1
|
Aslan ES, Meral G, Aydin E, Caglayan S, Altundag A, Demirkol S, Gormus G, Solak M, Ayaz F. The Effect of a Casein and Gluten-Free Diet on the Epigenetic Characteristics of FoxP3 in Patients With Hashimoto's Thyroiditis. Cureus 2024; 16:e63208. [PMID: 38952602 PMCID: PMC11216022 DOI: 10.7759/cureus.63208] [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] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Background Hashimoto's thyroiditis (HT) is an autoimmune thyroid disease characterized by inflammation and dysfunction of the thyroid gland, resulting in hypothyroidism, it results in impaired thyroid hormone generation and mimics hypothyroidism. The disease involves complex interactions among genetic, environmental, and epigenetic factors, particularly affecting the regulation of T regulatory (Treg) cells, including CD4 + foxp3 + T cells. Treg cells, defined as CD4 + T cells, rely on the expression of the foxp3 transcription factor, which is crucial for their development and differentiation. Disruptions in this regulation can lead to immune dysregulation and potential proinflammatory responses. The study focuses on investigating the impact of dietary patterns on the epigenetic changes in the foxp3 gene, a key player in the development of HT. The primary aim was to evaluate how eliminating gluten and casein proteins from dietary regimens may influence the methylation levels of the foxp3 gene, considering the potential link between these dietary components and the triggering of autoimmune diseases. Methods An epigenetic analysis of the foxp3 gene in HT patients who were strictly following a dietary plan compared with the control group. For the epigenetic study, a methylation analysis experiment was conducted. Results Our findings revealed a notable reduction in foxp3 gene methylation levels among HT patients who adhered to a diet excluding casein and gluten. The control maintained normal dietary guidelines and showed no significant alterations in methylation levels. Discussion The laboratory values showed a decrease in methylation levels of the foxp3 gene, with statistical significance indicated as *p<0.005, **p<0.001, ***p<0.0001, suggesting a potential enhancement in its expression which could have profound implications for immune system regulation. Disruptions in the foxp3 pathway are crucial in the development of autoimmune disorders, where altered activity hinders the regulation of T cell (Treg) development, ultimately contributing to conditions like HT disease. These findings imply that nutritional interventions, especially for individuals with HT, could potentially be a strategy for mitigating autoimmunity through epigenetic mechanisms.
Collapse
Affiliation(s)
- Elif S Aslan
- Molecular Biology and Genetics, Biruni University, Istanbul, TUR
| | - Gulsen Meral
- Molecular Biology and Genetics, Epigenetic Coaching, Norwich, GBR
- Molecular Biology and Genetics, Biruni University, Istanbul, TUR
| | - Ece Aydin
- Molecular Biology and Genetics, Biruni University, Istanbul, TUR
| | - Sinan Caglayan
- Endocrinology and Metabolic Diseases, Demiroğlu Bilim Üniversitesi, Istanbul, TUR
| | | | - Seyda Demirkol
- Computer Science with Data Science, University of Sunderland, Sunderland, GBR
| | - Gizem Gormus
- Nutrition and Dietetics, Florence Nightingale Community Hospital, Istanbul, TUR
| | | | - Furkan Ayaz
- Molecular Biology and Genetics, Biruni University, Istanbul, TUR
| |
Collapse
|
2
|
Metyas C, Aung TT, Cheung J, Joseph M, Ballester AM, Metyas S. Diet and Lifestyle Modifications for Fibromyalgia. Curr Rheumatol Rev 2024; 20:405-413. [PMID: 38279728 PMCID: PMC11107431 DOI: 10.2174/0115733971274700231226075717] [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: 08/16/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 01/28/2024]
Abstract
Fibromyalgia (FM) is a complex, widespread pain disorder characterized by symptoms such as fatigue, sleep deprivation, mental fog, mood swings, and headaches. Currently, there are only three FDA-approved medications for FM patients: duloxetine, milnacipran, and pregabalin, with outcomes frequently being inadequate. This research team aims to investigate the effects of diet and lifestyle modifications on FM, with emphasis on anti-inflammatory diet, antioxidants, and gluten-free diets, as well as supplementation with Magnesium, CQ10, and Vitamin D, microbiome, sleep, exercise, and cognitive behavioral therapy. We reviewed the pathophysiology of certain foods that can be proinflammatory with the release of cytokines leading to activation of pain, fatigue and aggravation of the majority of Fibromyalgia symptoms. A literature review was performed by identifying FM articles published between 1994 and 2022 via PubMed and EMBASE databases, with particular emphasis on randomized controlled trials, meta-analysis, and evidence-based treatment guidelines. This review article was completed by a comprehensive narrative review process, in which our team systematically examined relevant scientific literature to provide a comprehensive overview of the significant role that diet and other lifestyle modifications play in mediating symptoms of Fibromyalgia. We propose that diet modifications and lifestyle changes, such as sleep, exercise, and weight loss, can be important steps in managing FM.
