1
|
Feng J, Jia Y, Xu B, Bi X, Ge Z, Ma G, Xie Y, Wang C, Ma D. Quantitative proteomic analysis for characterization of protein components related to dough quality and celiac disease in wheat flour, dough, and heat-treated dough. Food Chem 2024; 461:140924. [PMID: 39181042 DOI: 10.1016/j.foodchem.2024.140924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/23/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
High-sensitivity 4D label-free proteomic technology was used to identify protein components related to gluten quality and celiac disease (CD) in strong-gluten wheat cultivar KX 3302 and medium-gluten wheat cultivar BN 207. The highly expressed storage protein components in KX3302 were high-molecular-weight-glutenin-subunits (HMW-GSs), α-gliadin, and globulin, whereas those in BN207 were γ-gliadin, low-molecular-weight-glutenin-subunits (LMW-GSs) and avenin-like proteins. In addition, BN207 had more upregulated metabolic proteins than KX3302. The abundance of storage proteins increased during dough formation. After heat treatment, the upregulated proteins accounted for 57.53 % of the total proteins, but the downregulated storage proteins accounted for 79.34 % of the total storage proteins. In cultivar KX3302, CD proteins mainly included α-gliadin and HMW-GSs, whereas in BN207, they were mainly γ-gliadin and LMW-GSs. Thermal treatment significantly reduces the expression levels of CD-related proteins. These findings provide a new perspective on reducing the content of CD-related proteins in wheat products.
Collapse
Affiliation(s)
- Jianchao Feng
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Yuku Jia
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Beiming Xu
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Xintong Bi
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Zifei Ge
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Geng Ma
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; The National Key Laboratory of Wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou 450046, China
| | - Yingxin Xie
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Chenyang Wang
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; The National Key Laboratory of Wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China
| | - Dongyun Ma
- College of Agronomy/National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450046, China; The National Key Laboratory of Wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou 450046, China; Technology Innovation Center of Henan Wheat, Henan Agricultural University, Zhengzhou 450046, China.
| |
Collapse
|
2
|
Goudarzi H, Ikeda A, Bamai YA, Yokota I, Miyashita C, Karmaus W, Kishi R, Konno S. Impact of wheat sensitization on wheeze and T2 phenotypes in general population of children. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100300. [PMID: 39170911 PMCID: PMC11338083 DOI: 10.1016/j.jacig.2024.100300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/24/2024] [Accepted: 05/16/2024] [Indexed: 08/23/2024]
Abstract
Background The association between sensitization to specific aeroallergens and outcomes in patients with asthma is well researched; however, the association between childhood-onset wheeze/asthma and sensitization to various aeroallergens and food allergens in the general pediatric population remains poorly understood. Objective We sought to investigate the association between sensitization to common aeroallergens and food allergens with wheeze and type 2 (T2) inflammation in the general pediatric population. Methods Specific IgEs against 9 aeroallergens and 4 food allergens were measured in the prospective Hokkaido birth cohort of 428 school-age children (age ∼10 years). Wheeze and other allergic symptoms were assessed using the International Study of Asthma and Allergies in Childhood questionnaire. Blood eosinophil count and fractional exhaled nitric oxide level were assessed as T2 biomarkers. The Isle of Wight birth cohort in the United Kingdom was used for replication analysis (n = 1032). Results The prevalence of sensitization to at least 1 aeroallergen and food allergen was 70.5% and 22.3%, respectively. A significant association between wheeze and sensitization to aeroallergens such as ragweed, Japanese cedar, mugwort, and pet dander was found. However, the association between wheeze and wheat sensitization was highly significant (Hokkaido birth cohort: odds ratio, 4.67; 95% CI, 1.98-11.01; Isle of Wight birth cohort, odds ratio, 4.01; 95% CI, 1.78-9.07). Sensitization to most aeroallergens, though not any food allergen, was associated with the T2-high phenotype. Conclusions Sensitization to wheat may be an important risk factor for wheeze/asthma development, especially the pathogenesis of T2-non/low asthma, independent of aeroallergens, in the general pediatric population.
Collapse
Affiliation(s)
- Houman Goudarzi
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Hokkaido University Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Atsuko Ikeda
- Hokkaido University Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yu Ait Bamai
- Hokkaido University Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Chihiro Miyashita
- Hokkaido University Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, Tenn
| | - Reiko Kishi
- Hokkaido University Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
3
|
Sharma L, Rahman F, Sharma RA. The emerging role of biotechnological advances and artificial intelligence in tackling gluten sensitivity. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39145745 DOI: 10.1080/10408398.2024.2392158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Gluten comprises an intricate network of hundreds of related but distinct proteins, mainly "gliadins" and "glutenins," which play a vital role in determining the rheological properties of wheat dough. However, ingesting gluten can trigger severe conditions in susceptible individuals, including celiac disease, wheat allergy, or non-celiac gluten sensitivity, collectively known as gluten-related disorders. This review provides a panoramic view, delving into the various aspects of gluten-triggered disorders, including symptoms, diagnosis, mechanism, and management. Though a gluten-free diet remains the primary option to manage gluten-related disorders, the emerging microbial and plant biotechnology tools are playing a transformative role in reducing the immunotoxicity of gluten. The enzymatic hydrolysis of gluten and the development of gluten-reduced/free wheat lines using RNAi and CRISPR/Cas technology are laying the foundation for creating safer wheat products. In addition to biotechnological interventions, the emerging artificial intelligence technologies are also bringing about a paradigm shift in the diagnosis and management of gluten-related disorders. Here, we provide a comprehensive overview of the latest developments and the potential these technologies hold for tackling gluten sensitivity.
Collapse
Affiliation(s)
- Lakshay Sharma
- Department of Biological Sciences, Birla Institute of Technology & Science Pilani (BITS Pilani), Pilani, India
| | - Farhanur Rahman
- Department of Biological Sciences, Birla Institute of Technology & Science Pilani (BITS Pilani), Pilani, India
| | - Rita A Sharma
- Department of Biological Sciences, Birla Institute of Technology & Science Pilani (BITS Pilani), Pilani, India
- National Agri-Food Biotechnology Institute (NABI), Mohali, India
| |
Collapse
|
4
|
Wall E, Semrad CE. Celiac Disease, Gluten Sensitivity, and Diet Management. Curr Gastroenterol Rep 2024; 26:191-199. [PMID: 38865028 DOI: 10.1007/s11894-024-00931-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE OF REVIEW Celiac disease is a common chronic inflammatory condition of the small bowel triggered by gluten in wheat, rye and barley in the diet. Non-celiac gluten sensitivity presents with symptoms similar to celiac disease with the ingestion of gluten or other components of wheat. In this article, we review challenges presented by a gluten free diet for the treatment of both disorders. RECENT FINDINGS Wheat is ubiquitous in the diet and medications/products. A registered dietitian is mandatory for patient education on the gluten free diet. Naturally gluten free foods provide a healthy diet for those with celiac disease. Whole grains labelled gluten free, including oats, are encouraged in the diet as refined grains may be deficient in fiber, protein, and micronutrients, particularly folate. Gluten contamination is the most common cause of persistent symptoms in celiac disease though shared equipment of food preparation may not be as large a problem as suspected. Most with celiac disease on a gluten free diet will fully recover and gain weight that poses a problem for those overweight to start. The gluten free diet may have a negative impact on quality of life for both celiac patients and their families. Those with hypervigilance of the gluten free diet and avoidance of dining out have the lowest quality of life. The gluten free diet is currently the only effective treatment for celiac disease. A registered dietitian is needed to educate patients on the complexity of the gluten free diet with a goal of healthy eating, maintaining a healthy weight, and avoiding disordered eating or diet hypervigilance; key to a good quality of life.
Collapse
Affiliation(s)
- Elizabeth Wall
- Clinical Nutrition, University of Chicago Medicine, Chicago, IL, USA
| | - Carol E Semrad
- Section of Gastroenterology, Hepatology and Nutrition, University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
5
|
Munteanu C, Schwartz B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int J Mol Sci 2024; 25:8250. [PMID: 39125822 PMCID: PMC11311432 DOI: 10.3390/ijms25158250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The intricate relationship between the gastrointestinal (GI) microbiome and the progression of chronic non-communicable diseases underscores the significance of developing strategies to modulate the GI microbiota for promoting human health. The administration of probiotics and prebiotics represents a good strategy that enhances the population of beneficial bacteria in the intestinal lumen post-consumption, which has a positive impact on human health. In addition, dietary fibers serve as a significant energy source for bacteria inhabiting the cecum and colon. Research articles and reviews sourced from various global databases were systematically analyzed using specific phrases and keywords to investigate these relationships. There is a clear association between dietary fiber intake and improved colon function, gut motility, and reduced colorectal cancer (CRC) risk. Moreover, the state of health is reflected in the reciprocal and bidirectional relationships among food, dietary antioxidants, inflammation, and body composition. They are known for their antioxidant properties and their ability to inhibit angiogenesis, metastasis, and cell proliferation. Additionally, they promote cell survival, modulate immune and inflammatory responses, and inactivate pro-carcinogens. These actions collectively contribute to their role in cancer prevention. In different investigations, antioxidant supplements containing vitamins have been shown to lower the risk of specific cancer types. In contrast, some evidence suggests that taking antioxidant supplements can increase the risk of developing cancer. Ultimately, collaborative efforts among immunologists, clinicians, nutritionists, and dietitians are imperative for designing well-structured nutritional trials to corroborate the clinical efficacy of dietary therapy in managing inflammation and preventing carcinogenesis. This review seeks to explore the interrelationships among dietary antioxidants, dietary fiber, and the gut microbiome, with a particular focus on their potential implications in inflammation and cancer.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
6
|
Lange S, Tsohataridis S, Boland N, Ngo L, Hahad O, Münzel T, Wild P, Daiber A, Schuppan D, Lurz P, Keppeler K, Steven S. Effects of Short-Term Gluten-Free Diet on Cardiovascular Biomarkers and Quality of Life in Healthy Individuals: A Prospective Interventional Study. Nutrients 2024; 16:2265. [PMID: 39064708 PMCID: PMC11279490 DOI: 10.3390/nu16142265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION The exposome concept includes nutrition as it significantly influences human health, impacting the onset and progression of diseases. Gluten-containing wheat products are an essential source of energy for the world's population. However, a rising number of non-celiac healthy individuals tend to reduce or completely avoid gluten-containing cereals for health reasons. AIM AND METHODS This prospective interventional human study aimed to investigate whether short-term gluten avoidance improves cardiovascular endpoints and quality of life (QoL) in healthy volunteers. A cohort of 27 participants followed a strict gluten-free diet (GFD) for four weeks. Endothelial function measured by flow-mediated vasodilation (FMD), blood testing, plasma proteomics (Olink®) and QoL as measured by the World Health Organisation Quality-of-Life (WHOQOL) survey were investigated. RESULTS GFD resulted in decreased leucocyte count and C-reactive protein levels along with a trend of reduced inflammation biomarkers determined by plasma proteomics. A positive trend indicated improvement in FMD, whereas other cardiovascular endpoints remained unchanged. In addition, no improvement in QoL was observed. CONCLUSION In healthy individuals, a short-term GFD demonstrated anti-inflammatory effects but did not result in overall cardiovascular improvement or enhanced quality of life.
