Celiac disease: Pathogenesis, disease management and new insights into the herbal-based treatments (2024)

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

Celiac disease: Pathogenesis, disease management and new insights into the herbal-based treatments (1)

Narra J. 2023 Aug; 3(2): e147.

Published online 2023 Jul 12. doi:10.52225/narra.v3i2.147

PMCID: PMC10919711

PMID: 38454981

Author information Article notes Copyright and License information PMC Disclaimer

Associated Data

Data Availability Statement

Abstract

Celiac disease (CD) is a gluten intolerance autoimmune disorder which its symptoms involve the gastrointestinal tract and sometimes the other organs. It is one of the most prevalent health problems rising in many populations as statistics show that in every 100 people about one person is suffering from CD. It has been observed that the persons who genetically contain the human leukocyte antigen (HLA) DQ2 and HLA DQ8 genes involved in the immune system haplotypes are more prone to develop an allergy to gluten. The only treatment currently available for CD is a strict gluten-free diet. However, recent research has shown promising new insights into the herbal-based treatments of CD. New insight on CD is now offering various prospects to manage its treatment, diagnosis, and serving in the development of advanced therapies. Several herbs and botanical extracts have demonstrated anti-inflammatory, immunomodulatory, and gut-healing properties that make them potential candidates for the management of CD. Here, we provide an updated review on pathogeneses and managements of CD. In particular, we summarize the current understandings of herbal-based treatments for CD and highlights their potential benefits.

Keywords: Celiac disease, autoimmune disorder, gluten, human leukocyte antigen, genetic

Introduction

Celiac disease (CD) (gluten-sensitive enteropathy) is a genetic, multisystemic autoimmune-mediated enteropathy that grounds inflammation of the small intestine due to the intake of gluten and restricts the absorption of dietary nutrients [1-3]. People suffering from CD are permanently gluten intolerant [4]. It was mentioned that polygenic predisposition, high pervasiveness (1%) and widely assorted expressions are responsible for other autoimmune disorders. The basis of the disease observed by diagnosis is characteristic or symptomless inflammation of the small intestinal mucosa which can be treated by a gluten-free diet [4-6].

At the time of World War II, Professor Dicke reported much less improvement in children with CD and later he described the involvement of wheat and rye in CD [1]. In the early 1950s, when it was first identified that gluten is responsible for CD, it has been advisable to the patients suffering from CD to follow the strict gluten-free diet (GFD). Elevated morbidity, mortality, and impaired quality of life are the result due to symptomatic and untreated CD. Charlotte Anderson also identified gluten as an aberrant component extracted from wheat. Later in the 1960s, physicians started to diagnose CD by small intestinal biopsies and further transglutaminase was recognized as the autoantigen in CD in 1997 [7, 8]. Earlier in the twentieth century, a new high-banana diet was recommended to reduce the high mortality rate of this disease. This diet specifically became the foundation stone of treatment for CD in which bread and cereals were excluded from this diet [6, 9]. European Society for Pediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) diagnostic instructions claims the diagnosis of autoantibodies in CD and suggested the use of GFD until the exact results are out. Patients with HLA-DQ2.5 and HLA-DQB1*02 genes show more severe histological damage when diagnosed [10, 11].

Pathogenesis

Pathogenesis of CD includes various crucial steps where T-cell epitopes peptides of gluten retaliate degradation of gastrointestinal. Gluten peptide deamidation is catalyzed by the tTG enzyme, which then easily gets attached to the predisposing HLA-DQ gene on intestinal antigen-presenting cells (APCs) [10]. When gluten-specific CD4+ T cells get activated, IFN-γ and IL-21 type cytokines are released which further activate B-cell and intraepithelial lymphocytes (IELs) that result in gastrointestinal lesions [12]. Giving a stress signal activated, IELs get converted into cytolytic NK-type cells which results in enterocytes damages. Interleukin-15 (IL-15) secretes effector T cells that do not get affected by the effect of regulatory T cells (Tregs). IL-15 provides mucosal specialized dendritic cells (DCs), encourages inflammatory reactions and inhibits differentiation of Tregs [13] (see Figure 1). The pathogenesis of CD is associated with an interaction between genes, gluten, and environmental influences.

Celiac disease: Pathogenesis, disease management and new insights into the herbal-based treatments (3)

Key steps in the pathogenesis of celiac disease. Peptides originating from gluten undergo modification by TG2 and are presented by antigen-presenting cells in the mesenteric lymph nodes (MLN) to CD4+ T cells within the context of HLA-DQ2. This interaction triggers a TH1 type response, resulting in the production of IFNγ and intestinal inflammation. The chronic inflammation further leads to the expansion and persistence of Vδ1+γδ T cells, which also contributes to IFNγ production. The expression of IL-15 and stress molecules on enterocytes is induced by gluten peptides. The increased levels of IL-15 promote a NK-like phenotype in CD8+ T cells, contributing directly to enterocyte death. A subset of CD8+γδ+ T cells is believed to have a regulatory role through the secretion of TGF-β. Plasma cells are also abundant in the lesion and express the immunodominant gluten peptide DQ2.5-glia-α1a. They are induced to secrete antibodies that bind to TG2 and other targets.

