Gluten, Inflammation, and Neurodegeneration (2024)

  • Journal List
  • Am J Lifestyle Med
  • v.16(1); Jan-Feb 2022
  • PMC8848113

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Gluten, Inflammation, and Neurodegeneration (1)

Link to Publisher's site

Am J Lifestyle Med. 2022 Jan-Feb; 16(1): 32–35.

Published online 2022 Jan 11. doi:10.1177/15598276211049345

PMCID: PMC8848113

PMID: 35185424

Ashok Philip, PhDGluten, Inflammation, and Neurodegeneration (2)

Director of Assessment and Professor of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, AR, USA (AP); Department of Pharmacy Practice, School of Pharmacy and Health Professions. Creighton University, Omaha, NE, USA (NDW)

Author information Copyright and License information PMC Disclaimer

Abstract

Growing evidence supports a potential link between dietary gluten intake and neurodegenerative disease in susceptible populations. Observational data supporting this link are described along with interventional study data evaluating the effects of restricting gluten from the diet in patients with neurologic disorders. Suggested underlying mechanisms between gluten intake and neurodegeneration are discussed.

Keywords: gluten, inflammation, neurodegeneration, neurologic disorders, celiac disease, Nonceliac gluten sensitivity

“...dietary gluten-induced chronic inflammation is linked to dysbiosis and a permeable gut...”

Gluten plays an established role in the pathogenesis of celiac disease (CD) and nonceliac gluten sensitivity (NCGS).1 Interestingly, a number of neurodegenerative conditions have been associated with grain intake or respond favorably when gluten is restricted from the diet.2-4 Additionally, rates of comorbid CD/NCGS and neurodegenerative disorders, including bipolar, major depressive disorder, anxiety, and autism spectrum disorders (ASD), have been observed in unusually high rates.5-8 These findings raise the question as to whether gluten may be implicated in the pathogenesis of neurodegenerative conditions.

Gluten, primarily comprising gliadin and glutenin proteins (8085%), is a member of the prolamin superfamily and is commonly found in wheat, barley, rye, and oats. The prolamin superfamily of proteins is characterized by a repetitive sequence of the amino acids glutamine and proline and is suspected to be responsible for gluten’s water insolubility. Gluten within grains is confined to the cells of the starchy endosperm where it supports germination. In food production, gluten proteins are valued for their cohesiveness and viscoelasticity, which allows for ideal dough and bread consistency.9,10 As a result, gluten is highly prevalent in a variety of common foods (bread, pasta, bakery items, and cereal) as well as used for sauce thickeners, fillers, and stabilizers.

Gluten consumed through the diet has been linked to a pathological immune response in susceptible populations.10-13 Upon consumption, gluten is partially hydrolyzed by proteases in the gastrointestinal tract to peptides of ∼1030 amino acids in length that cross the intestinal barrier via trans or paracellular transport. These peptides undergo deamidation by intestinal tissue transglutaminase-2 (tTG2) which increases their affinity for major histocompatibility complex II (MHC II) molecules and triggers an inflammatory response. In individuals with celiac disease (CD) who carry human leukocyte antigen class II with DQ2 and/or DQ8 molecules on antigen-presenting cells (HLA-DQ2 or DQ8 haplotype), these partially digested peptides are recognized by the DQ cell receptor and presented to T lymphocytes, resulting in an immune response. In patients with nonceliac gluten sensitivity (NCGS), who are DQ2 and DQ8 negative, it is suspected that gluten proteins in concert with other components of wheat activate an innate immune response in the body. Wheat germ agglutinin and amylase-trypsin inhibitors of wheat have also been shown to illicit an innate immune response.10-13 As a result, due to loss of tolerance to gluten proteins, individuals with CD and NCGS experience gastrointestinal distress, fatigue, and pain including inflammation and increased permeability of the intestinal mucosa.1

These inflammatory effects of gluten may not be limited to the gastrointestinal system. Increased intestinal permeability leads to entry of toxic digestive metabolites, bacteria, and bacterial toxins into the bloodstream which may eventually reach the central nervous system.1 Neurological issues like cerebellar ataxia, peripheral neuropathy, cognitive impairment, and neuropsychiatric diseases have been associated with CD, suggesting the possibility of gluten-mediated inflammation playing a role in loss of bloodbrain barrier (BBB) integrity.1,14-16 Furthermore, increased BBB permeability attributed to inflammation or bacteria is linked to other neurological disorders including ASD, dementia, Alzheimer’s disease, Parkinson’s disease (PD), depression, anxiety, and schizophrenia.15,17 The following describes observed relationships between gluten and neurodegenerative conditions as well as potential mechanisms by which dietary gluten may lead to neurodegeneration.