Collapse
Affiliation(s)
- Simon Lange
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Simeon Tsohataridis
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Niklas Boland
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Lisa Ngo
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Omar Hahad
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Philipp Wild
- Department of Preventive Cardiology and Medical Prevention, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Andreas Daiber
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Philipp Lurz
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Karin Keppeler
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Sebastian Steven
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
- Department of Cardiology, Goethe University Frankfurt, 60596 Main, Germany
| |
Collapse
|
7
|
Spertino M, Elisa RR, Pugliese N. Letter: Breaking new ground-Understanding the role of ATI-free diet in managing MASLD. Aliment Pharmacol Ther 2024; 60:302-303. [PMID: 38860611 DOI: 10.1111/apt.18064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
LINKED CONTENTThis article is linked to Armandi et al papers. To view these articles, visit https://doi.org/10.1111/apt.17941 and https://doi.org/10.1111/apt.18119
Collapse
Affiliation(s)
- Matteo Spertino
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
| | - Rossi Roberta Elisa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Gastroenterology and Endoscopy Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Nicola Pugliese
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
| |
Collapse
|
8
|
Galipeau HJ, Hinterleitner R, Leonard MM, Caminero A. Non-Host Factors Influencing Onset and Severity of Celiac Disease. Gastroenterology 2024; 167:34-50. [PMID: 38286392 DOI: 10.1053/j.gastro.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts; Center for Celiac Research and Treatment, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
9
|
Kranidioti H, Deutsch M. Editorial: Can dietary gluten restriction improve hepatic steatosis? Aliment Pharmacol Ther 2024; 59:1620-1621. [PMID: 38643496 DOI: 10.1111/apt.17969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
LINKED CONTENTThis article is linked to Armandi et al papers. To view these articles, visit https://doi.org/10.1111/apt.17941 and https://doi.org/10.1111/apt.18025.
Collapse
Affiliation(s)
- Hariklia Kranidioti
- 2nd Academic Department of Internal Medicine, General Hospital of Athens "Hippocration", National and Kapodistrian University of Athens, Athens, Greece
| | - Melanie Deutsch
- 2nd Academic Department of Internal Medicine, General Hospital of Athens "Hippocration", National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Alborzi Avanaki F, Ebrahimi Daryani N, Aletaha N, Hesabgar N, Rezaee-Zavareh MS, Hadi R. Short-term effect of gluten-free diet on disease severity, quality of life, and inflammatory markers among patients with mild to moderate ulcerative colitis: A triple-blind randomized placebo-controlled trial. Arab J Gastroenterol 2024:S1687-1979(24)00009-1. [PMID: 38714473 DOI: 10.1016/j.ajg.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 01/06/2024] [Accepted: 01/20/2024] [Indexed: 05/10/2024]
Abstract
BACKGROUND AND STUDY AIMS Diet is an important underlying factor in ulcerative colitis (UC) disease. The present study aimed to investigate the effect of a gluten-free diet (GFD) on disease severity, quality of life, and inflammatory markers in patients with UC. PATIENTS AND METHODS In this triple-blind randomized placebo-controlled clinical trial, we evaluated the effect of a GFD on the erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), fecal calprotectin, disease severity, and quality of life in patients with mild to moderate UC. Patients' quality of life and severity of symptoms were evaluated using the Inflammatory Bowel Disease Questionnaire (IBDQ) and Simple Clinical Colitis Activity Index (SCCAI), respectively. Patients received this regimen for six weeks and were evaluated before and after the intervention. RESULTS The mean age of patients (n = 26) was 39.31 years (standard deviation = 9.34). In both study groups, the mean ESR, CRP, IBDQ, and SCCAI showed no statistically significant improvement with the dietary intervention. Fecal calprotectin was increased in both groups without statistical significance. CONCLUSIONS We could not find any significant effect of GFD on inflammatory markers, quality of life, and disease severity among patients with mild to moderate UC. It is too early to suggest the gluten-free diet as a safe and beneficial regimen for UC patients. There is a need for further investigations with larger sample sizes and longer follow-ups as clinical trials and cohort studies to obtain more reliable results.
Collapse
Affiliation(s)
- Foroogh Alborzi Avanaki
- Department of Gastroenterology and Hepatology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Naser Ebrahimi Daryani
- Department of Gastroenterology and Hepatology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Aletaha
- Department of Gastroenterology and Hepatology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Hesabgar
- Department of Gastroenterology and Hepatology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Reza Hadi
- Department of Gastroenterology and Hepatology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Armandi A, Bespaljko H, Mang A, Huber Y, Michel M, Labenz C, Galle PR, Neerukonda M, Bugianesi E, Schuppan D, Schattenberg JM. Short-term reduction of dietary gluten improves metabolic-dysfunction associated steatotic liver disease: A randomised, controlled proof-of-concept study. Aliment Pharmacol Ther 2024; 59:1212-1222. [PMID: 38462919 DOI: 10.1111/apt.17941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/24/2023] [Accepted: 02/26/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND The current management of metabolic dysfunction-associated steatotic liver disease (MASLD) relies on lifestyle intervention. Prior studies have shown that nutritional wheat amylase trypsin inhibitors (ATI) activate toll-like receptor 4 on intestinal myeloid cells to enhance intestinal and extra-intestinal inflammation, including the promotion of murine MASLD, insulin resistance and liver fibrosis. AIMS We aimed to assess the impact of ATI (gluten)-free diet in liver as well as metabolic parameters of biopsy-proven MASLD patients. METHODS We performed a 6-week, proof-of-concept 1:1 randomised controlled trial of an ATI-free diet. The controls followed a balanced diet recommended by the German Nutrition Society. We assessed changes in controlled attenuation parameter (CAP), body mass index (BMI) and homeostatic model assessment of insulin resistance (HOMA-IR). Patient-reported outcomes were assessed by the CLDQ-NASH questionnaire. Forty-five patients were consecutively enrolled (21 in the intervention arm and 24 in the control arm). RESULTS Three patients from each arm discontinued the study. In the ATI-free diet group, a significant decrease in BMI (p = 0.018), CAP (p = 0.018) and HOMA-IR (p = 0.042) was observed at 6 weeks. The mean difference in CAP between the two arms at week 6 was 30.5 dB/m (p = 0.039), with a delta significantly higher in the ATI-free diet group (p = 0.043). Only an ATI-free diet could achieve a significant improvement in CLDQ-NASH domains (p value for total scoring: 0.013). CONCLUSIONS A short-term ATI-free diet leads to significant improvements in liver and metabolic parameters, as well as patient-reported outcomes with good tolerability. A larger follow-up study is justified to corroborate these findings. CLINICAL TRIAL NUMBER NCT04066400.
Collapse
Affiliation(s)
- Angelo Armandi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, University of Turin, Turin, Italy
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Helena Bespaljko
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Alexander Mang
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Yvonne Huber
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Maurice Michel
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Christian Labenz
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Peter R Galle
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Manjusha Neerukonda
- University Medical Center, Institute of Translational Immunology and Research Center for Immunotherapy, Johannes Gutenberg-University, Mainz, Germany
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, University of Turin, Turin, Italy
| | - Detlef Schuppan
- University Medical Center, Institute of Translational Immunology and Research Center for Immunotherapy, Johannes Gutenberg-University, Mainz, Germany
- Beth Israel Deaconess Medical Center, Division of Gastroenterology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jörn M Schattenberg
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Department of Internal Medicine II, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
12
|
Shea Z, Ogando do Granja M, Fletcher EB, Zheng Y, Bewick P, Wang Z, Singer WM, Zhang B. A Review of Bioactive Compound Effects from Primary Legume Protein Sources in Human and Animal Health. Curr Issues Mol Biol 2024; 46:4203-4233. [PMID: 38785525 PMCID: PMC11120442 DOI: 10.3390/cimb46050257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
The global demand for sustainable and nutritious food sources has catalyzed interest in legumes, known for their rich repertoire of health-promoting compounds. This review delves into the diverse array of bioactive peptides, protein subunits, isoflavones, antinutritional factors, and saponins found in the primary legume protein sources-soybeans, peas, chickpeas, and mung beans. The current state of research on these compounds is critically evaluated, with an emphasis on the potential health benefits, ranging from antioxidant and anticancer properties to the management of chronic diseases such as diabetes and hypertension. The extensively studied soybean is highlighted and the relatively unexplored potential of other legumes is also included, pointing to a significant, underutilized resource for developing health-enhancing foods. The review advocates for future interdisciplinary research to further unravel the mechanisms of action of these bioactive compounds and to explore their synergistic effects. The ultimate goal is to leverage the full spectrum of benefits offered by legumes, not only to advance human health but also to contribute to the sustainability of food systems. By providing a comprehensive overview of the nutraceutical potential of legumes, this manuscript sets a foundation for future investigations aimed at optimizing the use of legumes in the global pursuit of health and nutritional security.
Collapse
Affiliation(s)
- Zachary Shea
- United States Department of Agriculture–Agricultural Research Service, Raleigh Agricultural Research Station, Raleigh, NC 27606, USA;
| | - Matheus Ogando do Granja
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (M.O.d.G.); (E.B.F.); (Y.Z.); (P.B.); (Z.W.)
| | - Elizabeth B. Fletcher
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (M.O.d.G.); (E.B.F.); (Y.Z.); (P.B.); (Z.W.)
| | - Yaojie Zheng
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (M.O.d.G.); (E.B.F.); (Y.Z.); (P.B.); (Z.W.)
| | - Patrick Bewick
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (M.O.d.G.); (E.B.F.); (Y.Z.); (P.B.); (Z.W.)
| | - Zhibo Wang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (M.O.d.G.); (E.B.F.); (Y.Z.); (P.B.); (Z.W.)
- Donald Danforth Plant Science Center, Olivette, MO 63132, USA
| | - William M. Singer
- Center for Advanced Innovation in Agriculture, Virginia Tech, Blacksburg, VA 24061, USA;
| | - Bo Zhang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (M.O.d.G.); (E.B.F.); (Y.Z.); (P.B.); (Z.W.)
| |
Collapse
|
13
|
Müller I, Schmid B, Bosa L, Morlock GE. Screening of α-amylase/trypsin inhibitor activity in wheat, spelt and einkorn by high-performance thin-layer chromatography. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024. [PMID: 38687148 DOI: 10.1039/d4ay00402g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
α-Amylase/trypsin inhibitor proteins (ATI) are discussed as possible triggers for non-celiac gluten sensitivity. The potential of high-performance thin-layer chromatography (HPTLC) was studied for the first time to analyse the inhibitory properties of ATIs from flour of wheat, spelt, and einkorn. Inhibition by each flour of the digestive enzymes trypsin or α-amylase was determined by the reduction of released metabolisation products in comparison to non-digested flour, and positive (acarbose) and negative (water) controls. Firstly, amylolysis was carried out in miniaturized form on the HPTLC surface (HPTLC-nanoGIT) after in-vial pre-incubation of the amylase with the inhibitors from flour. α-Amylase inhibition was evident via the reduction of released saccharides, as analysed by normal phase HPTLC. A strong influence of the flour matrix on the assay results (individual saccharides) was evident, caused by an increased amylolysis of further polysaccharides present, making HPTLC analysis more reliable than currently used spectrophotometric sum value assays. The detection and visualization of such matrix influence helps to understand the problems associated with spectrophotometric assays. Only maltotriose was identified as a reliable marker of the amylolysis. The highest α-amylase inhibition and thus the lowest saccharide response was detected for maltotriose in refined spelt, whereas the lowest α-amylase inhibition and thus the highest saccharide response was detected for maltotriose in refined wheat. A comparison of refined and whole grain flours showed no clear trend in the responses. Secondly, trypsin inhibition and proteolysis were performed in-vial, and any inhibition was evident via the reduction of released peptides, analysed by reversed-phase HPTLC. Based on the product pattern of the proteolysis, einkorn and whole wheat showed the highest trypsin inhibition, whereas refined wheat and refined spelt showed the lowest inhibition. Advantageously, HPTLC analysis provided important information on changes in individual saccharides or peptides, which was more reliable and sustainable than spectrophotometric in-vial assays (only sum value) or liquid column chromatography analysis (targeting only the ATI proteins).