Genetics

Patients suffering from CD mainly have two predisposing genes human leukocyte antigen (HLA) class II genes which are HLA-DQ2 and HLA-DQ8 [4]. Both genes exist primarily in all patients with CD but many other non-HLA genes were also assumed to cause CD in different populations [6]. It is found that about 40% of people have at least one of these predisposing genes so the diagnosis of these genes is not sufficient to identify CD. However, if the presence of HLA-DQ2 and DQ8 are the same predictive value for CD, then there is a chance to get a negative predictive value for the CD when diagnosed [14, 15].

Gluten

The development of CD depends on the presence of predisposing genes and in addition, it also depends on exposure to gluten in the diet. Gliadins and glutenins are both types of protein that are present in gluten. Gliadins are monomeric proteins, whereas glutenins are defined as polymeric aggregated proteins. These proteins are not processed by the gastrointestinal tract since they contain proline and glutamine peptides which are alcohol soluble as they oppose deprivation by luminal border endopeptidases [16].

An essential step in the pathogenesis of CD takes place in lamina propria due to the activation of the tissue transglutaminase (tTG) enzyme. tTG enzyme deamidates gliadin with deamidase enzyme, which makes it immunogenic and affects the adaptive immune system. Gliadin activates adaptive immune response which encompasses antigen-depicting cells, such as macrophages, dendritic cells, and B cells [17]. These gliadin-specific CD41 TH1 cells and antigen-depicting cell interaction produce inflammatory cytokines, for example, IFN-g [18].

In addition, due to presence of gliadin, innate immune response increases the production of cytokines, macrophages, enterocytes, and dendritic cells in the epithelial constituent of the intestinal mucosa [19]. As a result, intraepithelial lymphocytes differentiate into NK-G2D markers represented as cytotoxic CD81 T cells. Direct ligands on the epithelial surface for NK-G2D are promoted by IL-15. This cascade of inflammatory mediators at last causes collective intestinal mucosa damage illustrated by crypt hyperplasia and villous atrophy [9, 17].

Environmental factors

Various environmental components add to the pathogenesis of CD. Earlier, in the 1950s, factors like breastfeeding, infection, and modifications in the intestinal microbiota were taken into consideration. It was also considered that intestinal infection in infants and adults increases the risk for the future development of CD. The fundamental principle behind these factors was that an augmented permeability of epithelial enterocytes increases the entrance of gluten protein like gliadin, which further activates tTG enzyme in lamina propria and these infections increase the production of interferon [12, 20]. Recently, some data also recommended that breastfeeding may have a defensive effect against the progression of CD at 4 to 6 months of age, especially if gluten is also provided in the diet. Infants at this age acquire immune tolerance against CD through breastfeeding [7].

Another environmental factor that is known to influence CD development is the intestinal microbiome which is also widely studied and investigated. It was found in some duodenal biopsy samples of children with the CD (with or without GFD) that there is an altered microbiome [21]. Further in this study, it was found that subjects who were not given a GFD show increased proportions of Bacteroides species and reduced Bifidobacterium species compared to those subjects who were provided with GFD and control subjects without CD. Then likewise, the Bacteroides versus Bifidobacterium species proportions were normalized in the subjects by recommending GFD. As concluded that the intestinal microbiome is a potential marker and has a pathogenic role in CD [22]. In a placebo-controlled trial study, patients with CD on GFD administered daily with Bifidobacterium species and their intestinal microbiome, cytokine, and secretory IgA profile were studied. It was concluded that Bacteroides species and secretory IgA levels decrease in stools. Although these findings reported promising results, more advanced research is needed before concluding immunopathogenic role of the intestinal microbiome in CD patients [21, 23].

Diagnosis

Various methods are used for diagnosis of CD including small intestinal biopsy and serologic screening which are extremely profound and specific for antibodies. With the invention of quick screening techniques and knowledge of CD, many individuals are diagnosed including those who are asymptomatic [24-26].

Small intestinal biopsy

Histologic analysis in small intestinal biopsy indicates villous atrophy, intraepithelial lymphocytosis, and crypt hyperplasia. It also shows the presence of tissue transglutaminase enzyme (tTG), deamidated gliadin peptides (DGP), CD-specific antibodies, and endomysial antibodies (EMA) [24].

Serologic tests

In the CD, gluten presence causes intestinal injury because of the antibodies produced by the autoimmune system. These specific antibodies are tested in serological tests [27]. The preliminary screening tests for CD are (1) Quantitative immunoglobulin A (IgA) test; (2) IgA anti-tissue transglutaminase antibodies (tTG-IgA); and (c) IgA anti-endomysial antibody test. There is an additional test IgA anti-EMA test for CD patients, especially for those who are suffering from diabetes mellitus (type 1). IgA anti-endomysial antibodies are present in about 90% of patients [28, 29].

Genetic testing

A quick specific test to diagnose CD is not available yet. However, commonly the presence of two pre-disposing genes, HLA-DQ2 and DQ8 alleles, are studied for diagnosis. Notably, the presence of one or both alleles are certain in a patient with CD, but about 50% of the general population shows the presence of these alleles as well, so these tests are not fully helpful in CD diagnosis. However, if genetic testing shows that both genes are negative in a patient, then the possibility of CD is very low [20, 29, 30].

Symptoms of celiac disease

The signs of CD differ amongst various patients and have different effects on the human body. For example, a patient may have symptoms like severe diarrhea with stomach pain while other patients may show liver, neurological, skin, or dental problems. The most common characteristic of CD symptoms are gastrointestinal, behavioral/psychiatric, and autoimmune symptoms [31].