Gluten and Neurodegenerative Conditions

Despite the paucity of evidence establishing a causal relationship between dietary gluten intake and neurological disorders, a number of studies demonstrate an association between the 2 and/or a beneficial effect on neurological symptoms when gluten is restricted from the diet. One example is the reduction in observed incidence of schizophrenia in populations who consume little to no grains compared to those consuming grain-rich diets.2 Patients with schizophrenia have markedly high levels of inflammation, and a gluten-free diet (GFD) has been shown to improve both psychiatric and gastrointestinal symptoms in these patients.3,18,19 Likewise, GFD has been associated with improvements in ASD.4,5,20,21 Unusually high rates of chronic diarrhea, constipation, and bloating have been observed in children with ASD.5 Interventional studies have demonstrated improvements in both the prevalence of gastrointestinal symptoms and ASD behaviors in children with ASD who followed GFD.4 Mood disorders (bipolar disorder, major depressive disorder, and anxiety) are commonly observed in individuals with CD and NCGS, with bipolar disorder 17 times more likely to affect individuals with CD relative to the general population.6,7 Increased levels of gluten-related antibodies are found in individuals with bipolar disorder, depression, anxiety, hyperactivity, as well as schizophrenia suggesting the correlation between gluten consumption and mood disorders.6-8

Putative Mechanisms

As stated earlier, intestinal tissue transglutaminase-2 (tTG2) is a calcium-dependent enzyme that mediates deamidation of glutamine residues present in gluten proteins, triggering gluten’s inflammatory response.16,22 tTG2 not only mediates the post-translational modification/deamidation of the gluten protein gliadin, but was shown to act as an autoantigen and due to its ability to induce the production of tTG2 autoantibodies.22,23 The presence of tTG2 autoantibodies were detected in almost 100% of patients diagnosed with CD and in a minor fraction of NCGS patients.23 Tissue transglutaminase is also involved in multiple neurodegenerative diseases. The neural isoform of tissue transglutaminase, tTG6, is implicated in the pathogenesis of Alzheimer’s disease (AD), Huntington’s disease (HD), and movement disorders like gluten ataxia and multiple sclerosis.24-28 Increased levels of circulating anti-tTG6 antibodies are also present in adult patients with schizophrenia.16,29

Overall, increased levels of tTG6 antibodies in CD patients with gluten ataxia and peripheral neuropathy suggest a plausible role for tTG6 in contributing toward these central and peripheral symptoms of CD.26,29 With regards to NCGS patients, it is suggested that loss of integrity of the intestinal barrier facilitates access of partially hydrolyzed immunogenic gluten peptides to both the systemic circulation and the brain. Access of these toxic gluten peptides and HLA-DQ2/DQ8 restricted CD4 T cells to the brain may contribute to the neurological issues observed in NCGS patients.30-33 It is also proposed that loss of intestinal barrier’s integrity aka leaky gut promotes increased access for circulating or gut-derived extracellular structures like biomolecular condensates (BMC) and extracellular vesicles (EV) to the brain of CD and NCGS patients.16 Given the critical role of tTG in mediating gluten metabolism, its autoantigen properties, its ability to cause central β-amyloid accumulation, and foster inflammation and cancer, tTG is being pursued as a molecular target for a variety of disease states.16,34

Additional mechanisms have been postulated to contribute to gluten sensitivity and neuronal dysfunction. One of them is the identification of a strong correlation between specific HLA haplotypes and dysregulation of the gut-brain axis in CD and NCGS patients diagnosed with ASD and/or Down’s syndrome (DS).16,35,36 Also, in NCGS patients with neurological issues, antibodies for glutamic acid decarboxylase (GAD), a key enzyme involved in the biosynthesis of the primary inhibitory neurotransmitter gamma-aminobutyric acid, were detected.16,37

