Gluten, in Ammation, and Neurodegeneration

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 4

American Journal of Lifestyle Medicine Jan • Feb 2022

Ashok Philip, PhD and


Nicole D. White, PharmD, CDCES, NBC-HWC, DipACLM

Gluten, Inflammation, and


Neurodegeneration
Abstract: Growing evidence question as to whether gluten may Gluten consumed through the diet
supports a potential link between be implicated in the pathogenesis of has been linked to a pathological
dietary gluten intake and neurodegenerative conditions. immune response in susceptible
neurodegenerative disease in Gluten, primarily comprising populations.10-13 Upon
susceptible populations. gliadin and glutenin proteins consumption, gluten is partially
Observational data supporting this (8085%), is a member of the hydrolyzed by proteases in the
link are described along with prolamin superfamily and is gastrointestinal tract to peptides of
interventional study data commonly found in wheat, barley, ∼1030 amino acids in length that
evaluating the effects of restricting rye, and oats. The prolamin cross the intestinal barrier via trans or
gluten from the diet in patients with superfamily of proteins is paracellular transport. These
neurologic disorders. Suggested characterized by a repetitive peptides undergo deamidation by
underlying mechanisms between sequence of the amino acids intestinal tissue transglutaminase-2
gluten intake and glutamine and proline and is (tTG2) which increases their affinity
neurodegeneration are discussed.

Keywords: gluten; inflammation;


neurodegeneration; neurologic
‘“...dietary gluten-induced chronic
disorders; celiac disease; Nonceliac inflammation is linked to dysbiosis and
gluten sensitivity

Gluten plays an established role in


a permeable gut...” ’
the pathogenesis of celiac disease suspected to be responsible for for major histocompatibility complex
(CD) and nonceliac gluten sensitivity gluten’s water insolubility. Gluten II (MHC II) molecules and triggers an
(NCGS).1 Interestingly, a number of within grains is confined to the cells inflammatory response. In
neurodegenerative conditions have of the starchy endosperm where it individuals with celiac disease (CD)
been associated with grain intake or supports germination. In food who carry human leukocyte antigen
respond favorably when gluten is production, gluten proteins are class II with DQ2 and/or DQ8
restricted from the diet.2-4 valued for their cohesiveness and molecules on antigen-presenting
Additionally, rates of comorbid CD/ viscoelasticity, which allows for ideal cells (HLA-DQ2 or DQ8 haplotype),
NCGS and neurodegenerative dough and bread consistency.9,10 As these partially digested peptides are
disorders, including bipolar, major a result, gluten is highly prevalent in recognized by the DQ cell receptor
depressive disorder, anxiety, and a variety of common foods (bread, and presented to T lymphocytes,
autism spectrum disorders (ASD), pasta, bakery items, and cereal) as resulting in an immune response. In
have been observed in unusually well as used for sauce thickeners, patients with nonceliac gluten
high rates.5-8 These findings raise the fillers, and stabilizers. sensitivity (NCGS), who are DQ2

DOI: 10.1177/15598276211049345. 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). Address correspondence to: Ashok Philip, Director
of Assessment and Professor of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, College of Pharmacy 4301 W. Markham | Little Rock, AR 72205, USA. e-
mail: aephilip@uams.edu.
For reprints and permissions queries, please visit SAGE’s Web site at http://www.sagepub.com/journalsPermissions.nav.
Copyright © 2022 The Author(s).