Some of gastrointestinal symptoms includes diarrhea, vomiting, lactose intolerant, abdominal distention, malabsorption, alteration in appetite, constipation, bacterial infection, flatulence, reflux heartburn, hepatitis, bloating, ulcers (mouth, esophageal and stomach), dyspepsia, irritable bowel syndrome, and inflammatory bowel disease (IBD) [3-34]. However, the behavior symptoms include hypochondria, incapability to concentrate, brain fog, anxiety, neurosis irritability, depression, attention-deficit/hyperactivity disorder (ADHD), obsessive-compulsive disorder (OCD), and autism spectrum disorder [35-37]. Hashimoto's disease, Grave’s disease, Sjogren's syndrome, hyperparathyroidism, dilated (congestive) cardiomyopathy, fibromyalgia, alopecia areata, rheumatoid arthritis, multiple sclerosis and systemic lupus erythema are some of the autoimmune response-based symptoms [38-40].

Treatment strategies of celiac disease

Medical nutrition therapy is the only established method for CD treatment [41]. Treatment of CD primarily includes a gluten-free diet (GFD) and it is compulsorily recommended to all those patients who are diagnosed to be suffering from CD. GFD includes avoiding all foods made from wheat, barley, and rye. The safe range for CD patients is 50-100 mg per day of gluten in the diet. Fish, plain meat, fruits, and various vegetables are free from gluten, so patients with CD can consume this type of diet. Also, CD patients are instructed to take multivitamins that are gluten-free [42-45]. Examples of various herbs that can be used for the management of CD are mentioned in Table 1 and their chemical structures are described in Figure 2.

Table1.

Examples of various herbs for the management of celiac disease [46]

Name of the herbsRole played
ChamomileCarminative
MeadowsweetAntacid and anti-inflammatory
Lemon balmReduce bloating
LiquoriceAntispasmodic
PeppermintCarminative and antispasmodic
FennelCarminative and antispasmodic
Slippery elmDemulcent action
Celiac disease: Pathogenesis, disease management and new insights into the herbal-based treatments (4)

The chemical structures of herbal plants constituents used in the treatment of celiac disease based on previous studies [47-49].

Chamomile (Matricaria chamomilla L.) that belongs to the Asteraceae family is a well-known therapeutic plant. It has been used in herbal treatment for thousands of years. Chamomile is used mainly as an antiseptic, anti-inflammatory, and antispasmodic, and very effectively for urinary tract swelling and menstrual pain. The other therapeutic properties include carminative, healing, and sedative activity [50-53].

Meadowsweet (Filipendula ulmaria L.) is used as a diuretic, anti-hemorrhoid, sedative, anti-inflammatory, wound-healing, analgesic, antiulcerogenic, hypoglycemic, and astringent in some European countries. Conventionally, it has been used for rheumatic pains, dyspepsia with hyperacidity and heartburn, and diarrhea (children) and specifically for the prevention and management of peptic ulcers [47-49, 54, 55].

Lemon balm (Melissa officinalis L.) has its place in the mint family. Lemon balm has medicinal properties like flatus relieving, digestion promotion, antidepressant, antioxidant, and antiviral activity. According to the German Commission E monograph, it is also prescribed in the gastrointestinal complaints especially cramps in the digestive tract and flatulent dyspepsia [56-59].

Liquorice (Glycyrrhiza glabra L.) is known for its ethnopharmacological value in traditional medicine across the world. Liquorice has been recommended as a preventive agent for duodenal and gastric ulcers. During allergy, it is used in dyspepsia as an anti-inflammatory agent. It is also employed as an emmenagogue, contraceptive, anti-asthmatic, laxative, galactagogue, and antiviral agent in folk therapy [60-62].

Peppermint (Mentha piperita L.) that belongs to the family Lamiaceae (Labiatae) is an interminable herb and is a combination of two plants viz. spearmint (Mentha spicata L.) and water mint (Mentha aquatica L). The plant is extensively used in folk or traditional medicine for the management of digestive and nervous system disorders. The herb also has antitumor and antimicrobial properties, chemopreventive potential, renal actions, antiallergenic effects, and is for lessening cramps, digestive complaints, anorexia, nausea, and diarrhea [63-65].

Fennel (Foeniculum vulgare Mill) is a therapeutic plant belonging to the Umbelliferae (Apiaceae) family. As a traditional medicine, it is used for many types of ailments like diseases related to digestive, respiratory, reproductive, and endocrine systems. In various countries, it is used to stimulate appetite, and treat abdominal pain, antiemetic, arthritis, cancer, stomach pain in children, irritable colon, constipation, depurative, emmenagogue, conjunctivitis, fever, gastritis, flatulence, insomnia, kidney ailments, laxative, liver pain, leucorrhea, and mouth ulcer [66-69].

Slippery elm (Ulmus rubra) is an herbal medicine obtained from its inner bark. According to the Natural Medicines Comprehensive Database, it is used for the treatment of sore throat, dysphasia, colic, loose motions, hemorrhoids, irritable bowel syndrome, septicemia of the urinary tract, syphilis, tapeworm expulsion, as well as protecting against stomach and duodenal ulcers, swelling of the colon, GI inflammation, and acidity [70-72].