The potential role of epigenetic mechanisms in regulating inflammatory responses has prompted focused attention toward micro-RNAs (MiRNAs) and CD.16 The MiRNAs are ∼20–23 nucleotides in length, small RNA molecules that mediate a number of cellular processes like cell proliferation, differentiation, apoptosis, cell signaling, and immune, including inflammatory, responses.16 Specifically, downregulation of miR-192-5p due to inflammation is suggestive of its role in maintaining intestinal homeostasis.16,38,39 Moreover, chronic inflammation of the intestine combined with a leaky gut results in gut dysbiosis, which may in turn promote microbial entry into systemic circulation. Especially, loss of intestinal barrier’s integrity has the potential to allow increased access of lipopolysaccharides (LPS) across the BBB, activate microglial cells, and ultimately cause neuronal inflammation and damage in CD patients.16,40

In CD and NCGS individuals, it is established that dietary gluten-induced chronic inflammation is linked to dysbiosis and a permeable gut.16 This in turn affects factors that regulate neuronal inflammation, cognition, and neurodegeneration. For example, at the molecular level, the genetic expression of peroxisome proliferator activated receptor gamma (PPARγ) is reduced in patients diagnosed with ulcerative colitis and CD. Because PPARγ is an anti-inflammatory and anti-dysbiotic molecule, it may serve as a potential molecular target for the management of gluten-induced intestinal inflammation and dysbiosis.16,41

Conclusion

While data supporting the direct link between gluten consumption and neurodegeneration is limited, available evidence highlights the relationship between gluten consumption, tTG antibody production, and disruption of the microbiota-gut-brain axis.1,16 The beneficial effects of GFD in patients with CD, schizophrenia, and ASD, especially in mitigating gastrointestinal and neurological symptoms, highlight the need for more exploration and identification of definitive evidence.42

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

1. Obrenovich M. Leaky gut, leaky brain?Microorganisms. 2018;6:107. [PMC free article] [PubMed] [Google Scholar]

2. Dohan FC, Harper EH, Clark MH, Rodrigue RB, Zigas V. Is schizophrenia rare if grain is rare?Biol Psychiatr. 1984;19:385-399. [PubMed] [Google Scholar]

3. Kelly DL, Demyanovich HK, Rodriguez KM, et al. Randomized controlled trial of a gluten-free diet in patients with schizophrenia positive for antigliadin antibodies (AGA IgG): a pilot feasibility study. J Psychiatr Neurosci. 2019;44(4):269-276. [PMC free article] [PubMed] [Google Scholar]

4. Ghalichi F, Ghaemmaghami J, Malek A, Ostadrahimi A. Effect of gluten free diet on gastrointestinal and behavioral indices for children with autism spectrum disorders: a randomized clinical trial. World Journal of Pediatrics. 2016;12(4):436-442. [PubMed] [Google Scholar]

5. Coury DL, Ashwood P, Fasano A, et al. Gastrointestinal conditions in children with autism spectrum disorder: developing a research Agenda. Pediatrics. 2012;130(2):S160-S168. [PubMed] [Google Scholar]

6. Giovanni Carta M, Conti A, Lecca F, et al. The burden of depressive and bipolar disorders in celiac disease. Clin Pract Epidemiol Ment Health. 2015;11:180-185. [PMC free article] [PubMed] [Google Scholar]

7. Busby E, Bold J, Fellows L, Rostami K. Mood disorders and gluten: it’s not all in your mind! A systematic review with meta-analysis. Nutrients. 2018;10(11):1708. [PMC free article] [PubMed] [Google Scholar]

8. Dickerson F, Stallings C, Origoni A, et al. Markers of gluten sensitivity and celiac disease in bipolar disorder. Bipolar Disord. 2011;13(1):52-58. [PubMed] [Google Scholar]

9. Shewry PR, Halford NG, Belton PS, Tatham AS. The structure and properties of gluten: an elastic protein from wheat grain. Philos Trans R Soc Lond Ser B Biol Sci. 2002;357:133-142. [PMC free article] [PubMed] [Google Scholar]

10. Balakireva A, Zamyatnin A. Properties of GLUTEN intolerance: gluten STRUCTURE, evolution, pathogenicity and detoxification capabilities. Nutrients. 2016;8(10):644. [PMC free article] [PubMed] [Google Scholar]