Collapse
Affiliation(s)
- Caroline Metyas
- Department of Rheumatology, Covina Arthritis Clinic, 500 West San Bernadino Rd, Suite A, United States
| | - Tun Tun Aung
- Department of Rheumatology, Covina Arthritis Clinic, 500 West San Bernadino Rd, Suite A, United States
| | - Jennifer Cheung
- Department of Rheumatology, Covina Arthritis Clinic, 500 West San Bernadino Rd, Suite A, United States
| | - Marina Joseph
- Department of Rheumatology, Covina Arthritis Clinic, 500 West San Bernadino Rd, Suite A, United States
| | - Andrew M Ballester
- Department of Rheumatology, Covina Arthritis Clinic, 500 West San Bernadino Rd, Suite A, United States
| | - Samy Metyas
- Department of Rheumatology, Covina Arthritis Clinic, 500 West San Bernadino Rd, Suite A, United States
| |
Collapse
|
3
|
Johansen VBI, Josefsen K, Antvorskov JC. The Impact of Dietary Factors during Pregnancy on the Development of Islet Autoimmunity and Type 1 Diabetes: A Systematic Literature Review. Nutrients 2023; 15:4333. [PMID: 37892409 PMCID: PMC10609322 DOI: 10.3390/nu15204333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
AIMS AND HYPOTHESIS The incidence of type 1 diabetes mellitus in children is considerably increasing in western countries. Thus, identification of the environmental determinants involved could ultimately lead to disease prevention. Here, we aimed to systematically review (PROSPERO ID: CRD42022362522) the current evidence of the association between maternal dietary factors during gestation and the risk of developing type 1 diabetes and/or islet autoimmunity (IA) in murine and human offspring. METHODS In accordance with PRISMA guidelines, the present systematic review searched PubMed and Scopus (n = 343) for different combinations of MeSH terms, such as type 1 diabetes, diet, islet autoimmunity, prenatal, nutrient, gluten, gliadin, vitamin, milk, and fibers. RESULTS We found that the most investigated dietary factors in the present literature were gluten, dietary advanced glycosylated end products (dAGEs), vitamin D, fatty acids, and iron. The results concerning prenatal exposure to a gluten-free environment showed a consistently protective effect on the development of IA. Prenatal exposures to vitamin D and certain fatty acids appeared to protect against the development of IA, whereas in utero iron and fat exposures correlated with increased risks of IA. CONCLUSION We conclude that a definite association is not established for most factors investigated as the literature represents a heterogeneous pool of data, although fetal exposures to some maternal dietary components, such as gluten, show consistent associations with increased risks of IA. We suggest that human prospective dietary intervention studies in both cohort and clinical settings are crucial to better evaluate critical and protective prenatal exposures from the maternal diet during pregnancy.
Collapse
Affiliation(s)
- Valdemar Brimnes Ingemann Johansen
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Department of Biology, Faculty of Science, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark; (K.J.); (J.C.A.)
| | - Knud Josefsen
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark; (K.J.); (J.C.A.)
| | - Julie Christine Antvorskov
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark; (K.J.); (J.C.A.)