Adjuvant therapies for the management of celiac disease

Oral enzyme supplementation

It has been reported that in vitro T cell toxicity is decreased by admixtures of purified prolyl endopeptidases (PEPs) and sprouting wheat proteases, by hydrolyzing gluten [59, 73]. PEPs obtained from various bacterial species like Flavobacterium meningosepticum, Myxococcusxanthus, and Sphingomonascapsulata, and some from fungal species like Aspergillus niger are widely reviewed [47, 63].

Anti-inflammatory therapy

Many proinflammatory cytokines and lymphocytes (e.g., TNFα, IFNγ, IL-15, CCL25, CXCL10) were found to have a significant contribution to the CD. Considering these as molecular targets, scientists had already developed the drugs for these inflammatory mediators and some research is still being carried out. Infliximab Remicade® (Janssen Biotech, Titusville NJ, USA), a monoclonal antibody blocking TNFα used in RA and Crohn’s disease, had positive effects on a patient with refractory CD [38, 54, 74].

Desensitization

A desensitizing immunization with Nexvax2 as an immune therapy and preventive drug to regain gluten resistance to three immunostimulatory gluten peptides from wheat and barley were synthesized [43, 68, 75, 76].

The paracellular permeability modulator

The gliadin-mediated opening of tight junction contributes to the increased permeability of the mucosa in active CD. The reliability of lactulose as a marker for increased paracellular leakage of gliadins was contested concerning the molecular weight of lactulose (342 Da) compared to immunogenic gliadin peptides such as p31-49 and 33mer (2,245 and 3,900 Da, respectively). A fluorescently labeled gliadin fragment may be of higher relevance [38, 77-79].

Polymeric binders

In the gastrointestinal tract, polymeric binders are considered to sequester gluten. Poly(hydroxyethyl methacrylate-co-styrene sulfonate) (P(HEMA-co-SS), BL-7010, BioLineRx) is responsible to formulate gliadin complex and it can decrease the production of the immune stimulatory complex in digestive fluid. It can sequester gluten presence in the stomach, which is a significant benefit of this polymeric binder [77-79].

Modified grains

Modified grains are developed using various techniques like breeding of selective wheat species or by using the small interfering RNA (siRNA) technology to mutate or silence immunostimulatory sequences [56, 58].

Preventing gluten entry through the intestinal epithelium

Zonulin inhibitor larazotide (AT-1001) is studied in the animal model and is capable of treating intestinal barrier defects. AT-1001 undertaken in phase II clinical trials is presently the best pharmacological agent to treat CD patients [67, 74-76].

Molecular targets of celiac disease

Consideration of the best possible aims of therapy and the techniques to evaluate effectiveness is an evolving area. Molecular targets for CD are mentioned in Table 2.

Table2.

Molecular targets of celiac disease

ApproachProposed mechanismReference(s)
EndopeptidasesEnzymatic degradation of gluten[68]
Tight junction regulatorsReduceparacellularpassage of gluten across the mucosa[57, 59]
Gluten binding agentsSequester gluten in the intestinal lumen[56, 57, 59]
Endorphin-based therapeutic vaccineGluten-specific CD4+ T cells are targeted by epitope-specific immunotherapy[79]
Inflammatory proteases inhibitionAnti-inflammatory effects and improved barrier function[68]
HLADQ2 blockersGliadin antagonist blocks the binding grooves of HLA molecules and peptides might suppress the antigen presentation process and block the cascade of T cell stimulation[57, 59]
Immune modulation to induce tolerance to glutenProtein-based desensitization therapy-Nexvax2 vaccine[63, 78, 79]

Therapies under clinical trials

Various drugs that are under clinical trials for the treatment of CD are represented in Table 3. Some drugs such as anokion, KAN-101, CALY-002, DONQ52, EQ102 and others have been developed from different companies and have been entered to phase 1 to phase 2 clinical trials.

Table3.

Various investigational drug molecules under clinical trials for celiac disease treatment

Drug nameInnovator company/sponsorTargetPhases of clinical trials
Anokion, KAN-101Subsidiary Kanyos Bio, Inc.Target immune CD cellsPhase 1b clinical trial
CALY-002Calypso BiotechInhibit the cytokine interleukin role in the immune systemPhase 1b clinical trial for safety and tolerability
DONQ52Chugai pharmaceuticalTarget immune complex causing CDConducting phase 1 clinical trial
EQ102Equillium biotechnology companyInhibit cytokines lL-15 and 1L-21 that drive cytotoxic T cell and optimal approach CDFebruary 2022 conducting phase 1 clinical trial
GSK2915393GlaxoSmithKline (GSK)Inhibit TG2 enzyme for inflammatory action response to gluten exposure in CDCompletion on phase 1 clinical trial study in 2021
IMU-856Immunic Therapeutic biopharmaceutical companyRestore intestinal barrier, compromise immune function, treat gastrointestinal disease, and treat CDIn September 2022, Immunic announced positive results part A and B of its phase 1 clinical trial and part C on-going
HU-Mik-Beta-1National Cancer Institute and Mayo ClinicMonoclonal antibody targets the cytokine receptor subunit IL-2/IL-15RBETA tested for CDPhase 1 trial
ImmunogenxIMGX003 Alvine PharmaceuticalBreak gluten protein down to relieve CD symptomsThe technology has extensively studied in phase 1 and phase 2
TAK-062Takeda PharmaceuticalsTAK-062 is a highly potent enzyme for gluten breakdown in CD with GFDCurrently conducting phase 2 clinical trial