11. Niland B, Cash BD. Health benefits and adverse effects of a gluten-free diet in non-celiac disease patients. Gastroenterol Hepatol. 2018;14(2):82-91. [PMC free article] [PubMed] [Google Scholar]

12. Gutiérrez S, Pérez-Andrés J, Martínez-Blanco H, et al. The human digestive tract has proteases capable of gluten hydrolysis. Mol Metabol. 2017;6(7):693-702. [PMC free article] [PubMed] [Google Scholar]

13. De Re V, Magris R, Cannizzaro R. New Insights into the pathogenesis of celiac disease. Front Med. 2017;4:137.: Article [PMC free article] [PubMed] [Google Scholar]

14. Pennisi M, Bramanti A, Cantone M, Pennisi G, Bella R, Lanza G. Neurophysiology of the “Celiac Brain”: disentangling gut-brain connections. Front Neurosci. 2017;11:498. [PMC free article] [PubMed] [Google Scholar]

15. Bella R, Lanza G, Cantone M, et al. Effect of a gluten-free diet on cortical excitability in adults with celiac disease. PLoS One. 2015;10:e0129218. [PMC free article] [PubMed] [Google Scholar]

16. Mohan M, Okeoma CM, Sestak K. Dietary gluten and neurodegeneration: a case for preclinical studies. Int J Mol Sci. 2020;21:5407. [PMC free article] [PubMed] [Google Scholar]

17. Shalev H, Serlin Y, Friedman A. Breaching the blood-brain barrier as a gate to psychiatric disorder. Cardiovascular Psychiatry and Neurology. 2009;2009:1-7. [PMC free article] [PubMed] [Google Scholar]

18. Levinta A, Mukovozov I, Tsoutsoulas C. Use of a gluten-free diet in schizophrenia: a systematic review. Advances in Nutrition. 2018;9(6):824-832. [PMC free article] [PubMed] [Google Scholar]

20. Lau NM, Green PHR, Taylor AK, et al. Markers of celiac disease and gluten sensitivity in children with autism. PLoS One. 2013;8(6):e66155. [PMC free article] [PubMed] [Google Scholar]

21. Adams J, Audhya T, Geis E, et al. Comprehensive nutritional and dietary intervention for autism spectrum disorder-a randomized, controlled 12-month trial. Nutrients. 2018;10(3):369. [PMC free article] [PubMed] [Google Scholar]

22. Caccamo D, Currò M, Ientile R. Potential of transglutaminase 2 as a therapeutic target. Expert Opin Ther Targets. 2010;14:989-1003. [PubMed] [Google Scholar]

23. Sestak K, Fortgang I. Celiac and non-celiac forms of gluten sensitivity: shifting paradigms of an old disease. Br Microbiol Res J. 2013;3:585-589. [PMC free article] [PubMed] [Google Scholar]

24. Griffin M, Casadio R, Bergamini CM. Transglutaminases: nature’s biological glues. Biochem J. 2002;368:377-396. [PMC free article] [PubMed] [Google Scholar]

25. Baizabal-Carvallo JF, Jankovic J. Movement disorders in autoimmune diseases. Mov Disord. 2012;27:935-946. [PubMed] [Google Scholar]

26. Hadjivassiliou M, Aeschlimann P, Sanders DS, et al. Transglutaminase 6 antibodies in the diagnosis of gluten ataxia. Neurology. 2013;80:1740-1745. [PubMed] [Google Scholar]

27. Vöroös P, Sziksz E, Himer L, et al. Expression of PARK7 is increased in celiac disease. Virchows Arch. 2013;463:401-408. [PubMed] [Google Scholar]

28. Cristofanilli M, Gratch D, Pagano B, et al. Transglutaminase-6 is an autoantigen in progressive multiple sclerosis and is upregulated in reactive astrocytes. Multiple Sclerosis Journal. 2016;23:1707-1715. [PubMed] [Google Scholar]

29. Zis P, Rao DG, Sarrigiannis PG, et al. Transglutaminase 6 antibodies in gluten neuropathy. Digestive and Liver DiseaseLiver Dis. 2017;49:1196-1200. [PubMed] [Google Scholar]