32
vol. 16 • no. 1 American Journal of Lifestyle Medicine

and DQ8 negative, it is suspected between the 2 and/or a beneficial autoantibodies.22,23 The presence of
that gluten proteins in concert with effect on neurological symptoms tTG2 autoantibodies were detected
other components of wheat activate when gluten is restricted from the in almost 100% of patients diagnosed
an innate immune response in the diet. One example is the reduction with CD and in a minor fraction of
body. Wheat germ agglutinin and in observed incidence of NCGS patients.23 Tissue
amylase-trypsin inhibitors of wheat schizophrenia in populations who transglutaminase is also involved in
have also been shown to illicit an consume little to no grains multiple neurodegenerative
innate immune response.10-13 As compared to those consuming diseases. The neural isoform of
a result, due to loss of tolerance to grain-rich diets.2 Patients with tissue transglutaminase, tTG6, is
gluten proteins, individuals with CD schizophrenia have markedly high implicated in the pathogenesis of
and NCGS experience gastrointestinal levels of inflammation, and Alzheimer’s disease (AD),
distress, fatigue, and pain including a gluten-free diet (GFD) has been Huntington’s disease (HD), and
inflammation and increased shown to improve both psychiatric movement disorders like gluten
permeability of the intestinal mucosa.1 and gastrointestinal symptoms in ataxia and multiple sclerosis.24-28
These inflammatory effects of these patients.3,18,19 Likewise, GFD Increased levels of circulating anti-
gluten may not be limited to the has been associated with tTG6 antibodies are also present in
gastrointestinal system. Increased improvements in ASD.4,5,20,21 adult patients with
intestinal permeability leads to entry Unusually high rates of chronic schizophrenia.16,29
of toxic digestive metabolites, diarrhea, constipation, and bloating Overall, increased levels of tTG6
bacteria, and bacterial toxins into the have been observed in children with antibodies in CD patients with gluten
bloodstream which may eventually ASD.5 Interventional studies have ataxia and peripheral neuropathy
reach the central nervous system.1 demonstrated improvements in both suggest a plausible role for tTG6 in
Neurological issues like cerebellar the prevalence of gastrointestinal contributing toward these central
ataxia, peripheral neuropathy, symptoms and ASD behaviors in and peripheral symptoms of CD.26,29
cognitive impairment, and children with ASD who followed With regards to NCGS patients, it is
neuropsychiatric diseases have been GFD.4 Mood disorders (bipolar suggested that loss of integrity of the
associated with CD, suggesting the disorder, major depressive disorder, intestinal barrier facilitates access of
possibility of gluten-mediated and anxiety) are commonly observed partially hydrolyzed immunogenic
inflammation playing a role in loss of in individuals with CD and NCGS, gluten peptides to both the systemic
bloodbrain barrier (BBB) with bipolar disorder 17 times more circulation and the brain. Access of
integrity.1,14-16 Furthermore, likely to affect individuals with CD these toxic gluten peptides and HLA-
increased BBB permeability relative to the general population.6,7 DQ2/DQ8 restricted CD4 T cells to
attributed to inflammation or Increased levels of gluten-related the brain may contribute to the
bacteria is linked to other antibodies are found in individuals neurological issues observed in
neurological disorders including with bipolar disorder, depression, NCGS patients.30-33 It is also
ASD, dementia, Alzheimer’s disease, anxiety, hyperactivity, as well as proposed that loss of intestinal
Parkinson’s disease (PD), schizophrenia suggesting the barrier’s integrity aka leaky gut
depression, anxiety, and correlation between gluten promotes increased access for
schizophrenia.15,17 The following consumption and mood disorders.6-8 circulating or gut-derived
describes observed relationships extracellular structures like
between gluten and biomolecular condensates (BMC)
Putative Mechanisms
neurodegenerative conditions as and extracellular vesicles (EV) to the
well as potential mechanisms by As stated earlier, intestinal tissue brain of CD and NCGS patients.16
which dietary gluten may lead to transglutaminase-2 (tTG2) is Given the critical role of tTG in
neurodegeneration. a calcium-dependent enzyme that mediating gluten metabolism, its
mediates deamidation of glutamine autoantigen properties, its ability to
residues present in gluten proteins, cause central β-amyloid
Gluten and
triggering gluten’s inflammatory accumulation, and foster
Neurodegenerative
response.16,22 tTG2 not only inflammation and cancer, tTG is
Conditions
mediates the post-translational being pursued as a molecular target
Despite the paucity of evidence modification/deamidation of the for a variety of disease states.16,34
establishing a causal relationship gluten protein gliadin, but was Additional mechanisms have been
between dietary gluten intake and shown to act as an autoantigen and postulated to contribute to gluten
neurological disorders, a number of due to its ability to induce the sensitivity and neuronal dysfunction.
studies demonstrate an association production of tTG2 One of them is the identification of