Conclusion and future perspectives

CD is an autoimmune allergy to dietary gluten and is very common nowadays. It can have typical intestinal manifestations. The therapy to treat this disease is to keep a patient on a GFD which lacks patient compliance. Novel therapeutic targets have been found from deep insights into the pathophysiology of CD. The new research on the molecular targets of CD has resulted in various new therapies like the engineering of gluten-free grains, reducing intestinal permeability by blocking the various receptors, formulation of new vaccines, and the use of probiotics for the degradation of immunodominant gliadin peptides. These therapies boosted the efficacy of CD management and helped in the improvement of patient conformity. However, while a GFD diet is the main key for CD therapy for the immediate foreseeable future, these treatment options are expected to get US Food and Drug Administration soon.

In CD, comprehension of severe gluten intolerance is a primary issue that needs more study. Enzyme supplementation therapy is a very assuring approach as a forthcoming treatment for CD patients which is already established to have both preclinical and clinical potentials. Recently many investigations are ongoing and recommended as various treatment strategies, but the studies are still in their initial days and their Phase III clinical trials are yet to be reached. The novel therapies for CD are still not studied for children. Hopefully, it is believed that soon we will be able to develop a good disease prevention therapy for CD with the evolving studies on the role of the microbiome and environmental factors and in what way they may influence gluten immunity.

Acknowledgments

The authors are very thankful to their respective institutions for providing support in the completion of this study.

Ethics approval

Not required.

Conflict of interest

All the authors declare that there are no conflicts of interest.

Funding

This study received no external funding.

Underlying data

All data underlying the results are available as part of the article and no additional source data are required.

How to cite

Mittal P, Arora D, Parashar S, et al. Celiac disease: Pathogenesis, disease management and new insights into the herbal-based treatments. Narra J 2023; 3 (2): e147 - http://doi.org/10.52225/narra.v3i2.147.

References

1. Popp A, Mäki M.. Gluten-induced extra-intestinal manifestations in potential celiac disease—celiac trait. Nutrients2019; 11(2):320. [PMC free article] [PubMed] [Google Scholar]

2. Salvador SM, Zigmundo GCDO, Bundrich L, et al.. Oral manifestations of celiac diseases: Case report. J Oral Surg Oral Med Oral Pathol2020; 130(3):e208. [Google Scholar]

3. Iqbal U, Chaudhary A, Karim MA, et al.. Association of autoimmune hepatitis and celiac disease: role of gluten-free diet in reversing liver dysfunction. J Investig Med High Impact Case Rep2017; 5(2):2324709617705679. [PMC free article] [PubMed] [Google Scholar]

4. Lionetti E, Antonucci N, Marinelli M, et al.. Nutritional status, dietary intake, and adherence to the mediterranean diet of children with celiac disease on a gluten-free diet: A case-control prospective study. Nutrients2020; 12(1):143. [PMC free article] [PubMed] [Google Scholar]

5. Koehler P, Wieser H, Konitzer K.. Celiac disease and gluten: Multidisciplinary challenges and opportunities, Vol. 1. Amsterdam: Elsevier. 2014. [Google Scholar]

6. Lacy B, DiBaise J, Pimentel M, et al.. Essential medical disorders of the stomach and small intestine. Cham: Springer; 2019. [Google Scholar]

7. Segura V, Ruiz-Carnicer Á, Sousa C, et al.. New insights into non-dietary treatment in celiac disease: Emerging therapeutic options. Nutrients2021; 13(7):2146. [PMC free article] [PubMed] [Google Scholar]

8. Martínez-Ojinaga E, Fernández-Prieto M, Molina M, et al.. Influence of HLA on clinical and analytical features of pediatric celiac disease. BMC Gastroenterol2019; 19:1–6. [PMC free article] [PubMed] [Google Scholar]

9. Norris JM, Barriga K, Hoffenberg EJ, et al.. Risk of celiac disease autoimmunity and timing of gluten introduction in the diet of infants at increased risk of disease. JAMA2005; 293(19):2343–2351. [PubMed] [Google Scholar]

10. Husby S, Koletzko S, Korponay-Szabó I, et al.. European Society Paediatric Gastroenterology, Hepatology and Nutrition guidelines for diagnosing coeliac disease 2020. J Pediatr Gastroenterol Nutr2020; 70(1):141–156. [PubMed] [Google Scholar]

11. Parzanese I, Qehajaj D, Patrinicola F, et al.. Celiac disease: From pathophysiology to treatment. World J Gastrointest Pathophysiol2017; 8(2):27. [PMC free article] [PubMed] [Google Scholar]

12. Elli L, Bascuñán K, di Lernia L, et al.. Safety of occasional ingestion of gluten in patients with celiac disease: A real-life study. BMC Medicine2020; 18:1–8. [PMC free article] [PubMed] [Google Scholar]

13. Tozzoli R, Bonaguri C, Melegari A, et al.. Current state of diagnostic technologies in the autoimmunology laboratory. Clin Chem Lab Med2013; 51(1):129–138. [PubMed] [Google Scholar]

14. Ciacci C, Zingone F.. New perspectives on the diagnosis of adulthood coeliac disease. In: Coeliac Disease and Gluten-Related Disorders. edn. Edited by ASD, Sanders., Oxford: Elsevier; 2022: 101–110. [Google Scholar]