30. Aziz I, Hadjivassiliou M, Sanders DS. The spectrum of noncoeliac gluten sensitivity. Nat Rev Gastroenterol Hepatol. 2015;12:516-526. [PubMed] [Google Scholar]

31. Daulatzai M. Non-celiac gluten sensitivity triggers gut dysbiosis, neuroinflammation, gut-brain axis dysfunction, and vulnerability for dementia. CNS Neurol Disord - Drug Targets. 2015;14:110-131. [PubMed] [Google Scholar]

32. Hadjivassiliou M, Sanders DS, Grünewald RA, Woodroofe N, Boscolo S, Aeschlimann D. Gluten sensitivity: from gut to brain. Lancet Neurol. 2010;9:318-330. [PubMed] [Google Scholar]

33. Lionetti E, Leonardi S, Franzonello C, Mancardi M, Ruggieri M, Catassi C. Gluten psychosis: confirmation of a new clinical entity. Nutrients. 2015;7:5532-5539. [PMC free article] [PubMed] [Google Scholar]

34. Gentile V, Cooper A. Transglutaminases - possible drug targets in human diseases. Curr Drug Targets - CNS Neurol Disord. 2004;3:99-104. [PubMed] [Google Scholar]

35. Bavykina IA, Zvyagin AA, Petrova IV, Nastausheva TL. Markers of gluten intolerance in children with autism spectrum disorders and down’syndrome. Zhurnal nevrologii i psikhiatrii im. S.S. Korsakova. 2018;118:64-68. [PubMed] [Google Scholar]

36. Bennabi M, Gaman A, Delorme R, et al. HLA-class II haplotypes and autism spectrum disorders. Sci Rep. 2018;8:7639. [PMC free article] [PubMed] [Google Scholar]

37. Hadjivassiliou M, Aeschlimann D, Grünewald RA, Sanders DS, Sharrack B, Woodroofe N. GAD antibody-associated neurological illness and its relationship to gluten sensitivity. Acta Neurol Scand. 2011;123:175-180. [PubMed] [Google Scholar]

38. Vaira V, Roncoroni L, Barisani D, et al. microRNA profiles in coeliac patients distinguish different clinical phenotypes and are modulated by gliadin peptides in primary duodenal fibroblasts. Clin Sci. 2013;126:417-423. [PubMed] [Google Scholar]

39. Wu F, Zikusoka M, Trindade A, et al. MicroRNAs are differentially expressed in ulcerative colitis and alter expression of macrophage inflammatory peptide-2α. Gastroenterology. 2008;135:1624-1635. [PubMed] [Google Scholar]

40. Mohan M, Chow C-E, Ryan C, et al. Dietary gluten-induced gut dysbiosis is accompanied by selective upregulation of micrornas with intestinal tight junction and bacteria-binding motifs in rhesus macaque model of celiac disease. Nutrients. 2016;8:684. [PMC free article] [PubMed] [Google Scholar]

41. Soares FLP, de Oliveira Matoso R, Teixeira LG, et al. Gluten-free diet reduces adiposity, inflammation and insulin resistance associated with the induction of PPAR-alpha and PPAR-gamma expression. J Nutr Biochem. 2013;24:1105-1111. [PubMed] [Google Scholar]

42. Ward ME, Murphy JT, Greenberg GR. Celiac disease and spinocerebellar degeneration with normal vitamin E status. Neurology. 1985;35:1199. [PubMed] [Google Scholar]

Articles from American Journal of Lifestyle Medicine are provided here courtesy of SAGE Publications

Gluten, Inflammation, and Neurodegeneration (2024)
Top Articles
Latest Posts
Article information

Author: Ouida Strosin DO

Last Updated:

Views: 6121

Rating: 4.6 / 5 (76 voted)

Reviews: 91% of readers found this page helpful

Author information

Name: Ouida Strosin DO

Birthday: 1995-04-27

Address: Suite 927 930 Kilback Radial, Candidaville, TN 87795

Phone: +8561498978366

Job: Legacy Manufacturing Specialist

Hobby: Singing, Mountain biking, Water sports, Water sports, Taxidermy, Polo, Pet

Introduction: My name is Ouida Strosin DO, I am a precious, combative, spotless, modern, spotless, beautiful, precious person who loves writing and wants to share my knowledge and understanding with you.