Collapse
Affiliation(s)
- Isabel Müller
- Chair of Food Science, Institute of Nutritional Science, Interdisciplinary Research Centre for Biosystems, Land Use and Nutrition, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany.
| | - Bianca Schmid
- Chair of Food Science, Institute of Nutritional Science, Interdisciplinary Research Centre for Biosystems, Land Use and Nutrition, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany.
| | - Loredana Bosa
- Chair of Food Science, Institute of Nutritional Science, Interdisciplinary Research Centre for Biosystems, Land Use and Nutrition, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany.
| | - Gertrud Elisabeth Morlock
- Chair of Food Science, Institute of Nutritional Science, Interdisciplinary Research Centre for Biosystems, Land Use and Nutrition, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany.
| |
Collapse
|
14
|
Wang Y, Shi D, Zou W, Jiang Y, Wang T, Chen X, Ma C, Li W, Chen T, Burrows JF, Wang L, Zhou M. An Effective Modification Strategy to Build Multifunctional Peptides Based on a Trypsin Inhibitory Peptide of the Kunitz Family. Pharmaceutics 2024; 16:597. [PMID: 38794259 PMCID: PMC11125039 DOI: 10.3390/pharmaceutics16050597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Peptides with antimicrobial activity or protease inhibitory activity are potential candidates to supplement traditional antibiotics or cancer chemotherapies. However, the potential of many peptides are limited by drawbacks such as cytotoxicity or susceptibility to hydrolysis. Therefore, strategies to modify the structure of promising peptides may represent an effective approach for developing more promising clinical candidates. In this study, the mature peptide OSTI-1949, a Kunitz-type inhibitor from Odorrana schmackeri, and four designed analogues were successfully synthesised. In contrast to the parent peptide, the analogues showed impressive multi-functionality including antimicrobial, anticancer, and trypsin inhibitory activities. In terms of safety, there were no obvious changes observed in the haemolytic activity at the highest tested concentration, and the analogue OSTI-2461 showed an increase in activity against cancer cell lines without cytotoxicity to normal cells (HaCaT). In summary, through structural modification of a natural Kunitz-type peptide, the biological activity of analogues was improved whilst retaining low cytotoxicity. The strategy of helicity enhancement by forming an artificial α-helix and ß-sheet structure provides a promising way to develop original bioactive peptides for clinical therapeutics.
Collapse
Affiliation(s)
- Ying Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Daning Shi
- Chinese Academy of Agricultural Sciences, No.12 Zhongguancun South Street, Haidian District, Beijing 100081, China;
| | - Wanchen Zou
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yangyang Jiang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Tao Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Xiaoling Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Chengbang Ma
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Tianbao Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - James F. Burrows
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Lei Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Mei Zhou
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| |
Collapse
|
15
|
San Mauro Martín I, López Oliva S, Garicano Vilar E, Sánchez Niño GM, Penadés BF, Terrén Lora A, Sanz Rojo S, Collado Yurrita L. Effects of Gluten on Gut Microbiota in Patients with Gastrointestinal Disorders, Migraine, and Dermatitis. Nutrients 2024; 16:1228. [PMID: 38674918 PMCID: PMC11053402 DOI: 10.3390/nu16081228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
As gluten may trigger gastrointestinal disorders (GIDs), its presence or absence in the diet can change the diversity and proportion of gut microbiota. The effects of gluten after six weeks of a double-blind, placebo-controlled intervention with a gluten-free diet (GFD) were studied in participants with GIDs suffering from migraines and atopic dermatitis (n = 46). Clinical biomarkers, digestive symptoms, stool, the Migraine Disability Assessment questionnaire, and zonulin levels were analyzed. Next-generation sequencing was used to amplify the 16S rRNA gene of bacteria and the internal transcribed spacer (ITS) regions of fungi. The GFD increased Chao1 fungal diversity after the intervention, while the fungal composition showed no changes. Bacterial diversity and composition remained stable, but a positive association between bacterial and fungal Chao1 diversity and a negative association between Dothideomycetes and Akkermansia were observed. GIDs decreased in both groups and migraines improved in the placebo group. Our findings may aid the development of GID treatment strategies.
Collapse
Affiliation(s)
- Ismael San Mauro Martín
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | - Sara López Oliva
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | - Elena Garicano Vilar
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | - Guerthy Melissa Sánchez Niño
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | - Bruno F. Penadés
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | - Ana Terrén Lora
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | - Sara Sanz Rojo
- Research Centers in Nutrition and Health (CINUSA Group), Paseo de la Habana 43, 28036 Madrid, Spain; (I.S.M.M.); (S.L.O.); (E.G.V.); (G.M.S.N.); (B.F.P.); (A.T.L.); (S.S.R.)
| | | |
Collapse
|
16
|
Jansson-Knodell CL, Rubio-Tapia A. Gluten-related Disorders From Bench to Bedside. Clin Gastroenterol Hepatol 2024; 22:693-704.e1. [PMID: 37879521 DOI: 10.1016/j.cgh.2023.09.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/27/2023]
Abstract
Celiac disease, non-celiac gluten sensitivity, and wheat allergy comprise 3 of the main conditions with wheat- and gluten-containing foods as the symptom trigger. Distinguishing between these entities can be daunting. In this review, we compare and contrast celiac disease, non-celiac gluten sensitivity, and wheat allergy to allow clinicians to determine which diagnosis fits their patient to facilitate high-quality management and longitudinal care.
Collapse
Affiliation(s)
- Claire L Jansson-Knodell
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - Alberto Rubio-Tapia
- Celiac Disease Program, Division of Gastroenterology, Hepatology, and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
17
|
de Graaf MC, Timmers E, Bonekamp B, van Rooy G, Witteman BJ, Shewry PR, Lovegrove A, America AH, Gilissen LJ, Keszthelyi D, Brouns FJ, Jonkers DMAE. Two randomized crossover multicenter studies investigating gastrointestinal symptoms after bread consumption in individuals with noncoeliac wheat sensitivity: do wheat species and fermentation type matter? Am J Clin Nutr 2024; 119:896-907. [PMID: 38373694 DOI: 10.1016/j.ajcnut.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Many individuals reduce their bread intake because they believe wheat causes their gastrointestinal (GI) symptoms. Different wheat species and processing methods may affect these responses. OBJECTIVES We investigated the effects of 6 different bread types (prepared from 3 wheat species and 2 fermentation conditions) on GI symptoms in individuals with self-reported noncoeliac wheat sensitivity (NCWS). METHODS Two parallel, randomized, double-blind, crossover, multicenter studies were conducted. NCWS individuals, in whom coeliac disease and wheat allergy were ruled out, received 5 slices of yeast fermented (YF) (study A, n = 20) or sourdough fermented (SF) (study B, n = 20) bread made of bread wheat, spelt, or emmer in a randomized order on 3 separate test days. Each test day was preceded by a run-in period of 3 d of a symptom-free diet and separated by a wash-out period of ≥7 d. GI symptoms were evaluated by change in symptom score (test day minus average of the 3-d run-in period) on a 0-100 mm visual analogue scale (ΔVAS), comparing medians using the Friedman test. Responders were defined as an increase in ΔVAS of ≥15 mm for overall GI symptoms, abdominal discomfort, abdominal pain, bloating, and/or flatulence. RESULTS GI symptoms did not differ significantly between breads of different grains [YF bread wheat median ΔVAS 10.4 mm (IQR 0.0-17.8 mm), spelt 4.9 mm (-7.6 to 9.4 mm), emmer 11.0 mm (0.0-21.3 mm), P = 0.267; SF bread wheat 10.5 mm (-3.1 to 31.5 mm), spelt 11.3 mm (0.0-15.3 mm), emmer 4.0 mm (-2.9 to 9.3 mm), P = 0.144]. The number of responders was also comparable for both YF (6 to wheat, 5 to spelt, and 7 to emmer, P = 0.761) and SF breads (9 to wheat, 7 to spelt, and 8 to emmer, P = 0.761). CONCLUSIONS The majority of NCWS individuals experienced some GI symptoms for ≥1 of the breads, but on a group level, no differences were found between different grains for either YF or SF breads. CLINICAL TRIAL REGISTRY clinicaltrials.gov, NCT04084470 (https://classic. CLINICALTRIALS gov/ct2/show/NCT04084470).
Collapse
Affiliation(s)
- Marlijne Cg de Graaf
- Department of Gastroenterology-Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Emma Timmers
- Department of Gastroenterology-Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Bo Bonekamp
- Department of Gastroenterology-Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Gonny van Rooy
- Department of Gastroenterology-Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Ben Jm Witteman
- Division Gastroenterology-Hepatology, Gelderse Vallei Hospital, Ede, The Netherlands; Division of Human Nutrition, Wageningen University & Research, Wageningen, The Netherlands
| | | | | | - Antoine Hp America
- Business Unit Bioscience, Plant Sciences Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Luud Jwj Gilissen
- Plant Breeding, Wageningen University & Research, Wageningen, The Netherlands
| | - Daniel Keszthelyi
- Department of Gastroenterology-Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Fred Jph Brouns
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands; Department of Human Biology, Maastricht University, Maastricht, The Netherlands
| | - Daisy M A E Jonkers
- Department of Gastroenterology-Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands.
| |
Collapse
|
18
|
Manza F, Lungaro L, Costanzini A, Caputo F, Volta U, De Giorgio R, Caio G. Gluten and Wheat in Women's Health: Beyond the Gut. Nutrients 2024; 16:322. [PMID: 38276560 PMCID: PMC10820448 DOI: 10.3390/nu16020322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 01/27/2024] Open
Abstract
Since the rise of awareness of gluten/wheat-related disorders in the academic and clinical field in the last few decades, misinformation regarding the gluten-free diet (GFD) and its impact on health has been spreading among the general population. Despite the established link between gluten and celiac disease (CD), where a GFD is mandatory to reach clinical and histological remission, things are more complicated when it comes to non-celiac gluten/wheat sensitivity (NCGWS) and other autoimmune/dysimmune disorders. In the last conditions, a beneficial effect of gluten withdrawal has not been properly assessed, but still is often suggested without strong supporting evidence. In this context, women have always been exposed, more than men, to higher social pressure related to nutritional behaviors and greater engagement in controlling body weight. With this narrative review, we aim to summarize current evidence on the adherence to a GFD, with particular attention to the impact on women's health.
Collapse
Affiliation(s)
- Francesca Manza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (F.C.); (R.D.G.)
| | - Lisa Lungaro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (F.C.); (R.D.G.)
| | - Anna Costanzini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (F.C.); (R.D.G.)
| | - Fabio Caputo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (F.C.); (R.D.G.)
| | - Umberto Volta
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (F.C.); (R.D.G.)
| | - Giacomo Caio
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (F.C.); (R.D.G.)
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital—Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
19
|
Zevallos VF, Yogev N, Hauptmann J, Nikolaev A, Pickert G, Heib V, Fittler N, Steven S, Luessi F, Neerukonda M, Janoschka C, Tobinski AM, Klotz L, Waisman A, Schuppan D. Dietary wheat amylase trypsin inhibitors exacerbate CNS inflammation in experimental multiple sclerosis. Gut 2023; 73:92-104. [PMID: 37595983 PMCID: PMC10715558 DOI: 10.1136/gutjnl-2023-329562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
OBJECTIVE Wheat has become a main staple globally. We studied the effect of defined pro-inflammatory dietary proteins, wheat amylase trypsin inhibitors (ATI), activating intestinal myeloid cells via toll-like receptor 4, in experimental autoimmune encephalitis (EAE), a model of multiple sclerosis (MS). DESIGN EAE was induced in C57BL/6J mice on standardised dietary regimes with defined content of gluten/ATI. Mice received a gluten and ATI-free diet with defined carbohydrate and protein (casein/zein) content, supplemented with: (a) 25% of gluten and 0.75% ATI; (b) 25% gluten and 0.19% ATI or (c) 1.5% purified ATI. The effect of dietary ATI on clinical EAE severity, on intestinal, mesenteric lymph node, splenic and central nervous system (CNS) subsets of myeloid cells and lymphocytes was analysed. Activation of peripheral blood mononuclear cells from patients with MS and healthy controls was compared. RESULTS Dietary ATI dose-dependently caused significantly higher EAE clinical scores compared with mice on other dietary regimes, including on gluten alone. This was mediated by increased numbers and activation of pro-inflammatory intestinal, lymph node, splenic and CNS myeloid cells and of CNS-infiltrating encephalitogenic T-lymphocytes. Expectedly, ATI activated peripheral blood monocytes from both patients with MS and healthy controls. CONCLUSIONS Dietary wheat ATI activate murine and human myeloid cells. The amount of ATI present in an average human wheat-based diet caused mild intestinal inflammation, which was propagated to extraintestinal sites, leading to exacerbation of CNS inflammation and worsening of clinical symptoms in EAE. These results support the importance of the gut-brain axis in inflammatory CNS disease.