33
American Journal of Lifestyle Medicine Jan • Feb 2022

a strong correlation between specific molecular target for the management with autism spectrum disorder:
HLA haplotypes and dysregulation of gluten-induced intestinal developing a research Agenda.
Pediatrics. 2012;130(2):S160-S168.
of the gut-brain axis in CD and NCGS inflammation and dysbiosis.16,41
patients diagnosed with ASD and/or 6. Giovanni Carta M, Conti A, Lecca F,
et al. The burden of depressive and
Down’s syndrome (DS).16,35,36 Also, Conclusion bipolar disorders in celiac disease. Clin
in NCGS patients with neurological Pract Epidemiol Ment Health. 2015;11:
While data supporting the direct
issues, antibodies for glutamic acid 180-185.
link between gluten consumption
decarboxylase (GAD), a key enzyme 7. Busby E, Bold J, Fellows L, Rostami K.
and neurodegeneration is limited,
involved in the biosynthesis of the Mood disorders and gluten: it’s not all
available evidence highlights the in your mind! A systematic review with
primary inhibitory neurotransmitter
relationship between gluten meta-analysis. Nutrients. 2018;10(11):
gamma-aminobutyric acid, were
consumption, tTG antibody 1708.
detected.16,37
production, and disruption of the 8. Dickerson F, Stallings C, Origoni A,
The potential role of epigenetic
microbiota-gut-brain axis.1,16 The et al. Markers of gluten sensitivity and
mechanisms in regulating celiac disease in bipolar disorder.
beneficial effects of GFD in patients
inflammatory responses has Bipolar Disord. 2011;13(1):52-58.
with CD, schizophrenia, and ASD,
prompted focused attention toward 9. Shewry PR, Halford NG, Belton PS,
especially in mitigating
micro-RNAs (MiRNAs) and CD.16 Tatham AS. The structure and
gastrointestinal and neurological
The MiRNAs are ∼20–23 nucleotides properties of gluten: an elastic protein
symptoms, highlight the need for from wheat grain. Philos Trans R Soc
in length, small RNA molecules that
more exploration and identification Lond Ser B Biol Sci. 2002;357:133-142.
mediate a number of cellular
of definitive evidence.42 10. Balakireva A, Zamyatnin A. Properties
processes like cell proliferation,
of GLUTEN intolerance: gluten
differentiation, apoptosis, cell STRUCTURE, evolution, pathogenicity
signaling, and immune, including Declaration of Conflicting and detoxification capabilities.
inflammatory, responses.16 Interests Nutrients. 2016;8(10):644.
Specifically, downregulation of miR- The author(s) declared no potential conflicts of interest 11. Niland B, Cash BD. Health benefits and
192-5p due to inflammation is with respect to the research, authorship, and/or adverse effects of a gluten-free diet in
suggestive of its role in maintaining publication of this article. non-celiac disease patients.
Gastroenterol Hepatol. 2018;14(2):
intestinal homeostasis.16,38,39
82-91.