15. Caio G, Volta U, Sapone A, et al.. Celiac disease: a comprehensive current review. BMC Medicine2019; 17:1–20. [PMC free article] [PubMed] [Google Scholar]

16. Ludvigsson JF, Murray JA. Epidemiology of celiac disease. Gastroenterol Clin2019; 48(1):1–18. [PubMed] [Google Scholar]

17. Mazzarella GJW, Jo GW. Effector and suppressor T cells in celiac disease. World J Gastroenterol2015; 21(24):7349. [PMC free article] [PubMed] [Google Scholar]

18. Barone MV, Troncone R, Auricchio S.. Gliadin peptides as triggers of the proliferative and stress/innate immune response of the celiac small intestinal mucosa. Int J Mol Sci2014; 15(11):20518–20537. [PMC free article] [PubMed] [Google Scholar]

19. Serena G, Lima R, Fasano A.. Genetic and environmental contributors for celiac disease. Curr Allergy Asthma Rep2019; 19:1–10. [PubMed] [Google Scholar]

20. Ting YT, Dahal-Koirala S, Kim HSK, et al.. A molecular basis for the T cell response in HLA-DQ2. 2 mediated celiac disease. Proc Natl Acad Sci2020; 117(6):3063–3073. [PMC free article] [PubMed] [Google Scholar]

21. Labruna G, Nanayakkara M, Pagliuca C, et al.. Celiac disease-associated Neisseria flavescens decreases mitochondrial respiration in CaCo-2 epithelial cells: Impact of Lactobacillus paracasei CBA L74 on bacterial-induced cellular imbalance. Cell Microbiol2019; 21(8):e13035. [PMC free article] [PubMed] [Google Scholar]

22. Lebwohl B, Green PHR. New developments in celiac disease. Gastroenterol Clin North Am2019; 48(1):xv–xvi. [PubMed] [Google Scholar]

23. Villella VR, Venerando A, Cozza G, et al.. A pathogenic role for cystic fibrosis transmembrane conductance regulator in celiac disease. EMBO J2019; 38(2):e100101. [PMC free article] [PubMed] [Google Scholar]

24. Izzo FC, Lodi GC, Vázquez de Ágredos Pascual ML. New insights into the composition of historical remedies and pharmaceutical formulations: The identification of natural resins and balsams by gas chromatographic-mass spectrometric investigations. Archaeol Anthropol Sci2021; 13:1–17. [Google Scholar]

25. Ben Houmich T, Admou B.. Celiac disease: Understandings in diagnostic, nutritional, and medicinal aspects. Int J Immunopathol Pharmacol2021; 35:20587384211008709. [PMC free article] [PubMed] [Google Scholar]

26. Leffler DA, Schuppan D.. Update on serologic testing in celiac disease. Off J Am Coll Gastroenterol2010; 105(12):2520–2524. [PubMed] [Google Scholar]

27. Husby S, Murray JA, Katzka DA. AGA clinical practice update on diagnosis and monitoring of celiac disease—changing utility of serology and histologic measures: expert review. Gastroenterol2019; 156(4):885–889. [PMC free article] [PubMed] [Google Scholar]

28. Grewal AK, Singh TG, Sharma D, et al.. Mechanistic insights and perspectives involved in neuroprotective action of quercetin. Biomed Pharmacother2021; 140:111729. [PubMed] [Google Scholar]

29. Bogaert L, Cauchie M, Van Hoovels L, et al.. Optimization of serologic diagnosis of celiac disease in the pediatric setting. Autoimmunity Rev2020; 19(5):102513. [PubMed] [Google Scholar]

30. Flores-Silva PC, Berrios JDJ, Pan J, et al.. Gluten-free spaghetti made with chickpea, unripe plantain and maize flours: functional and chemical properties and starch digestibility. Int J Food Sci Technol2014; 49(9):1985–1991. [Google Scholar]

31. Chauhan A, Saxena D, Singh S.. Total dietary fibre and antioxidant activity of gluten free cookies made from raw and germinated amaranth (Amaranthus spp.) flour. LWT Food Sci Technol2015; 63(2):939–945. [Google Scholar]

32. Khurana B, Lomash A, Khalil S, et al.. Evaluation of the impact of celiac disease and its dietary manipulation on children and their caregivers. Indian J Gastroenterol2015; 34:112–116. [PubMed] [Google Scholar]

33. Parvaiz M, Hussain K, Khalid S, et al.. A review: Medicinal importance of Glycyrrhiza glabra L.(Fabaceae family). Global J Pharmacol2014; 8(1):8–13. [Google Scholar]

34. Agarwal S, Kovilam O, Zach TL, et al.. Immunopathogenesis and therapeutic approaches in pediatric celiac disease. Expert Rev Clin Immunol2016; 12(8):857–869. [PMC free article] [PubMed] [Google Scholar]

35. Gismondi A, D’Agostino A, Di Marco G, et al.. Back to the roots: dental calculus analysis of the first documented case of coeliac disease. Archaeol Anthropol Sci2020; 12:1–10. [Google Scholar]

36. Thapa K, Khan H, Singh TG, et al.. Traumatic brain injury: Mechanistic insight on pathophysiology and potential therapeutic targets. J Mol Neurosci2021; 71(9):1725–1742. [PubMed] [Google Scholar]