Collapse
Affiliation(s)
- Victor F Zevallos
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Department of Applied and Health Sciences, Northumbria University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Nir Yogev
- Institute for Molecular Medicine, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Department of Dermatology, University of Cologne, Koln, Germany
| | - Judith Hauptmann
- Institute for Molecular Medicine, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Alexei Nikolaev
- Institute for Molecular Medicine, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Geethanjali Pickert
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Valeska Heib
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Nicola Fittler
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Felix Luessi
- Department of Neurology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Manjusha Neerukonda
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | | | - Ann-Marie Tobinski
- Institute for Molecular Medicine, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Luisa Klotz
- Neurology Department, University Hospital Munster, Munster, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg Universitat Mainz, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Caminero A. Wheat proteins as triggers of central nervous system inflammation. Gut 2023; 73:5-6. [PMID: 37751934 DOI: 10.1136/gutjnl-2023-330848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023]
Affiliation(s)
- Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
21
|
Kalinovskii AP, Sintsova OV, Gladkikh IN, Leychenko EV. Natural Inhibitors of Mammalian α-Amylases as Promising Drugs for the Treatment of Metabolic Diseases. Int J Mol Sci 2023; 24:16514. [PMID: 38003703 PMCID: PMC10671682 DOI: 10.3390/ijms242216514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
α-Amylase is a generally acknowledged molecular target of a distinct class of antidiabetic drugs named α-glucosidase inhibitors. This class of medications is scarce and rather underutilized, and treatment with current commercial drugs is accompanied by unpleasant adverse effects. However, mammalian α-amylase inhibitors are abundant in nature and form an extensive pool of high-affinity ligands that are available for drug discovery. Individual compounds and natural extracts and preparations are promising therapeutic agents for conditions associated with impaired starch metabolism, e.g., diabetes mellitus, obesity, and other metabolic disorders. This review focuses on the structural diversity and action mechanisms of active natural products with inhibitory activity toward mammalian α-amylases, and emphasizes proteinaceous inhibitors as more effective compounds with significant potential for clinical use.
Collapse
Affiliation(s)
- Aleksandr P. Kalinovskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Oksana V. Sintsova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia; (O.V.S.); (I.N.G.)
| | - Irina N. Gladkikh
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia; (O.V.S.); (I.N.G.)
| | - Elena V. Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia; (O.V.S.); (I.N.G.)
| |
Collapse
|
22
|
Brouns F, Van Haaps A, Keszthelyi D, Venema K, Bongers M, Maas J, Mijatovic V. Diet associations in endometriosis: a critical narrative assessment with special reference to gluten. Front Nutr 2023; 10:1166929. [PMID: 37731404 PMCID: PMC10507348 DOI: 10.3389/fnut.2023.1166929] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Endometriosis is characterized by the presence of endometrium-like tissue outside the uterus. The etiology remains largely unknown. Despite adequate treatment, patients can still experience symptoms or side effects resulting in therapy incompliance and in self-management strategies such as dietary measures is increasing. A gluten free diet is thought to be contributory in reducing endometriosis-related pain, thereby optimizing quality of life. However, data is conflicting and currently provides no evidence for causality. This narrative review aims to put the effect of dietary self-management strategies on endometriosis in a balanced perspective, especially the effect of gluten and a gluten free diet. Several studies have found a strong overlap in symptoms, metabolic and immune responses associated with endometriosis and those associated with celiac disease, ulcerative colitis, Crohn's disease, irritable bowel syndrome and non-celiac wheat sensitivity. However, it remains unclear whether these diseases and/or disorders are causal to an increased risk of endometriosis. Some studies have found a positive effect on the risk of endometriosis, endometriosis-related symptoms and quality of life (QoL) when women either avoided certain nutrients or foods, or applied a specific nutrient supplementation. This includes the avoidance of red meat, an increasing intake of foods rich in anti-oxidants, omega-3, micronutrients and dietary fibers (e.g., fruit, vegetables) and the appliance of a gluten free diet. However, data from the available studies were generally graded of low quality and it was noted that placebo and/or nocebo effects influenced the reported positive effects. In addition, such effects were no longer seen when adjusting for confounders such as overweight, when a translation was made from in vitro to in vivo, or when the nutrients were not supplemented as isolated sources but as part of a mixed daily diet. Finally, some studies showed that long-term adherence to a gluten free diet is often associated with an impaired diet quality and nutrient intake, leading to negative health outcomes and reduced QoL. Concluding, scientific evidence on the efficacy of dietary interventions on well-defined clinical endpoints of endometriosis is lacking and recommending a gluten free diet to women solely diagnosed with endometriosis should therefore not be advised.
Collapse
Affiliation(s)
- Fred Brouns
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Annelotte Van Haaps
- Endometriosis Center, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Daniel Keszthelyi
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University, Maastricht, Netherlands
| | - Marlies Bongers
- Department of Obstetrics and Gynecology, Máxima Medical Center, Veldhoven, Netherlands
- Grow-School of Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Jacques Maas
- Grow-School of Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology MUMC+, Maastricht, Netherlands
| | - Velja Mijatovic
- Endometriosis Center, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| |
Collapse
|
23
|
Geisslitz S, Pronin D, Neerukonda M, Curella V, Neufang S, Koch S, Weichert H, Weber H, Börner A, Schuppan D, Scherf KA. Breeding from 1891 to 2010 did not increase the content of amylase/trypsin-inhibitors in wheat (Triticum aestivum). NPJ Sci Food 2023; 7:43. [PMID: 37612428 PMCID: PMC10447418 DOI: 10.1038/s41538-023-00219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
The prevalence of hypersensitivities towards wheat has increased in the last decades. Apart from celiac disease these include allergic and other inflammatory reactions summarized under the term non-celiac wheat sensitivity. One suspected trigger is the family of amylase/trypsin-inhibitors (ATIs), non-gluten proteins that are prominent wheat allergens and that activate the toll-like receptor 4 on intestinal immune cells to promote intestinal and extra-intestinal inflammation. We therefore quantified 13 ATIs in 60 German hexaploid winter wheat cultivars originating from 1891 to 2010 and harvested in three years by targeted liquid chromatography-tandem mass spectrometry combined with stable isotope dilution assay using specific marker peptides as internal standards. The total ATI content and that of the two major ATIs 0.19 and CM3 did not change from old cultivars (first registered from 1891 to 1950) to modern cultivars (1951-2010). There were also no significant changes in ATI distribution.
Collapse
Affiliation(s)
- Sabrina Geisslitz
- Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20 a, 76131, Karlsruhe, Germany.
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354, Freising, Germany.
| | - Darina Pronin
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354, Freising, Germany
| | - Manjusha Neerukonda
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Valentina Curella
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sibylle Neufang
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sandra Koch
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Heiko Weichert
- Department of Molecular Genetics, Leibniz Institute of Plant Genetics and Crop Plant Research, Corrensstr. 3, 06466 Seeland/OT Gatersleben, Germany
| | - Hans Weber
- Department of Molecular Genetics, Leibniz Institute of Plant Genetics and Crop Plant Research, Corrensstr. 3, 06466 Seeland/OT Gatersleben, Germany
| | - Andreas Börner
- Genebank Department, Leibniz Institute of Plant Genetics and Crop Plant Research, Corrensstr. 3, 06466 Seeland/OT Gatersleben, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Katharina Anne Scherf
- Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20 a, 76131, Karlsruhe, Germany.
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354, Freising, Germany.
| |
Collapse
|
24
|
Rizwan MZ, Kerbus R, Kamstra K, Keerthisinghe P, Tups A. Dietary wheat gluten induces astro- and microgliosis in the hypothalamus of male mice. J Neuroendocrinol 2023; 35:e13326. [PMID: 37534400 DOI: 10.1111/jne.13326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 08/04/2023]
Abstract
Gluten, which is found in cereals such as wheat, rye and barley, makes up a major dietary component in most western nations, and has been shown to promote body mass gain and peripheral inflammation in mice. In the current study, we investigated the impact of gluten on central inflammation that is typically associated with diet-induced obesity. While we found no effect of gluten when added to a low-fat diet (LFD), male mice fed high fat diet (HFD) enriched with gluten increased body mass and adiposity compared with mice fed HFD without gluten. We furthermore found that gluten, when added to the LFD, increases circulating C-reactive protein levels. Gluten regardless of whether it was added to LFD or HFD led to a profound increase in the number of microglia and astrocytes in the arcuate nucleus of the hypothalamus, as detected by immunohistochemistry for ionised calcium binding adaptor molecule 1 (Iba-1) and glial fibrillary acidic protein (GFAP), respectively. In mice fed LFD, gluten mimicked the immunogenic effects of HFD exposure and when added to HFD led to a further increase in the number of immunoreactive cells. Taken together, our results confirm a moderate obesogenic effect of gluten when fed to mice exposed to HFD and for the first-time report gluten-induced astro- and microgliosis suggesting the development of hypothalamic injury in rodents.
Collapse
Affiliation(s)
- Mohammed Z Rizwan
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Romy Kerbus
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Kaj Kamstra
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Pramuk Keerthisinghe
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Alexander Tups
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
25
|
D’Amico V, Gänzle M, Call L, Zwirzitz B, Grausgruber H, D’Amico S, Brouns F. Does sourdough bread provide clinically relevant health benefits? Front Nutr 2023; 10:1230043. [PMID: 37545587 PMCID: PMC10399781 DOI: 10.3389/fnut.2023.1230043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/27/2023] [Indexed: 08/08/2023] Open
Abstract
During the last decade, scientific interest in and consumer attention to sourdough fermentation in bread making has increased. On the one hand, this technology may favorably impact product quality, including flavor and shelf-life of bakery products; on the other hand, some cereal components, especially in wheat and rye, which are known to cause adverse reactions in a small subset of the population, can be partially modified or degraded. The latter potentially reduces their harmful effects, but depends strongly on the composition of sourdough microbiota, processing conditions and the resulting acidification. Tolerability, nutritional composition, potential health effects and consumer acceptance of sourdough bread are often suggested to be superior compared to yeast-leavened bread. However, the advantages of sourdough fermentation claimed in many publications rely mostly on data from chemical and in vitro analyzes, which raises questions about the actual impact on human nutrition. This review focuses on grain components, which may cause adverse effects in humans and the effect of sourdough microbiota on their structure, quantity and biological properties. Furthermore, presumed benefits of secondary metabolites and reduction of contaminants are discussed. The benefits claimed deriving from in vitro and in vivo experiments will be evaluated across a broader spectrum in terms of clinically relevant effects on human health. Accordingly, this critical review aims to contribute to a better understanding of the extent to which sourdough bread may result in measurable health benefits in humans.