- Steno Diabetes Center, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark
| |
Collapse
|
4
|
Arora S, Tayade A, Bhardwaj T, Pathak SS. Unveiling the Link: A Comprehensive Narrative Review of the Relationship Between Type 1 Diabetes Mellitus and Celiac Disease. Cureus 2023; 15:e47726. [PMID: 38022113 PMCID: PMC10676227 DOI: 10.7759/cureus.47726] [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: 08/21/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune condition with a genetic predisposition. It has underlying autoimmune destruction of the pancreatic cells that produce insulin. It is often accompanied by other autoimmune conditions. This article focuses on celiac disease (CD), also an autoimmune disease. It is caused by gluten exposure. Both these conditions have genetic predisposing factors. Apart from the genetic background, aberrant small intestine immune response, inflammation, and different grades of enteropathy present in T1DM and CD are the same. With a mean frequency of 8%, the CD frequency of T1DM ranges from 3 to 16%. All T1DM patients should undergo serological testing for CD using antibodies to tissue transglutaminase at the time of T1DM onset. Individuals with T1DM and those accompanied by CD must follow a diet with no gluten. To outline the steps that can avert the development of these disorders and reduce the morbidity of the affected people, a complete understanding of the intricate pathophysiology of T1DM and its connection to CD has been undertaken in this review. The use of resources, such as PubMed and Google Scholar, has made this possible.
Collapse
Affiliation(s)
- Sanvi Arora
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ayush Tayade
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Tanya Bhardwaj
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Swanand S Pathak
- Pharmacology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
5
|
Johansen VBI, Færø D, Buschard K, Kristiansen K, Pociot F, Kiilerich P, Josefsen K, Haupt-Jorgensen M, Antvorskov JC. A Gluten-Free Diet during Pregnancy and Early Life Increases Short Chain Fatty Acid-Producing Bacteria and Regulatory T Cells in Prediabetic NOD Mice. Cells 2023; 12:1567. [PMID: 37371037 PMCID: PMC10297205 DOI: 10.3390/cells12121567] [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: 05/24/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The incidence of the autoimmune disease type 1 diabetes is increasing, likely caused by environmental factors. A gluten-free diet has previously been shown to ameliorate autoimmune diabetes in non-obese diabetic (NOD) mice and humans. Although the exact mechanisms are not understood, interventions influencing the intestinal microbiota early in life affect the risk of type 1 diabetes. Here, we characterize how NOD mice that are fed a gluten-free (GF) diet differ from NOD mice that are fed a gluten-containing standard (STD) diet in terms of their microbiota composition by 16S rRNA gene amplicon sequencing and pancreatic immune environment by real-time quantitative PCR at the prediabetic stage at 6 and 13 weeks of age. Gut microbiota analysis revealed highly distinct microbiota compositions in both the cecum and the colon of GF-fed mice compared with STD-fed mice. The microbiotas of the GF-fed mice were characterized by an increased Firmicutes/Bacteroidetes ratio, an increased abundance of short chain fatty acid (particularly butyrate)-producing bacteria, and a reduced abundance of Lactobacilli compared with STD mice. We found that the insulitis score in the GF mice was significantly reduced compared with the STD mice and that the markers for regulatory T cells and T helper 2 cells were upregulated in the pancreas of the GF mice. In conclusion, a GF diet during pre- and early post-natal life induces shifts in the cecal and colonic microbiota compatible with a less inflammatory environment, providing a likely mechanism for the protective effect of a GF diet in humans.
Collapse
Affiliation(s)
| | - Daisy Færø
- Department of Pathology, Bartholin Institute, Rigshospitalet, 2100 Copenhagen, Denmark; (D.F.); (K.B.); (K.J.); (M.H.-J.)
| | - Karsten Buschard
- Department of Pathology, Bartholin Institute, Rigshospitalet, 2100 Copenhagen, Denmark; (D.F.); (K.B.); (K.J.); (M.H.-J.)
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, August Krogh Building, University of Copenhagen, Universitetsparken 13, 2200 Copenhagen, Denmark; (K.K.); (P.K.)
| | - Flemming Pociot
- Steno Diabetes Center, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark;
| | - Pia Kiilerich
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, August Krogh Building, University of Copenhagen, Universitetsparken 13, 2200 Copenhagen, Denmark; (K.K.); (P.K.)