Moreover, chronic inflammation of
Funding 12. Gutiérrez S, Pérez-Andrés J, Martı́nez-
the intestine combined with a leaky
Blanco H, et al. The human digestive
gut results in gut dysbiosis, which The author(s) received no financial support for the
tract has proteases capable of gluten
may in turn promote microbial entry research, authorship, and/or publication of this article.
hydrolysis. Mol Metabol. 2017;6(7):
into systemic circulation. Especially, 693-702.
loss of intestinal barrier’s integrity 13. De Re V, Magris R, Cannizzaro R. New
has the potential to allow increased References Insights into the pathogenesis of celiac
access of lipopolysaccharides (LPS) disease. Front Med. 2017;4:137.: Article
1. Obrenovich M. Leaky gut, leaky brain?
across the BBB, activate microglial Microorganisms. 2018;6:107. 14. Pennisi M, Bramanti A, Cantone M,
cells, and ultimately cause neuronal Pennisi G, Bella R, Lanza G.
2. Dohan FC, Harper EH, Clark MH, Neurophysiology of the “Celiac Brain”:
inflammation and damage in CD Rodrigue RB, Zigas V. Is schizophrenia disentangling gut-brain connections.
patients.16,40 rare if grain is rare? Biol Psychiatr. Front Neurosci. 2017;11:498.
In CD and NCGS individuals, it is 1984;19:385-399.
15. Bella R, Lanza G, Cantone M, et al.
established that dietary gluten- 3. Kelly DL, Demyanovich HK, Rodriguez Effect of a gluten-free diet on cortical
induced chronic inflammation is KM, et al.. Randomized controlled trial excitability in adults with celiac
of a gluten-free diet in patients with
linked to dysbiosis and a permeable schizophrenia positive for antigliadin
disease. PLoS One. 2015;10:e0129218.
gut.16 This in turn affects factors that antibodies (AGA IgG): a pilot feasibility 16. Mohan M, Okeoma CM, Sestak K.
regulate neuronal inflammation, study. J Psychiatr Neurosci. 2019;44(4): Dietary gluten and neurodegeneration:
cognition, and neurodegeneration. 269-276. a case for preclinical studies. Int J Mol
Sci. 2020;21:5407.
For example, at the molecular level, 4. Ghalichi F, Ghaemmaghami J, Malek A,
the genetic expression of Ostadrahimi A. Effect of gluten free diet 17. Shalev H, Serlin Y, Friedman A.
on gastrointestinal and behavioral Breaching the blood-brain barrier as
peroxisome proliferator activated a gate to psychiatric disorder.
indices for children with autism
receptor gamma (PPARγ) is reduced spectrum disorders: a randomized Cardiovascular Psychiatry and
in patients diagnosed with ulcerative clinical trial. World Journal of Neurology. 2009;2009:1-7.
colitis and CD. Because PPARγ is an Pediatrics. 2016;12(4):436-442. 18. Levinta A, Mukovozov I, Tsoutsoulas C.
anti-inflammatory and anti-dysbiotic 5. Coury DL, Ashwood P, Fasano A, et al. Use of a gluten-free diet in
molecule, it may serve as a potential Gastrointestinal conditions in children schizophrenia: a systematic review.