Collapse
Affiliation(s)
- Vera D’Amico
- Department of Food Science and Technology, BOKU–University of Natural Resources and Life Sciences, Vienna, Austria
| | - Michael Gänzle
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Lisa Call
- Department of Crop Sciences, BOKU–University of Natural Resources and Life Sciences, Tulln, Austria
| | - Benjamin Zwirzitz
- Department of Food Science and Technology, BOKU–University of Natural Resources and Life Sciences, Vienna, Austria
| | - Heinrich Grausgruber
- Department of Crop Sciences, BOKU–University of Natural Resources and Life Sciences, Tulln, Austria
| | - Stefano D’Amico
- Institute for Animal Nutrition and Feed, AGES–Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Fred Brouns
- Department of Human Biology, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
26
|
Bascuñán KA, Orosteguí C, Rodríguez JM, Roncoroni L, Doneda L, Elli L, Araya M. Heavy Metal and Rice in Gluten-Free Diets: Are They a Risk? Nutrients 2023; 15:2975. [PMID: 37447301 DOI: 10.3390/nu15132975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
A gluten-free diet (GFD) is the treatment of choice for gluten-related disorders. It has been associated with macro- and micronutrient deficiencies. Recently, consumption of arsenic-contaminated rice has raised concern because of the potential greater risk that it may represent for people on GFDs, whose rice consumption is high, since it is a fundamental cereal in GFDs. We reviewed the data published over the last 20 years in Medline and Scielo, in English, French and Spanish, on four metals (As, Hg, Cd, and Pb), to assess whether the evidence suggests that celiac disease or consumption of a GFD is associated with increased levels of blood/urinary metal concentrations. The review revealed a few articles that were directly related to the four metals and their relationships with a GFD. The evidence supports that rice-based products are a relevant source of As and other metals. Clinical studies and evaluations based on NHANES have indicated that persons on GFDs have higher As and Hg blood/urinary levels, suggesting that the diet and not the disease is responsible for it. The levels described are statistically significant compared to those of persons on complete diets, but far from toxic levels. The question of whether higher exposure to heavy metals associated with a GFD is biologically relevant remains unanswered and deserves study.
Collapse
Affiliation(s)
- Karla A Bascuñán
- Department of Nutrition, School of Medicine, University of Chile, Santiago 8380453, Chile
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| | - Claudia Orosteguí
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| | - Juan Manuel Rodríguez
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| | - Leda Roncoroni
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy
| | - Luisa Doneda
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Magdalena Araya
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| |
Collapse
|
27
|
Jahn N, Longin CFH, Scherf KA, Geisslitz S. No correlation between amylase/trypsin-inhibitor content and amylase inhibitory activity in hexaploid and tetraploid wheat species. Curr Res Food Sci 2023; 7:100542. [PMID: 38115899 PMCID: PMC10728332 DOI: 10.1016/j.crfs.2023.100542] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 12/21/2023] Open
Abstract
Wheat amylase/trypsin-inhibitors (ATI) are known triggers for wheat-related disorders. The aims of our study were to determine (1) the inhibitory activity against different α-amylases, (2) the content of albumins and globulins (ALGL) and total ATI and (3) to correlate these parameters in wholegrain flour of hexaploid, tetraploid and diploid wheat species. The amount of ATI within the ALGL fraction varied from 0.8% in einkorn to 20% in spelt. ATI contents measured with reversed-phase high-performance liquid chromatography (RP-HPLC) revealed similar contents (1.2-4.2 mg/g) compared to the results determined by LC-MS/MS (0.2-5.2 mg/g) for all wheat species except einkorn. No correlation was found between ALGL content and inhibitory activity. In general, hexaploid cultivars of spelt and common wheat had the highest inhibitory activities, showing values between 897 and 3564 AIU/g against human salivary α-amylase. Tetraploid wheat species durum and emmer had lower activities (170-1461 AIU/g), although a few emmer cultivars showed similar activities at one location. In einkorn, no inhibitory activity was found. No correlation was observed between the ATI content and the inhibitory activity against the used α-amylases, highlighting that it is very important to look at the parameters separately.
Collapse
Affiliation(s)
- Nora Jahn
- Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20 a, 76131, Karlsruhe, Germany
| | | | - Katharina A. Scherf
- Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20 a, 76131, Karlsruhe, Germany
| | - Sabrina Geisslitz
- Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20 a, 76131, Karlsruhe, Germany
| |
Collapse
|
28
|
Engel S, Klotz L, Wirth T, Fleck AK, Pickert G, Eschborn M, Kreuzburg S, Curella V, Bittner S, Zipp F, Schuppan D, Luessi F. Attenuation of immune activation in patients with multiple sclerosis on a wheat-reduced diet: a pilot crossover trial. Ther Adv Neurol Disord 2023; 16:17562864231170928. [PMID: 37384112 PMCID: PMC10293514 DOI: 10.1177/17562864231170928] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/03/2023] [Indexed: 06/30/2023] Open
Abstract
Background Western lifestyle has been associated with an increase in relapsing-remitting multiple sclerosis (RRMS). In mice, dietary wheat amylase-trypsin inhibitors (ATIs) activate intestinal myeloid cells and augment T cell-mediated systemic inflammation. Objective The aim of this study was to assess whether a wheat- and thus ATI-reduced diet might exert beneficial effects in RRMS patients with modest disease activity. Methods In this 6-month, crossover, open-label, bicentric proof-of-concept trial, 16 RRMS patients with stable disease course were randomized to either 3 months of a standard wheat-containing diet with consecutive switch to a > 90% wheat-reduced diet, or vice versa. Results The primary endpoint was negative, as the frequency of circulating pro-inflammatory T cells did not decrease during the ATI-reduced diet. We did, however, observe decreased frequencies of CD14+ CD16++ monocytes and a concomitant increase in CD14++ CD16- monocytes during the wheat-reduced diet interval. This was accompanied by an improvement in pain-related quality of life in health-related quality of life assessed (SF-36). Conclusion Our results suggest that the wheat- and thus ATI-reduced diet was associated with changes in monocyte subsets and improved pain-related quality of life in RRMS patients. Thus, a wheat (ATI)-reduced diet might be a complementary approach accompanying immunotherapy for some patients. Registration German Clinical Trial Register (No. DRKS00027967).
Collapse
Affiliation(s)
- Sinah Engel
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Timo Wirth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Ann-Katrin Fleck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Geethanjali Pickert
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie Eschborn
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Samia Kreuzburg
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Valentina Curella
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Felix Luessi
- Department of Neurology and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| |
Collapse
|
29
|
Wenger M, Grosse-Kathoefer S, Kraiem A, Pelamatti E, Nunes N, Pointner L, Aglas L. When the allergy alarm bells toll: The role of Toll-like receptors in allergic diseases and treatment. Front Mol Biosci 2023; 10:1204025. [PMID: 37426425 PMCID: PMC10325731 DOI: 10.3389/fmolb.2023.1204025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Toll-like receptors of the human immune system are specialized pathogen detectors able to link innate and adaptive immune responses. TLR ligands include among others bacteria-, mycoplasma- or virus-derived compounds such as lipids, lipo- and glycoproteins and nucleic acids. Not only are genetic variations in TLR-related genes associated with the pathogenesis of allergic diseases, including asthma and allergic rhinitis, their expression also differs between allergic and non-allergic individuals. Due to a complex interplay of genes, environmental factors, and allergen sources the interpretation of TLRs involved in immunoglobulin E-mediated diseases remains challenging. Therefore, it is imperative to dissect the role of TLRs in allergies. In this review, we discuss i) the expression of TLRs in organs and cell types involved in the allergic immune response, ii) their involvement in modulating allergy-associated or -protective immune responses, and iii) how differential activation of TLRs by environmental factors, such as microbial, viral or air pollutant exposure, results in allergy development. However, we focus on iv) allergen sources interacting with TLRs, and v) how targeting TLRs could be employed in novel therapeutic strategies. Understanding the contributions of TLRs to allergy development allow the identification of knowledge gaps, provide guidance for ongoing research efforts, and built the foundation for future exploitation of TLRs in vaccine design.
Collapse
|
30
|
Wang Z, Shea Z, Rosso L, Shang C, Li J, Bewick P, Li Q, Zhao B, Zhang B. Development of new mutant alleles and markers for KTI1 and KTI3 via CRISPR/Cas9-mediated mutagenesis to reduce trypsin inhibitor content and activity in soybean seeds. FRONTIERS IN PLANT SCIENCE 2023; 14:1111680. [PMID: 37223818 PMCID: PMC10200896 DOI: 10.3389/fpls.2023.1111680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/31/2023] [Indexed: 05/25/2023]
Abstract
The digestibility of soybean meal can be severely impacted by trypsin inhibitor (TI), one of the most abundant anti-nutritional factors present in soybean seeds. TI can restrain the function of trypsin, a critical enzyme that breaks down proteins in the digestive tract. Soybean accessions with low TI content have been identified. However, it is challenging to breed the low TI trait into elite cultivars due to a lack of molecular markers associated with low TI traits. We identified Kunitz trypsin inhibitor 1 (KTI1, Gm01g095000) and KTI3 (Gm08g341500) as two seed-specific TI genes. Mutant kti1 and kti3 alleles carrying small deletions or insertions within the gene open reading frames were created in the soybean cultivar Glycine max cv. Williams 82 (WM82) using the CRISPR/Cas9-mediated genome editing approach. The KTI content and TI activity both remarkably reduced in kti1/3 mutants compared to the WM82 seeds. There was no significant difference in terms of plant growth or maturity days of kti1/3 transgenic and WM82 plants in greenhouse condition. We further identified a T1 line, #5-26, that carried double homozygous kti1/3 mutant alleles, but not the Cas9 transgene. Based on the sequences of kti1/3 mutant alleles in #5-26, we developed markers to co-select for these mutant alleles by using a gel-electrophoresis-free method. The kti1/3 mutant soybean line and associated selection markers will assist in accelerating the introduction of low TI trait into elite soybean cultivars in the future.
Collapse
Affiliation(s)
- Zhibo Wang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Zachary Shea
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Luciana Rosso
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Chao Shang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Jianyong Li
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, United States
| | - Patrick Bewick
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Qi Li
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Bingyu Zhao
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Bo Zhang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
31
|
Rej A, Penny HA. Current evidence for dietary therapies in irritable bowel syndrome. Curr Opin Gastroenterol 2023; 39:219-226. [PMID: 36976876 DOI: 10.1097/mog.0000000000000930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
PURPOSE OF REVIEW Diet appears to trigger symptoms in the majority of individuals with irritable bowel syndrome (IBS) and is associated with a reduced quality of life. There has been a recent focus on the role of dietary therapies to manage individuals with IBS. The aim of this review is to discuss the utility of traditional dietary advice (TDA), low-FODMAP diet (LFD) and gluten-free diet (GFD) in IBS. RECENT FINDINGS Several recent randomized controlled trials (RCTs) have been published demonstrating the efficacy of the LFD and GFD in IBS, with the evidence base for TDA being predominantly based on clinical experience, with emerging RCTs evaluating TDA. Only one RCT has been published to date comparing TDA, LFD and GFD head to head, with no difference noted between diets in terms of efficacy. However, TDA has been noted to be more patient-friendly and is commonly implemented as a first-line dietary therapy. SUMMARY Dietary therapies have been demonstrated to improve symptoms in patients with IBS. In view of insufficient evidence to recommend one diet over another currently, specialist dietetic input in conjunction with patient preference is required to determine implementation of dietary therapies. Novel methods of dietetic delivery are required in view of the lack of dietetic provision to deliver these therapies.
Collapse
Affiliation(s)
- Anupam Rej
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals, Sheffield
| | - Hugo A Penny
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals, Sheffield
- Academic Unit of Gastroenterology, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| |
Collapse
|
32
|
Kaushik M, Mulani E, Mahendru-Singh A, Makharia G, Mohan S, Mandal PK. Comparative Expression Profile of Genes Encoding Intolerant Proteins in Bread vs. Durum Wheat During Grain Development. JOURNAL OF PLANT GROWTH REGULATION 2023; 42:3200-3210. [DOI: 10.1007/s00344-022-10785-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 08/23/2022] [Indexed: 08/30/2023]
|
33
|
Cenni S, Sesenna V, Boiardi G, Casertano M, Russo G, Reginelli A, Esposito S, Strisciuglio C. The Role of Gluten in Gastrointestinal Disorders: A Review. Nutrients 2023; 15:nu15071615. [PMID: 37049456 PMCID: PMC10096482 DOI: 10.3390/nu15071615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Gluten is only partially digested by intestinal enzymes and can generate peptides that can alter intestinal permeability, facilitating bacterial translocation, thus affecting the immune system. Few studies addressed the role of diet with gluten in the development of intestinal inflammation and in other gastrointestinal disorders. The aim of this narrative review was to analyse the role of gluten in several gastrointestinal diseases so as to give a useful overview of its effectiveness in the prevention and management of these disorders.
