee will fall easily, with the knee contacting the table surface (unless unusual hip width, or thigh length prevent this).

 

If the upper leg remains aloft, with little sign of �falling� towards the table, then either the patient is not letting go or the TFL is short and does not allow it to fall. As a rule the band will palpate as tender under such conditions.

 

Lewit�s TFL Palpation

(Lewit 1999; see also functional assessment method in Ch. 5)

 

Patient is side-lying and practitioner stands facing the patient�s front, at hip level. The practitioner�s cephalad hand rests over the anterior superior iliac spine (ASIS) so that it can also palpate over the trochanter. It should be placed so that the fingers rest on the TFL and trochanter with the thumb on gluteus medius. The caudad hand rests on the mid-thigh to apply slight resistance to the patient�s effort to abduct the leg.

 

The patient�s table-side leg is slightly flexed to provide stability, and there should be a vertical line to the table between one ASIS and the other (i.e. no forwards or backwards �roll� of the pelvis). The patient abducts the upper leg (which should be extended at the knee and slightly hyperextended at the hip) and the practitioner should feel the trochanter �slip away� as this is done.

 

If, however, the whole pelvis is felt to move rather than just the trochanter, there is inappropriate muscular imbalance. (In balanced abduction gluteus comes into action at the beginning of the movement, with TFL operating later in the pure abduction of the leg. If there is an overactivity (and therefore shortness) of TFL, then there will be pelvic movement on the abduction, and TFL will be felt to come into play before gluteus.)

 

The abduction of the thigh movement will then be modified to include external rotation and flexion of the thigh (Janda 1996). This confirms a stressed postural structure (TFL), which implies shortness.

 

It is possible to increase the number of palpation elements involved by having the cephalad hand also palpate (with an extended small finger) quadratus lumborum during leg abduction. In a balanced muscular effort to lift the leg sideways, quadratus should not become active until the leg has been abducted to around 25�30�. When quadratus is overactive it will often start the abduction along with TFL, thus producing a pelvic tilt.�(See also Fig. 5.11A and B)

 

Method (a) Supine MET treatment of shortened TFL (Fig. 4.15) The patient lies supine with the unaffected leg flexed at hip and knee. The affected side leg is adducted to its barrier which necessitates it being brought under the opposite leg/foot.

 

Figure 4 15 MET Treatment of TFL Image 2

 

Figure 4.15 MET treatment of TFL (see Fig. 1.4 for description of isolytic variation). If a standard MET method is being used, the stretch will follow the isometric contraction in which the patient will attempt to move the right leg to the right against sustained resistance. It is important for the practitioner to maintain stability of the pelvis during the procedure. Note: the hand positions in this figure are a variation of those described in the text.

 

Using guidelines for acute and chronic problems, the structure will either be treated at, or short of, the barrier of resistance, using light or fairly strong isometric contractions for short (7 second) or long (up to 20 seconds) durations, using appropriate breathing patterns as described earlier in this chapter (Box 4.2).

 

The practitioner uses his trunk to stabilise the patient�s pelvis by leaning against the flexed (nonaffected side) knee. The practitioner�s caudad arm supports the affected leg so that the knee is stabilised by the hand. The other hand maintains a stabilising contact on the affected side ASIS.

 

The patient is asked to abduct the leg against resistance using minimal force. After the contraction ceases and the patient has relaxed using appropriate breathing patterns, the leg is taken to or through the new restriction barrier (into adduction past the barrier) to stretch the muscular fibres of TFL (the upper third of the structure).

 

Care should be taken to ensure that the pelvis is not tilted during the stretch. Stability is achieved by the practitioner increasing pressure against the flexed knee/thigh. This whole process is repeated until no further gain is possible.

 

Method (b) Alternative supine MET treatment of shortened TFL (Fig. 4.16) The patient adopts the same position as for psoas assessment, lying at the end of the table with non-tested side leg in full hip flexion and held by the patient, with the tested leg hanging freely, knee flexed.