- Department for Congenital Disorders, Danish Center for Neonatal Screening, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Knud Josefsen
- Department of Pathology, Bartholin Institute, Rigshospitalet, 2100 Copenhagen, Denmark; (D.F.); (K.B.); (K.J.); (M.H.-J.)
| | - Martin Haupt-Jorgensen
- Department of Pathology, Bartholin Institute, Rigshospitalet, 2100 Copenhagen, Denmark; (D.F.); (K.B.); (K.J.); (M.H.-J.)
| | - Julie Christine Antvorskov
- Department of Pathology, Bartholin Institute, Rigshospitalet, 2100 Copenhagen, Denmark; (D.F.); (K.B.); (K.J.); (M.H.-J.)
- Steno Diabetes Center, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark;
| |
Collapse
|
6
|
Aguilar EC, Fernandes-Braga W, Leocádio PCL, Campos GP, Lemos VS, de Oliveira RP, Caetano de Faria AM, Dos Santos Aggum Capettini L, Alvarez-Leite JI. Dietary gluten worsens hepatic steatosis by increasing inflammation and oxidative stress in ApoE-/- mice fed a high-fat diet. Food Funct 2023; 14:3332-3347. [PMID: 36940107 DOI: 10.1039/d3fo00149k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disorder in the world. We have seen that gluten intake exacerbated obesity and atherosclerosis in apolipoprotein E knockout (ApoE-/-) mice. In this study, we investigated the effect of gluten consumption on inflammation and oxidative stress in the liver of mice with NAFLD. Male ApoE-/- mice were fed a gluten-free (GF-HFD) or gluten-containing (G-HFD) high-fat diet for 10 weeks. Blood, liver, and spleen were collected to perform the analyses. The animals of the gluten group had increased hepatic steatosis, followed by increased serum AST and ALT. Gluten intake increased hepatic infiltration of neutrophils, macrophages, and eosinophils, as well as the levels of chemotaxis-related factors CCL2, Cxcl2, and Cxcr3. The production of the TNF, IL-1β, IFNγ, and IL-4 cytokines in the liver was also increased by gluten intake. Furthermore, gluten exacerbated the hepatic lipid peroxidation and nitrotyrosine deposition, which were associated with increased production of ROS and nitric oxide. These effects were related to increased expression of NADPH oxidase and iNOS, as well as decreased activity of superoxide dismutase and catalase enzymes. There was an increased hepatic expression of the NF-κB and AP1 transcription factors, corroborating the worsening effect of gluten on inflammation and oxidative stress. Finally, we found an increased frequency of CD4+FOXP3+ lymphocytes in the spleen and increased gene expression of Foxp3 in the livers of the G-HFD group. In conclusion, dietary gluten aggravates NAFLD, exacerbating hepatic inflammation and oxidative stress in obese ApoE-deficient mice.
Collapse
Affiliation(s)
- Edenil Costa Aguilar
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais/UFMG, Caixa Postal 486, 30161-970 Belo Horizonte, Brazil.
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais/UFMG, Caixa Postal 486, 30161-970 Belo Horizonte, Brazil. .,Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, USA
| | - Paola Caroline Lacerda Leocádio
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais/UFMG, Caixa Postal 486, 30161-970 Belo Horizonte, Brazil.
| | - Gianne Paul Campos
- Department of Pharmacology, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| | - Virginia Soares Lemos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| | | | - Ana Maria Caetano de Faria
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais/UFMG, Caixa Postal 486, 30161-970 Belo Horizonte, Brazil.
| | | | - Jacqueline I Alvarez-Leite
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais/UFMG, Caixa Postal 486, 30161-970 Belo Horizonte, Brazil.