Collapse
Affiliation(s)
- Sabrina Cenni
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Veronica Sesenna
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
| | - Giulia Boiardi
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
| | - Marianna Casertano
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giuseppina Russo
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alfonso Reginelli
- Radiology Unit, Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Napoli, Italy
| | - Susanna Esposito
- Department of Medicine and Surgery, Pediatric Clinic, University of Parma, 43126 Parma, Italy
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: ; Tel.: +39-081-5665464; Fax: +39-081-7462679
| |
Collapse
|
34
|
Afzal M, Sielaff M, Distler U, Schuppan D, Tenzer S, Longin CFH. Reference proteomes of five wheat species as starting point for future design of cultivars with lower allergenic potential. NPJ Sci Food 2023; 7:9. [PMID: 36966156 PMCID: PMC10039927 DOI: 10.1038/s41538-023-00188-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/06/2023] [Indexed: 03/27/2023] Open
Abstract
Wheat is an important staple food and its processing quality is largely driven by proteins. However, there is a sizable number of people with inflammatory reactions to wheat proteins, namely celiac disease, wheat allergy and the syndrome of non-celiac wheat sensitivity. Thus, proteome profiles should be of high importance for stakeholders along the wheat supply chain. We applied liquid chromatography-tandem mass spectrometry-based proteomics to establish the flour reference proteome for five wheat species, ancient to modern, each based on 10 cultivars grown in three diverse environments. We identified at least 2540 proteins in each species and a cluster analyses clearly separated the species based on their proteome profiles. Even more, >50% of proteins significantly differed between species - many of them implicated in products' quality, grain-starch synthesis, plant stress regulation and proven or potential allergic reactions in humans. Notably, the expression of several important wheat proteins was found to be mainly driven by genetics vs. environmental factors, which enables selection and refinement of improved cultivars for the wheat supply chain as long as rapid test methods will be developed. Especially einkorn expressed 5.4 and 7.2-fold lower quantities of potential allergens and immunogenic amylase trypsin inhibitors, respectively, than common wheat, whereas potential allergen content was intermediate in tetraploid wheat species. This urgently warrants well-targeted clinical studies, where the developed reference proteomes will help to design representative test diets.
Collapse
Affiliation(s)
- Muhammad Afzal
- State Plant Breeding Institute, University of Hohenheim, Fruwirthstr. 21, 70599, Stuttgart, Germany
| | - Malte Sielaff
- Institute for Immunology and Research Center for Immune Therapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Ute Distler
- Institute for Immunology and Research Center for Immune Therapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Stefan Tenzer
- Institute for Immunology and Research Center for Immune Therapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - C Friedrich H Longin
- State Plant Breeding Institute, University of Hohenheim, Fruwirthstr. 21, 70599, Stuttgart, Germany.
| |
Collapse
|
35
|
Di Nardo G, Cremon C, Staiano A, Stanghellini V, Borrelli O, Strisciuglio C, Romano C, Mallardo S, Scarpato E, Marasco G, Salvatore S, Zenzeri L, Felici E, Pensabene L, Sestito S, Francavilla R, Quitadamo P, Baldassarre M, Giorgio V, Tambucci R, Ziparo C, Parisi P, Barbaro MR, Barbara G. Role of inflammation in pediatric irritable bowel syndrome. Neurogastroenterol Motil 2023; 35:e14365. [PMID: 35340083 DOI: 10.1111/nmo.14365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 02/09/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND IBS affects a large number of children throughout the world and is thought to be the result of disturbed neuroimmune function along with the brain-gut axis. Although the underlying pathophysiologic mechanisms are not clear, the role of low-grade inflammation and mucosal immune activation in IBS symptom generation has become evident also in subsets of pediatric patients. Animal models provided meaningful insight in the causal relationship between abnormal mucosal immune activation and changes in gastrointestinal (GI) sensory-motor function. Likewise, the development of long-standing GI symptoms fulfilling the current criteria for functional GI disorders after infection gastroenteritis and in patients with IBD or celiac disease in remission further supports this hypothesis. Immune activation, its impact on gut sensory-motor function, and potential implications for symptom generation emerged in both children and adults with IBS. PURPOSE The aim of this review is to summarize the main evidence on the presence of low-grade inflammation and immune activation in children with IBS, its possible role in symptom generation, and its potential implication for new therapeutic strategies.
Collapse
Affiliation(s)
- Giovanni Di Nardo
- NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Cesare Cremon
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, "Federico II", University of Naples, Naples, Italy
| | | | - Osvaldo Borrelli
- Division of Neurogastroenterology and Motility, Department of Paediatric Gastroenterology, UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialistic Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudio Romano
- Pediatric Gastroenterology Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi", University of Messina, Messina, Italy
| | - Saverio Mallardo
- Pediatric Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Elena Scarpato
- Department of Translational Medical Science, "Federico II", University of Naples, Naples, Italy
| | - Giovanni Marasco
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Silvia Salvatore
- Pediatric Department, Ospedale "F. Del Ponte", University of Insubria, Varese, Italy
| | - Letizia Zenzeri
- NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea University Hospital, Sapienza University of Rome, Rome, Italy.,Pediatric Emergency Unit, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Enrico Felici
- Pediatric and Pediatric Emergency Unit, "Umberto Bosio" Center for Digestive Diseases, The Children Hospital, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Licia Pensabene
- Department of Medical and Surgical Sciences, Pediatric Unit, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Simona Sestito
- Department of Medical and Surgical Sciences, Pediatric Unit, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Ruggiero Francavilla
- Pediatric Section, Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Paolo Quitadamo
- Department of Pediatrics, A.O.R.N. Santobono-Pausilipon, Naples, Italy
| | - Mariella Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | | | - Renato Tambucci
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Chiara Ziparo
- NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Pasquale Parisi
- NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | | | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | |
Collapse
|
36
|
Funsten MC, Yurkovetskiy LA, Kuznetsov A, Reiman D, Hansen CHF, Senter KI, Lee J, Ratiu J, Dahal-Koirala S, Antonopoulos DA, Dunny GM, Sollid LM, Serreze D, Khan AA, Chervonsky AV. Microbiota-dependent proteolysis of gluten subverts diet-mediated protection against type 1 diabetes. Cell Host Microbe 2023; 31:213-227.e9. [PMID: 36603588 PMCID: PMC9911364 DOI: 10.1016/j.chom.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/02/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023]
Abstract
Diet and commensals can affect the development of autoimmune diseases like type 1 diabetes (T1D). However, whether dietary interventions are microbe-mediated was unclear. We found that a diet based on hydrolyzed casein (HC) as a protein source protects non-obese diabetic (NOD) mice in conventional and germ-free (GF) conditions via improvement in the physiology of insulin-producing cells to reduce autoimmune activation. The addition of gluten (a cereal protein complex associated with celiac disease) facilitates autoimmunity dependent on microbial proteolysis of gluten: T1D develops in GF animals monocolonized with Enterococcus faecalis harboring secreted gluten-digesting proteases but not in mice colonized with protease deficient bacteria. Gluten digestion by E. faecalis generates T cell-activating peptides and promotes innate immunity by enhancing macrophage reactivity to lipopolysaccharide (LPS). Gnotobiotic NOD Toll4-negative mice monocolonized with E. faecalis on an HC + gluten diet are resistant to T1D. These findings provide insights into strategies to develop dietary interventions to help protect humans against autoimmunity.
Collapse
Affiliation(s)
- Matthew C Funsten
- Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA; Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Leonid A Yurkovetskiy
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA; Committee on Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | - Andrey Kuznetsov
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Derek Reiman
- Toyota Technological Institute at Chicago, Chicago, IL 60637, USA
| | - Camilla H F Hansen
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA; Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Katharine I Senter
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Jean Lee
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA; Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Jeremy Ratiu
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Shiva Dahal-Koirala
- KG Jebsen Coeliac Disease Research Centre and Department of Immunology, University of Oslo and University of Oslo Hospital, 0372 Oslo, Norway
| | | | - Gary M Dunny
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ludvig M Sollid
- KG Jebsen Coeliac Disease Research Centre and Department of Immunology, University of Oslo and University of Oslo Hospital, 0372 Oslo, Norway
| | | | - Aly A Khan
- Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA; Department of Pathology, The University of Chicago, Chicago, IL 60637, USA; Institute for Population and Precision Health, The University of Chicago, Chicago, IL 60637, USA; Department of Family Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Alexander V Chervonsky
- Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA; Department of Pathology, The University of Chicago, Chicago, IL 60637, USA; Committee on Microbiology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
37
|
Investigation of TLR4 Antagonists for Prevention of Intestinal Inflammation. Inflammation 2023; 46:103-114. [PMID: 35867263 PMCID: PMC9971050 DOI: 10.1007/s10753-022-01714-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/08/2022] [Accepted: 07/04/2022] [Indexed: 11/05/2022]
Abstract
Activation of toll-like receptor 4 (TLR4) has been shown to be a major influence on the inflammatory signalling pathways in intestinal mucositis (IM), as demonstrated by TLR4 knock-out mice. Pharmacological TLR4 inhibition has thus been postulated as a potential new therapeutic approach for the treatment of IM but specific TLR4 inhibitors have yet to be investigated. As such, we aimed to determine whether direct TLR4 antagonism prevents inflammation in pre-clinical experimental models of IM. The non-competitive and competitive TLR4 inhibitors, TAK-242 (10 µM) and IAXO-102 (10 µM), respectively, or vehicle were added to human T84, HT-29, and U937 cell lines and mouse colonic explants 1 h before the addition of lipopolysaccharide (LPS) (in vitro: 100 µg/mL; ex vivo: 10 µg/mL), SN-38 (in vitro: 1 µM or 1 nM; ex vivo: 2 µM), and/or tumour necrosis factor-alpha (TNF-α) (5 µg/mL). Supernatant was collected for human IL-8 and mouse IL-6 enzyme-linked immunosorbent assays (ELISAs), as a measure of inflammatory signalling. Cell viability was measured using XTT assays. Explant tissue was used in histopathological and RT-PCR analysis for genes of interest: TLR4, MD2, CD14, MyD88, IL-6, IL-6R, CXCL2, CXCR1, CXCR2. SN-38 increased cytostasis compared to vehicle (P < 0.0001). However, this was not prevented by either antagonist (P > 0.05) in any of the 3 cell lines. Quantitative histological assessment scores showed no differences between vehicle and treatment groups (P > 0.05). There were no differences in in vitro IL-8 (P > 0.05, in all 3 cells lines) and ex vivo IL-6 (P > 0.05) concentrations between vehicle and treatment groups. Transcript expression of all genes was similar across vehicle and treatment groups (P > 0.05). TLR4 antagonism using specific inhibitors TAK-242 and IAXO-102 was not effective at blocking IM in these pre-clinical models of mucositis. This work indicates that specific epithelial inhibition of TLR4 with these compounds is insufficient to manage mucositis-related inflammation. Rather, TLR4 signalling through immune cells may be a more important target to prevent IM.
Collapse
|
38
|
Abstract
Proteases are an evolutionarily conserved family of enzymes that degrade peptide bonds and have been implicated in several common gastrointestinal (GI) diseases. Although luminal proteolytic activity is important for maintenance of homeostasis and health, the current review describes recent advances in our understanding of how overactivity of luminal proteases contributes to the pathophysiology of celiac disease, irritable bowel syndrome, inflammatory bowel disease and GI infections. Luminal proteases, many of which are produced by the microbiota, can modulate the immunogenicity of dietary antigens, reduce mucosal barrier function and activate pro-inflammatory and pro-nociceptive host signaling. Increased proteolytic activity has been ascribed to both increases in protease production and decreases in inhibitors of luminal proteases. With the identification of strains of bacteria that are important sources of proteases and their inhibitors, the stage is set to develop drug or microbial therapies to restore protease balance and alleviate disease.