34
vol. 16 • no. 1 American Journal of Lifestyle Medicine

Advances in Nutrition. 2018;9(6): 28. Cristofanilli M, Gratch D, Pagano B, 36. Bennabi M, Gaman A, Delorme R, et al.
824-832. et al.. Transglutaminase-6 is an HLA-class II haplotypes and autism
19. Vellinga J. Can gluten cause mental autoantigen in progressive multiple spectrum disorders. Sci Rep. 2018;8:
health disorder?. https://www. sclerosis and is upregulated in reactive 7639.
tcimedicine.com/post/can-gluten- astrocytes. Multiple Sclerosis Journal. 37. Hadjivassiliou M, Aeschlimann D,
cause-mental-health-disorders. https:// 2016;23:1707-1715. Grünewald RA, Sanders DS, Sharrack B,
www.tcimedicine.com/post/can- 29. Zis P, Rao DG, Sarrigiannis PG, et al. Woodroofe N. GAD antibody-
gluten-cause-mental-health-disorders Transglutaminase 6 antibodies in associated neurological illness and its
Accessed Aug 15, 2021. gluten neuropathy. Digestive and Liver relationship to gluten sensitivity. Acta
20. Lau NM, Green PHR, Taylor AK, et al. DiseaseLiver Dis. 2017;49:1196-1200. Neurol Scand. 2011;123:175-180.
Markers of celiac disease and gluten 30. Aziz I, Hadjivassiliou M, Sanders DS. 38. Vaira V, Roncoroni L, Barisani D, et al.
sensitivity in children with autism. PLoS The spectrum of noncoeliac gluten microRNA profiles in coeliac patients
One. 2013;8(6):e66155. sensitivity. Nat Rev Gastroenterol distinguish different clinical
21. Adams J, Audhya T, Geis E, et al. Hepatol. 2015;12:516-526. phenotypes and are modulated by
Comprehensive nutritional and dietary 31. Daulatzai M. Non-celiac gluten gliadin peptides in primary duodenal
intervention for autism spectrum sensitivity triggers gut dysbiosis, fibroblasts. Clin Sci. 2013;126:
disorder-a randomized, controlled 12- neuroinflammation, gut-brain axis 417-423.
month trial. Nutrients. 2018;10(3):369. dysfunction, and vulnerability for 39. Wu F, Zikusoka M, Trindade A, et al.
22. Caccamo D, Currò M, Ientile R. dementia. CNS Neurol Disord - Drug MicroRNAs are differentially expressed
Potential of transglutaminase 2 as Targets. 2015;14:110-131. in ulcerative colitis and alter expression
a therapeutic target. Expert Opin Ther 32. Hadjivassiliou M, Sanders DS, of macrophage inflammatory peptide-
Targets. 2010;14:989-1003. Grünewald RA, Woodroofe N, Boscolo 2α. Gastroenterology. 2008;135:
S, Aeschlimann D. Gluten sensitivity: 1624-1635.
23. Sestak K, Fortgang I. Celiac and non- 40. Mohan M, Chow C-E, Ryan C, et al.
from gut to brain. Lancet Neurol. 2010;
celiac forms of gluten sensitivity: Dietary gluten-induced gut dysbiosis is
9:318-330.
shifting paradigms of an old disease. Br accompanied by selective
Microbiol Res J. 2013;3:585-589. 33. Lionetti E, Leonardi S, Franzonello C,
upregulation of micrornas with
Mancardi M, Ruggieri M, Catassi C.
24. Griffin M, Casadio R, Bergamini CM. intestinal tight junction and bacteria-
Gluten psychosis: confirmation of
Transglutaminases: nature’s biological binding motifs in rhesus macaque
a new clinical entity. Nutrients. 2015;7:
glues. Biochem J. 2002;368:377-396. model of celiac disease. Nutrients.
5532-5539.
2016;8:684.
25. Baizabal-Carvallo JF, Jankovic J. 34. Gentile V, Cooper A.
Movement disorders in autoimmune 41. Soares FLP, de Oliveira Matoso R,
Transglutaminases - possible drug Teixeira LG, et al. Gluten-free diet
diseases. Mov Disord. 2012;27:935-946. targets in human diseases. Curr Drug reduces adiposity, inflammation and
26. Hadjivassiliou M, Aeschlimann P, Targets - CNS Neurol Disord. 2004;3: insulin resistance associated with the
Sanders DS, et al. Transglutaminase 6 99-104. induction of PPAR-alpha and PPAR-
antibodies in the diagnosis of gluten 35. Bavykina IA, Zvyagin AA, Petrova IV, gamma expression. J Nutr Biochem.
ataxia. Neurology. 2013;80:1740-1745. Nastausheva TL. Markers of gluten 2013;24:1105-1111.
27. Vöroös P, Sziksz E, Himer L, et al. intolerance in children with autism 42. Ward ME, Murphy JT, Greenberg GR.
Expression of PARK7 is increased in spectrum disorders and down’syndrome. Celiac disease and spinocerebellar
celiac disease. Virchows Arch. 2013; Zhurnal nevrologii i psikhiatrii im. S.S. degeneration with normal vitamin E
463:401-408. Korsakova. 2018;118:64-68. status. Neurology. 1985;35:1199.

35

You might also like