37. Rashtak S, Murray JA. coeliac disease, new approaches to therapy. Aliment Pharmacol Ther2012; 35(7):768–781. [PMC free article] [PubMed] [Google Scholar]

38. Samaşca G, Sur G, Lupan I.. Current trends and investigative developments in celiac disease. Immunol Invest2013; 42(4):273–284. [PubMed] [Google Scholar]

39. Deng L, Wojciech L, Gascoigne NR, et al.. New insights into the interactions between Blastocystis, the gut microbiota, and host immunity. PLoS Pathogens2021; 17(2):e1009253. [PMC free article] [PubMed] [Google Scholar]

40. Kupper C.Dietary guidelines and implementation for celiac disease. Gastroenterol2005; 128(4):S121–S127. [PubMed] [Google Scholar]

41. Mohn A, Cerruto M, Iafusco D, et al.. Celiac disease in children and adolescents with type I diabetes: importance of hypoglycemia. J Pediatr Gastroenterol Nutr2001; 32(1):37–40. [PubMed] [Google Scholar]

42. Stern M, Cicl*tira PJ, van Eckert R, et al.. Analysis and clinical effects of gluten in coeliac disease. Eur J Gastroenterol Hepatol2001; 13(6):741–747. [PubMed] [Google Scholar]

43. Tosun MS, Ertekin V, Selimoglu MA. Autoimmune hepatitis associated with celiac disease in childhood. Eur J Gastroenterol Hepatol2010; 22(7):898–899. [PubMed] [Google Scholar]

44. Dias R, Pereira CB, Pérez-Gregorio R, et al.. Recent advances on dietary polyphenol’s potential roles in celiac disease. Trends Food Sci Technol2021; 107:213–225. [Google Scholar]

45. Someya N, Endo MY, f*ckuba Y, et al.. Blood flow responses in celiac and superior mesenteric arteries in the initial phase of digestion. Am J Physiol Regul Integr Comp Physiol2008; 294(6):R1790–R1796. [PubMed] [Google Scholar]

46. Daveson AJ, Jones DM, Gaze S, et al.. Effect of hookworm infection on wheat challenge in celiac disease–a randomised double-blinded placebo controlled trial. PloS ONE2011; 6(3):e17366. [PMC free article] [PubMed] [Google Scholar]

47. Auricchio R, Troncone R.. Can celiac disease be prevented?Front Immunol2021; 12:672148. [PMC free article] [PubMed] [Google Scholar]

48. Baranenko D, Bespalov V, Nadtochii L, et al.. Development of encapsulated extracts on the basis of meadowsweet (Filipendula ulmaria) in the composition of functional foods with oncoprotective properties. Agro Res2019; 17(5):1829–1838. [Google Scholar]

49. Edwards SE, Rocha da Costa I, Williamson EM, et al.. Phytopharmacy: An evidence-based guide to herbal medicinal products. Chicester: John Wiley & Sons; 2015. [Google Scholar]

50. El Mihyaoui A, Esteves da Silva JC, Charfi S, et al.. Chamomile (Matricaria chamomilla L.): A review of ethnomedicinal use, phytochemistry and pharmacological uses. Life2022; 12(4):479. [PMC free article] [PubMed] [Google Scholar]

51. Catani MV, Rinaldi F, Tullio V, et al.. Comparative analysis of phenolic composition of six commercially available chamomile (Matricaria chamomilla L.) extracts: Potential biological implications. Int J Mol Sci2021; 22(19):10601. [PMC free article] [PubMed] [Google Scholar]

52. Piknová Ľ, Brežná B, Kuchta T.. Detection of gluten-containing cereals in food by 5′-nuclease real-time polymerase chain reaction. J Food Nutr Res2008; 47(3):114–119. [Google Scholar]

53. de Sousa JS. Enzymatic treatment of celiac disease. J Pediatr Gastroenterol Nutr2004; 38(2):229. [PubMed] [Google Scholar]

54. Marti T, Molberg Ø, Li Q, et al.. Prolyl endopeptidase-mediated destruction of T cell epitopes in whole gluten: chemical and immunological characterization. J Pharmacol Exp Ther2005; 312(1):19–26. [PubMed] [Google Scholar]

55. Matuz J, Bartók T, Mórocz-Salamon K, et al.. Structure and potential allergenic character of cereal proteins I. Protein content and amino acid composition. Cereal Res Comm2000; 28:263–270. [Google Scholar]

56. Stepniak D, Spaenij-Dekking L, Mitea C, et al.. Highly efficient gluten degradation with a newly identified prolyl endoprotease: implications for celiac disease. Am J Physiol Gastroenterol Liver Physiol2006; 291(4):G621–G629. [PubMed] [Google Scholar]

57. Pastorino G, Cornara L, Soares S, et al.. Liquorice (Glycyrrhiza glabra): A phytochemical and pharmacological review. Phytother Res2018; 32(12):2323–2339. [PMC free article] [PubMed] [Google Scholar]

58. Karkanis A, Martins N, Petropoulos S, et al.. Phytochemical composition, health effects, and crop management of liquorice (Glycyrrhiza glabra L.): A medicinal plant. Food Rev Int2018; 34(2):182–203. [Google Scholar]

59. Gowthaman V, Sharma D, Biswas A, et al.. Liquorice (Glycyrrhiza glabra) herb as a poultry feed additive: A review. Lett Animal Biol2021; 1(2):14–20. [Google Scholar]