Collapse
Affiliation(s)
- Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mabel Guzman
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada
| | - Josie Libertucci
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Alan E. Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada,CONTACT Alan E. Lomax Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, K7L 2V7, Canada
| |
Collapse
|
39
|
Hov JR. Editorial: dietary interventions to understand and treat primary sclerosing cholangitis-is gluten-free diet a starting point? Aliment Pharmacol Ther 2023; 57:265-266. [PMID: 36565005 DOI: 10.1111/apt.17326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Johannes R Hov
- Norwegian PSC Research Center, Section of Gastroenterology and Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Shewry P. Wheat grain proteins: Past, present, and future. Cereal Chem 2023; 100:9-22. [PMID: 37064052 PMCID: PMC10087814 DOI: 10.1002/cche.10585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/06/2022]
Abstract
Background and Objectives Research on wheat grain proteins is reviewed, including achievements over the past century and priorities for future research. The focus is on three groups of proteins that have major impacts on wheat quality and utilization: the gluten proteins which determine dough viscoelasticity but also trigger celiac disease in susceptible individuals, the puroindolines which are major determinants of grain texture and the amylase/trypsin inhibitors which are food and respiratory allergens and are implicated in triggering celiac disease and nonceliac wheat sensitivity. Findings Although earlier work focused on protein structure and properties, the development of genomics and high-sensitivity proteomics has resulted in the availability of a vast amount of information on the amino acid sequences of individual wheat proteins, including allelic variants of gluten proteins which are associated with good processing quality and of puroindolines, which are associated with a hard or soft grain texture, and on protein expression and polymorphism. Conclusions However, our ability to exploit this knowledge is limited by a lack of detailed understanding of the structure:function relationships of wheat proteins. In particular, we need to understand how the three-dimensional structures of the individual proteins determine their interactions with other grain components (to determine functional properties) and with the immune systems of susceptible consumers (to trigger adverse responses), how these interactions are affected by allelic variation, and how they can be manipulated. Significance and Novelty The article, therefore, identifies priorities for future research which should enable the adoption of a more rational approach to improving the quality of wheat grain proteins.
Collapse
|
41
|
Liwinski T, Hübener S, Henze L, Hübener P, Heinemann M, Tetzlaff M, Hiller MI, Jagemann B, Surabattula R, Leeming D, Karsdal M, Monguzzi E, Schachschal G, Rösch T, Bang C, Franke A, Lohse AW, Schuppan D, Schramm C. A prospective pilot study of a gluten-free diet for primary sclerosing cholangitis and associated colitis. Aliment Pharmacol Ther 2023; 57:224-236. [PMID: 36266939 DOI: 10.1111/apt.17256] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/02/2022] [Accepted: 10/08/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Primary sclerosing cholangitis (PSC) is a progressive bile duct disease associated with inflammatory bowel disease (PSC-IBD). AIM To investigate whether patients with PSC-IBD benefit from a gluten-free and amylase trypsin inhibitor (ATI)-free diet (GFD). METHODS We performed a prospective clinical pilot study administering an eight-week GFD. The primary outcomes were colonic inflammation assessed by proctosigmoidoscopy, and liver stiffness (surrogate for fibrosis, inflammation and cholestasis) measured by transient elastography before and after GFD. Amongst the secondary (exploratory) outcomes were colonic mucosal and serum cytokine/chemokine changes, the intestinal microbiome and transcriptome dynamics, and shifts in serum markers of hepatic fibrogenesis. RESULTS Fifteen patients with PSC-IBD completed the study. The study did not meet its primary outcome: the endoscopic score and liver stiffness remained unchanged. However, the expression of pro-inflammatory mucosal cytokines and chemokines such as IL6, IL8, CCL2, and TNFα was significantly down-regulated. Two critical markers of liver fibrosis and matrix remodelling, thrombospondin-2 and -4, decreased significantly. The microbiota composition changed slightly, including a decrease in the pathogen Romboutsia ilealis. The intestinal transcriptome indicated a gut barrier improvement. Pruritus, fatigue, overall well-being, faecal calprotectin levels, and serum alkaline phosphatase did not change significantly. CONCLUSIONS This study did not demonstrate a clinical improvement with short-term GFD in patients with PSC-IBD. However, a gluten/ATI-free diet may improve biomarkers of intestinal inflammation and barrier function in these patients with associated changes in the enteric microbiota. Further investigation of the therapeutic potential of the GFD in PSC-IBD is warranted.
Collapse
Affiliation(s)
- Timur Liwinski
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Center for Affective, Stress and Sleep Disorders (ZASS), University Psychiatric Clinics (UPK) Basel, University of Basel, Basel, Switzerland
| | - Sina Hübener
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lara Henze
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Hübener
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melina Heinemann
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Tetzlaff
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marie I Hiller
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bettina Jagemann
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rambabu Surabattula
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Diana Leeming
- Research and Development, Nordic Bioscience, Biomarkers and Research A/S, Herlev, Denmark
| | - Morten Karsdal
- Research and Development, Nordic Bioscience, Biomarkers and Research A/S, Herlev, Denmark
| | - Erika Monguzzi
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Guido Schachschal
- Department of Interdisciplinary Endoscopy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Rösch
- Department of Interdisciplinary Endoscopy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Corinna Bang
- Institute for Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute for Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Ansgar W Lohse
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, Hamburg, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany.,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christoph Schramm
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, Hamburg, Germany.,Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
42
|
Talipova D, Smagulova A, Poddighe D. Toll-like Receptors and Celiac Disease. Int J Mol Sci 2022; 24:265. [PMID: 36613709 PMCID: PMC9820541 DOI: 10.3390/ijms24010265] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated disorder triggered by dietary gluten intake in some genetically predisposed individuals; however, the additional non-HLA-related genetic factors implicated in CD immunopathogenesis are not well-defined. The role of the innate immune system in autoimmunity has emerged in the last few years. Genetic polymorphisms of some pattern-recognition receptors, including toll-like receptors (TLRs), have been associated with several autoimmune disorders. In this review, we summarize and discuss the evidence from basic research and clinical studies as regards the potential role of TLRs in CD immunopathogenesis. The evidence supporting the role of TLRs in CD immunopathogenesis is limited, especially in terms of basic research. However, differences in the expression and activation of TLRs between active CD patients from one side, and controls and treated CD patients from the other side, have been described in some clinical studies. Therefore, TLRs may be part of those non-HLA-related genetic factors implicated in CD etiopathogenesis, considering their potential role in the interaction between the host immune system and some environmental factors (including viral infections and gut microbiota), which are included in the list of candidate agents potentially contributing to the determination of CD risk in genetically predisposed individuals exposed to dietary gluten intake. Further basic research and clinical studies focused on TLRs in the context of CD and other gluten-related disorders are needed.
Collapse
Affiliation(s)
- Diana Talipova
- Department of Medicine, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
| | - Aiganym Smagulova
- Department of Medicine, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
| | - Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan
| |
Collapse
|
43
|
Allergenic food protein consumption is associated with systemic IgG antibody responses in non-allergic individuals. Immunity 2022; 55:2454-2469.e6. [PMID: 36473469 DOI: 10.1016/j.immuni.2022.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/01/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Although food-directed immunoglobulin E (IgE) has been studied in the context of allergies, the prevalence and magnitude of IgG responses against dietary antigens are incompletely characterized in the general population. Here, we measured IgG binding against food and environmental antigens obtained from allergen databases and the immune epitope database (IEDB), represented in a phage displayed library of 58,233 peptides. By profiling blood samples of a large cohort representing the average adult Israeli population (n = 1,003), we showed that many food antigens elicited systemic IgG in up to 50% of individuals. Dietary intake of specific food protein correlated with antibody binding, suggesting that diet can shape the IgG epitope repertoire. Our work documents abundant systemic IgG responses against food antigens and provides a reference map of the exact immunogenic epitopes on a population scale, laying the foundation to unravel the role of food- and environmental antigen-directed antibody binding in disease contexts.
Collapse
|
44
|
Borrelli DE Andreis F, Schiepatti A, Gibiino G, Fabbri C, Baiardi P, Biagi F. Is it time to rethink the burden of non-coeliac gluten sensitivity? A systematic review. Minerva Gastroenterol (Torino) 2022; 68:442-449. [PMID: 34929997 DOI: 10.23736/s2724-5985.21.03077-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Non-coeliac gluten sensitivity (NCGS) is still a poorly defined clinical condition. This review aims to describe the clinical features of subjects with a symptomatic response to gluten intake, and to estimate the prevalence of NCGS. EVIDENCE ACQUISITION Literature search was conducted in accordance with PRISMA recommendations. The PubMed database was searched for original articles until 1st June 2020. EVIDENCE SYNTHESIS We identified 30 relevant articles, including 14 studies that investigated NCGS through a double-blind, placebo-controlled crossover trial (DBPCC), and 16 that examined the role of gluten in causing symptoms without a DBPCC. We found that regardless of the diagnostic work up, gluten-sensitive patients were predominately middle-aged females complaining of abdominal pain, bloating and diarrhea. The pooled prevalence of NCGS after DBPCC was 24% (5-34%). Subjects with irritable bowel syndrome or self-reporting gluten intolerance accounted for the vast majority of the patients who did not start a DBPCC. A symptomatic response to a gluten-free diet (GFD) occurred in between 7% and 93% of patients. No data on long-term outcomes of NCGS individuals were reported. CONCLUSIONS Clinical features of NCGS patients did not differ among all the included studies, whereas prevalence figures are rather heterogeneous. Long-term benefit of a GFD on these patients still needs to be ascertained.
Collapse
Affiliation(s)
- Federica Borrelli DE Andreis
- Gastroenterology Unit, IRCCS Pavia Institute, Istituti Clinici Scientifici Maugeri, University of Pavia, Pavia, Italy
| | - Annalisa Schiepatti
- Gastroenterology Unit, IRCCS Pavia Institute, Istituti Clinici Scientifici Maugeri, University of Pavia, Pavia, Italy -
| | - Giulia Gibiino
- Gastroenterology and Digestive Endoscopy Unit, Morgagni-Pierantoni Hospital, Forlì, Italy.,M. Bufalini Hospital, AUSL Romagna, Cesena, Italy
| | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Morgagni-Pierantoni Hospital, Forlì, Italy.,M. Bufalini Hospital, AUSL Romagna, Cesena, Italy
| | - Paola Baiardi
- Scientific Direction, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Federico Biagi
- Gastroenterology Unit, IRCCS Pavia Institute, Istituti Clinici Scientifici Maugeri, University of Pavia, Pavia, Italy
| |
Collapse
|
45
|
Malkovics T, Joura MI, Koszorú K, Sárdy M. [Dermatitis herpetiformis and other forms of wheat sensitivity]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2022; 74:955-960. [PMID: 37882829 PMCID: PMC10661880 DOI: 10.1007/s00105-023-05243-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Wheat sensitivity is a collective term for several, especially gastrointestinal, diseases that occur as part of a hypersensitivity reaction after wheat consumption. The symptoms, which are mostly similar to those of irritable bowel syndrome, are often accompanied by skin lesions. In addition to celiac disease and dermatitis herpetiformis, the cutaneous manifestation of celiac disease, wheat sensitivity also includes nonceliac gluten sensitivity (NCGS), allergic nickel contact mucositis, wheat allergy, amylase-trypsin inhibitor intolerance, and fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) intolerance. OBJECTIVES This review article aims to provide an overview of the clinical, especially dermatological and gastrointestinal features of the different forms of wheat sensitivity. Diagnosis and therapeutic management are also discussed. MATERIALS AND METHODS A selective literature search was carried out with evaluation of our own clinical data. RESULTS The skin lesions in dermatitis herpetiformis are very disease-specific. In contrast, wheat allergy often shares signs and symptoms with many other diseases. Other forms of wheat sensitivity cause primarily gastrointestinal abnormalities, but extra-intestinal manifestations can also occur. Their diagnosis is often complex and requires cross-disciplinary collaboration with experts in gastroenterology. The therapy consists of a wheat- or gluten-free diet. CONCLUSIONS Knowledge of the different and frequently occurring dermatological signs of wheat sensitivity is of great importance, because dermatological manifestations associated with gastrointestinal pathology, intolerance reactions, and allergies appear more and more frequently.