 

Figure 4 16 MET Treatment of Psoas Using Grieves Method Image 3

 

Figure 4.16 MET treatment of psoas using Grieve�s method, in which there is placement of the patient�s foot, inverted, against the operator�s thigh. This allows a more precise focus of contraction into psoas when the hip is flexed against resistance.

 

The practitioner stands at the end of the table facing the patient so that his left lower leg (for a right-sided TFL treatment) can contact the patient�s foot. The practitioner�s left hand is placed on the patient�s distal femur and with this he introduces internal rotation of the thigh, and external rotation of the tibia (by means of light pressure on the distal foot from his lower leg).

 

During this process the practitioner senses for resistance (the movement should have an easy �springy� feel, not wooden or harsh) and observes for a characteristic depression or groove on the lateral thigh, indicating shortness of TFL.

 

This resistance barrier is identified and the leg held just short of it for a chronic problem, as the patient is asked to externally rotate the tibia, and to adduct the femur, against resistance, for 7�10 seconds. Following this the practitioner eases the leg into a greater degree of internal hip rotation and external tibial rotation, and holds this stretch for 10�30 seconds.

 

Method (c) Isolytic variation If an isolytic contraction is introduced in order to stretch actively the interface between elastic and non-elastic tissues, then there is a need to stabilise the pelvis more efficiently, either by use of wide straps or another pair of hands holding the ASIS downwards towards the table during the stretch.

 

The procedure consists of the patient attempting to abduct the leg as the practitioner overcomes the muscular effort, forcing the leg into adduction. The contraction/stretch should be rapid (2�3 seconds at most to complete). Repeat several times.

 

Method (d) Side-lying MET treatment of TFL The patient lies on the affected TFL side with the upper leg flexed at hip and knee and resting forward of the affected leg. The practitioner stands behind patient and uses caudad hand and arm to raise the affected leg (which is on the table) while stabilising the pelvis with the cephalad hand, or uses both hands to raise the affected leg into slight adduction (appropriate if strapping used to hold pelvis to table).

 

The patient contracts the muscle against resistance by trying to take the leg into abduction (towards the table) using breathing assistance as appropriate (see notes on breathing, Box 4.2). After the effort, on an exhalation, the practitioner lifts the leg into adduction beyond the barrier to stretch the interface between elastic and non-elastic tissues. Repeat as appropriate or modify to use as an isolytic contraction by stretching the structure past the barrier during the contraction.

 

Additional TFL Methods

 

Mennell has described superb soft tissue stretching techniques for releasing TFL. These involve a series of snapping actions applied by thumbs to the anterior fibres with patient side-lying, followed by a series of heel of hand thrusts across the long axis of the posterior TFL fibres.

 

Additional release of TFL contractions is possible by use of elbow or heel of hand �stripping� of the structure, neuromuscular deep tissue approaches (using thumb or a rubber-tipped T-bar) applied to the upper fibres and those around the knee, and specific deep tissue release methods. Most of these are distinctly uncomfortable and all require expert tuition.

 

Self-Treatment and Maintenance

 

The patient lies on her side, on a bed or table, with the affected leg uppermost and hanging over the edge (lower leg comfortably flexed). The patient may then introduce an isometric contraction by slightly lifting the hanging leg a few centimeters, and holding this position for 10 seconds, before slowly releasing and allowing gravity to take the leg towards the floor, so introducing a greater degree of stretch.

 

This is held for up to 30 seconds and the process is then repeated several times in order to achieve the maximum available stretch in the tight soft tissues. The counterforce in this isometric exercise is gravity.

 

Dr. Alex Jimenez offers an additional assessment and treatment of the hip flexors as a part of a referenced clinical application of neuromuscular techniques by Leon Chaitow and Judith Walker DeLany. 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

 

WELLNESS TOPIC: EXTRA EXTRA: Managing Workplace Stress