60. Stenman SM, Venäläinen JI, Lindfors K, et al.. Enzymatic detoxification of gluten by germinating wheat proteases: implications for new treatment of celiac disease. Ann Med2009; 41(5):390–400. [PubMed] [Google Scholar]

61. Tsai M-L, Wu C-T, Lin T-F, et al.. Chemical composition and biological properties of essential oils of two mint species. Trop J Pharm Res2013; 12(4):577–582. [Google Scholar]

62. Zhao H, Ren S, Yang H, et al.. Peppermint essential oil: Its phytochemistry, biological activity, pharmacological effect and application. Biomed Pharmacother2022; 154:113559. [PubMed] [Google Scholar]

63. Bai JC, Fried M, Corazza GR, et al.. World Gastroenterology Organisation global guidelines on celiac disease. J Clin Gastroenterol2013; 47(2):121–126. [PubMed] [Google Scholar]

64. Howlett K, Galbo H, Lorentsen J, et al.. Effect of adrenaline on glucose kinetics during exercise in adrenalectomised humans. J Physiol1999; 519(Pt 3):911. [PMC free article] [PubMed] [Google Scholar]

65. Kagnoff MF. Overview and pathogenesis of celiac disease. Gastroenterol2005; 128(4):S10–S18. [PubMed] [Google Scholar]

66. Khan RU, Fatima A, Naz S, et al.. Perspective, opportunities and challenges in using fennel (Foeniculum vulgare) in poultry health and production as an eco-friendly alternative to antibiotics: A review. Antibiotics2022; 11(2):278. [PMC free article] [PubMed] [Google Scholar]

67. Makharia GK. Current and emerging therapy for celiac disease. Front Med2014; 1:6. [PMC free article] [PubMed] [Google Scholar]

68. Pinier M, Fuhrmann G, Verdu E, et al.. Prevention measures and exploratory pharmacological treatments of celiac disease. Off J Am Coll Gastroenterol2010; 105(12):2551–2561. [PubMed] [Google Scholar]

69. Setty M, Hormaza L, Guandalini S.. Celiac disease: risk assessment, diagnosis, and monitoring. Mol Diagnos Ther2008; 12:289–298. [PubMed] [Google Scholar]

70. Braun L.Slippery elm. J Complement Med2006; 5(1). [Google Scholar]

71. Bonamico M, Thanasi E, Mariani P, et al.. Duodenal bulb biopsies in celiac disease: a multicenter study. J Pediatr Gastroenterol Nutr2008; 47(5):618–622. [PubMed] [Google Scholar]

72. Ciacci C, Maiuri L, Caporaso N, et al.. Celiac disease: in vitro and in vivo safety and palatability of wheat-free sorghum food products. Clin Nutr2007; 26(6):799–805. [PubMed] [Google Scholar]

73. Ludvigsson J, Green P.. Clinical management of coeliac disease. J Intern Med2011; 269(6):560–571. [PubMed] [Google Scholar]

74. Confino-Cohen R, Chodick G, Shalev V, et al.. Chronic urticaria and autoimmunity: associations found in a large population study. J Aller Clin Immunol2012; 129(5):1307–1313. [PubMed] [Google Scholar]

75. Lebreton C, Ménard S, Abed J, et al.. Interactions among secretory immunoglobulin A, CD71, and transglutaminase-2 affect permeability of intestinal epithelial cells to gliadin peptides. Gastroenterol2012; 143(3):698–707. [PubMed] [Google Scholar]

76. Mazumdar K, Alvarez X, Borda JT, et al.. Visualization of transepithelial passage of the immunogenic 33-residue peptide from α-2 gliadin in gluten-sensitive macaques. PloS ONE2010; 5(4):e10228. [PMC free article] [PubMed] [Google Scholar]

77. Pavone L, Fiumara A, Bottaro G, et al.. Autism and celiac disease: failure to validate the hypothesis that a link might exist. Biol Psychiat1997; 42(1):72–75. [PubMed] [Google Scholar]

78. Mukhtar M, Bilal M, Rahdar A, et al.. Nanomaterials for diagnosis and treatment of brain cancer: Recent updates. Chemosensors2020; 8(4):117. [Google Scholar]

79. Singh S, Singh TG. Role of nuclear factor kappa B (NF-κB) signalling in neurodegenerative diseases: an mechanistic approach. Current Neuropharmacol2020; 18(10):918–935. [PMC free article] [PubMed] [Google Scholar]

Articles from Narra J are provided here courtesy of Narra Sains Indonesia

Celiac disease: Pathogenesis, disease management and new insights into the herbal-based treatments (2024)
Top Articles
Latest Posts
Article information

Author: Rob Wisoky

Last Updated:

Views: 6551

Rating: 4.8 / 5 (68 voted)

Reviews: 83% of readers found this page helpful

Author information

Name: Rob Wisoky

Birthday: 1994-09-30

Address: 5789 Michel Vista, West Domenic, OR 80464-9452

Phone: +97313824072371

Job: Education Orchestrator

Hobby: Lockpicking, Crocheting, Baton twirling, Video gaming, Jogging, Whittling, Model building

Introduction: My name is Rob Wisoky, I am a smiling, helpful, encouraging, zealous, energetic, faithful, fantastic person who loves writing and wants to share my knowledge and understanding with you.