Collapse
Affiliation(s)
- T Malkovics
- Klinik für Dermatologie, Venerologie und Dermatoonkologie, Fakultät für Medizin, Semmelweis Universität, Mária u. 41, 1085, Budapest, Ungarn.
| | - M I Joura
- Klinik für Dermatologie, Venerologie und Dermatoonkologie, Fakultät für Medizin, Semmelweis Universität, Mária u. 41, 1085, Budapest, Ungarn
- Universitätsklinik für Dermatologie, Medizinische Universität Wien, Wien, Österreich
| | - K Koszorú
- Klinik für Dermatologie, Venerologie und Dermatoonkologie, Fakultät für Medizin, Semmelweis Universität, Mária u. 41, 1085, Budapest, Ungarn
| | - M Sárdy
- Klinik für Dermatologie, Venerologie und Dermatoonkologie, Fakultät für Medizin, Semmelweis Universität, Mária u. 41, 1085, Budapest, Ungarn
- Klinik für Dermatologie und Allergologie, Klinikum der Universität München (LMU), München, Deutschland
| |
Collapse
|
46
|
Simonetti E, Bosi S, Negri L, Dinelli G. Amylase Trypsin Inhibitors (ATIs) in a Selection of Ancient and Modern Wheat: Effect of Genotype and Growing Environment on Inhibitory Activities. PLANTS (BASEL, SWITZERLAND) 2022; 11:3268. [PMID: 36501308 PMCID: PMC9738579 DOI: 10.3390/plants11233268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Wheat amylase-trypsin inhibitors (ATIs) are a family of plant defense proteins with an important role in human health for their involvement in allergies, celiac disease and non-celiac wheat sensitivity. Information about the differences in ATI activities among wheat genotypes and the influence of the growing environment is scarce. Therefore, ten selected wheat accessions with different ploidy level and year of release, previously characterized for their ATI gene sequences, were grown during three consecutive crop years at two growing areas and used for in vitro ATI activities. The contributions of the genotype and the crop year were significant for both activities. The hexaploid wheat genotypes showed the highest inhibitory activities. Einkorn had a peculiar behavior showing the lowest alpha-amylase inhibitory activity, but the highest trypsin inhibitory activity. It was not possible to observe any trend in ATI activities as a function of the release year of the wheat samples. The two inhibitory activities were differently affected by the growing conditions and were negatively correlated with the protein content. This information can be important in understanding the extent of variation of ATI inhibitory properties in relation to the wheat genotype and the growing environment and the impact of ATIs, if any, on human health and nutrition.
Collapse
Affiliation(s)
| | - Sara Bosi
- Correspondence: ; Tel.: +39-051-2096669; Fax: +39-051-2096241
| | | | | |
Collapse
|
47
|
Aguilera-Lizarraga J. Gut reactions: emerging mechanisms of abdominal pain from food intake. Am J Physiol Gastrointest Liver Physiol 2022; 323:G401-G409. [PMID: 36126222 DOI: 10.1152/ajpgi.00173.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Abdominal pain, which is a form of visceral pain, is a highly prevalent symptom worldwide frequently occurring following food ingestion. Its pathophysiology is complex, and many factors, including intestinal environmental cues, the immune system, or the molecular composition of foods, can influence the development of postprandial abdominal pain. Because of the poor efficacy of drug treatments, current strategies are often limited to the exclusion of culprit food(s) from the diet. However, there are two important limitations to this approach. First, patients suffering from food-induced abdominal pain usually recognize several food items as the cause of their gastrointestinal symptoms. Second, not all offending foods can always be identified by these patients. Newly identified mechanisms involving neuroimmune interactions and their communication with the intestinal microbiota shed light on the development of new therapeutic strategies. In this Mini-Review, these novel mechanisms and relevance of such findings are highlighted.
Collapse
Affiliation(s)
- Javier Aguilera-Lizarraga
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
48
|
Shah A, Kang S, Talley NJ, Do A, Walker MM, Shanahan ER, Koloski NA, Jones MP, Keely S, Morrison M, Holtmann GJ. The duodenal mucosa associated microbiome, visceral sensory function, immune activation and psychological comorbidities in functional gastrointestinal disorders with and without self-reported non-celiac wheat sensitivity. Gut Microbes 2022; 14:2132078. [PMID: 36303431 PMCID: PMC9621048 DOI: 10.1080/19490976.2022.2132078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Frequently, patients with functional gastrointestinal disorders (FGIDs) report intolerance of wheat products. We compared gastrointestinal symptoms, sensory function, psychiatric comorbidities, gut-homing immune cells, and duodenal mucosa-associated microbiome (d-MAM) in FGID patients and controls with and without self-reported wheat sensitivity (SR-NCWS). We recruited 40 FGID patients and 20 controls referred by GPs for treatment. Gastrointestinal/extraintestinal symptoms, visceral sensory function, psychological comorbidities, and SR-NCWS were assessed in a standardized approach. Peripheral gut homing T-cells (CD4+α4+β7+CCR9+/CD8+α4+β7+CCR9+) were quantified, and the d-MAM was assessed by DNA sequencing for 46 subjects. Factors of bacterial genera were extracted utilizing factor analysis with varimax rotation and factors univariately associated with FGID or SR-NCWS included in a subsequent multivariate analysis of variance to identify statistically independent discriminators. Anxiety scores (p < .05) and increased symptom responses to a nutrient challenge (p < .05) were univariately associated with FGID. Gut homing T-cells were increased in FGID patients with SR-NCWS compared to other groups (p all <0.05). MANOVA revealed that anxiety (p = .03), visceral sensory function (p = 0.007), and a d-MAM factor comprise members of the Alloprevotella, Prevotella, Peptostreptococcus, Leptotrichia, and Veillonella lineages were significantly (p = .001) associated with FGID, while gut homing CD4+α4+ β7+CCR9+ T-cells were associated (p = .002) with SR-NCWS. Compared to controls, patients with and without SR-NCWS show that there are shifts in the amplicon sequence variants within specific bacterial genera between the FGID subgroups (particularly Prevotella and Streptococcus) as well as distinct bacterial taxa discriminatory for the two different FGID subtypes. Compared to controls, both FGID patients with and without SR-NCWS have an increased symptom response to a standardized nutrient challenge and increased anxiety scores. The FGID patients with SR-NCWS - as compared to FGID without SR-NCWS (and controls without SR-NCWS) - have increased gut homing T-cells. The d-MAM profiles suggest species and strain-based variations between the two FGID subtypes and in comparison to controls.
Collapse
Affiliation(s)
- Ayesha Shah
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia,Translational Research Institute Queensland, Australia,Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Seungha Kang
- Faculty of Medicine, University of Queensland, Brisbane, Australia,University of Queensland Diamantina Institute, Woolloongabba, Australia
| | - Nicholas J Talley
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Anh Do
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia,Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Marjorie M Walker
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Erin R Shanahan
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia,Translational Research Institute Queensland, Australia
| | - Natasha A Koloski
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia,Translational Research Institute Queensland, Australia,Faculty of Medicine, University of Queensland, Brisbane, Australia,College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Michael P Jones
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Mark Morrison
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia,Faculty of Medicine, University of Queensland, Brisbane, Australia,University of Queensland Diamantina Institute, Woolloongabba, Australia,CONTACT Mark Morrison
| | - Gerald J Holtmann
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia,Translational Research Institute Queensland, Australia,Faculty of Medicine, University of Queensland, Brisbane, Australia,Gerald J Holtmann Princess Alexandra Hospital, Brisbane Department of Gastroenterology and Hepatology & University of Queensland 199 Ipswich Road, Woolloongabba, Queensland, Australia
| |
Collapse
|
49
|
Inflammatory Bowel Disease and Customized Nutritional Intervention Focusing on Gut Microbiome Balance. Nutrients 2022; 14:nu14194117. [PMID: 36235770 PMCID: PMC9572914 DOI: 10.3390/nu14194117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/24/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Inflammatory bowel disease (IBD) represents a chronic relapsing–remitting condition affecting the gastrointestinal system. The specific triggering IBD elements remain unknown: genetic variability, environmental factors, and alterations in the host immune system seem to be involved. An unbalanced diet and subsequent gut dysbiosis are risk factors, too. This review focuses on the description of the impact of pro- and anti-inflammatory food components on IBD, the role of different selected regimes (such as Crohn’s Disease Exclusion Diet, Immunoglobulin Exclusion Diet, Specific Carbohydrate Diet, LOFFLEX Diet, Low FODMAPs Diet, Mediterranean Diet) in the IBD management, and their effects on the gut microbiota (GM) composition and balance. The purpose is to investigate the potential positive action on IBD inflammation, which is associated with the exclusion or addition of certain foods or nutrients, to more consciously customize the nutritional intervention, taking also into account GM fluctuations during both disease flare-up and remission.
Collapse
|
50
|
Rej A, Avery A, Aziz I, Black CJ, Bowyer RK, Buckle RL, Seamark L, Shaw CC, Thompson J, Trott N, Williams M, Sanders DS. Diet and irritable bowel syndrome: an update from a UK consensus meeting. BMC Med 2022; 20:287. [PMID: 36096789 PMCID: PMC9469508 DOI: 10.1186/s12916-022-02496-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
There has been a renewed interest in the role of dietary therapies to manage irritable bowel syndrome (IBS), with diet high on the agenda for patients. Currently, interest has focussed on the use of traditional dietary advice (TDA), a gluten-free diet (GFD) and the low FODMAP diet (LFD). A consensus meeting was held to assess the role of these dietary therapies in IBS, in Sheffield, United Kingdom.Evidence for TDA is from case control studies and clinical experience. Randomised controlled trials (RCT) have demonstrated the benefit of soluble fibre in IBS. No studies have assessed TDA in comparison to a habitual or sham diet. There have been a number of RCTs demonstrating the efficacy of a GFD at short-term follow-up, with a lack of long-term outcomes. Whilst gluten may lead to symptom generation in IBS, other components of wheat may also play an important role, with recent interest in the role of fructans, wheat germ agglutinins, as well as alpha amylase trypsin inhibitors. There is good evidence for the use of a LFD at short-term follow-up, with emerging evidence demonstrating its efficacy at long-term follow-up. There is overlap between the LFD and GFD with IBS patients self-initiating gluten or wheat reduction as part of their LFD. Currently, there is a lack of evidence to suggest superiority of one diet over another, although TDA is more acceptable to patients.In view of this evidence, our consensus group recommends that dietary therapies for IBS should be offered by dietitians who first assess dietary triggers and then tailor the intervention according to patient choice. Given the lack of dietetic services, novel approaches such as employing group clinics and online webinars may maximise capacity and accessibility for patients. Further research is also required to assess the comparative efficacy of dietary therapies to other management strategies available to manage IBS.
Collapse
Affiliation(s)
- A Rej
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK.
| | - A Avery
- Division of Nutritional Sciences, School of Biosciences, University of Nottingham, Nottingham, UK
| | - I Aziz
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - C J Black
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - R K Bowyer
- Department of Nutrition and Dietetics, Royal United Hospitals NHS Foundation Trust, Bath, UK
| | - R L Buckle
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - L Seamark
- Specialist Gastroenterology Community Dietetic Service, Somerset Partnership NHS Foundation Trust, Bridgwater, UK
| | - C C Shaw
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - J Thompson
- Information Manager/Specialist Gastroenterology Dietitian, Guts UK Charity, 3 St Andrews Place, London, NW1 4LB, UK
| | - N Trott
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - M Williams
- Specialist Gastroenterology Community Dietetic Service, Somerset Partnership NHS Foundation Trust, Bridgwater, UK
| | - D S Sanders
- Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| |
Collapse
|