| |
Collapse
|
7
|
Cheng YW, Luo Y, Zheng SJ, Xiao JH. Combination therapy with human amniotic epithelial cells and hyaluronic acid promotes immune balance recovery in type 1 diabetic rats through local engraftment. Scand J Immunol 2023; 97:e13246. [PMID: 36575914 DOI: 10.1111/sji.13246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
Stem cell engraftment is currently a promising approach for type 1 diabetes mellitus (T1DM) treatment. In our previous study, engraftment of a combination of human amniotic epithelial cells (hAECs) and hyaluronic acid (HA) showed potent anti-diabetic effect in streptozotocin (STZ)-induced T1DM mice via tail vein injection. Here, we adopted a different route of stem cell delivery, that is via pancreatic subcapsular transplantation. This combined local engraftment of hAECs and HA in STZ-induced T1DM rats showed potent anti-diabetic activity, leading to stronger hypoglycaemia, more intact islet structure and increased number of insulin-positive cells compared with those with hAECs or insulin treatments. Engraftment of hAECs alone increased the proportion of Th1 and T-reg cells and decreased the proportion of Th2 and Th17 cells to protect islet β cells in STZ-induced T1DM rats, whereas the combined engraftment of hAECs and HA showed more potent regulatory capacity, considerably decreased the level of TNF-α and IL-17 and increased the level of TGF-β1 compared with those by other treatments. The potent synergistic effect of HA contributed to the recovery of immune balance in the diabetic rat model, thereby suggesting a new strategy for effective treatment of T1DM.
Collapse
Affiliation(s)
- Ya-Wei Cheng
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Luo
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Zunyi Municipal Key Laboratory of Medicinal Biotechnology & Guizhou Provincial Research Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shu-Juan Zheng
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jian-Hui Xiao
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Zunyi Municipal Key Laboratory of Medicinal Biotechnology & Guizhou Provincial Research Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
8
|
Li F, Hu X, Wu Z, Yang Q, Sa Q, Ren W, Wang T, Ji Z, Li N, Huang J, Lei L. Untargeted metabolomics reveals alternations in metabolism of bovine mammary epithelial cells upon IFN-γ treatment. BMC Vet Res 2023; 19:44. [PMID: 36765367 PMCID: PMC9921584 DOI: 10.1186/s12917-023-03588-2] [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: 09/10/2022] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND IFN-γ is a pleiotropic cytokine that has been shown to affect multiple cellular functions of bovine mammary epithelial cells (BMECs) including impaired milk fat synthesis and induction of malignant transformation via depletion of arginine, one of host conditionally essential amino acids. But the molecular mechanisms of these IFN-γ induced phenotypes are still unknown. METHODS BMECs were treated with IFN-γ for 6 h, 12 h, and 24 h. The metabolomic profiling in BMECs upon IFN-γ induction were assessed using untargeted ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) metabolomic analysis. Key differentially expressed metabolites (DEMs) were quantified by targeted metabolomics. RESULTS IFN-γ induction resulted in significant differences in the contents of metabolites. Untargeted analysis identified 221 significantly DEMs, most of which are lipids and lipid-like molecules, organic acids and derivatives, organ heterocyclic compounds and benzenoids. According to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, DEMs were enriched in fructose and mannose metabolism, phosphotransferase system (PTS), β-alanine metabolism, arginine and proline metabolism, methane metabolism, phenylalanine metabolism, and glycolysis/gluconeogenesis. Quantification of selected key DEMs by targeted metabolomics showed significantly decreased levels of D-(-)-mannitol, argininosuccinate, and phenylacetylglycine (PAG), while increased levels in S-hydroxymethylglutathione (S-HMG) and 2,3-bisphospho-D-glyceric acid (2,3-BPG). CONCLUSIONS These results provide insights into the metabolic alterations in BMECs upon IFN-γ induction and indicate potential theoretical basis for clarifying IFN-γ-induced diseases in mammary gland.
Collapse
Affiliation(s)
- Fengyang Li
- grid.64924.3d0000 0004 1760 5735State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, 1977 Xinzhu Road, Changchun, 130062 China
| | - Xiuhong Hu
- grid.64924.3d0000 0004 1760 5735Department of First Hospital, Jilin University, 1 Xinmin Street, Changchun, 130021 China ,Shannan Hospital, Shannan, 856099 China
| | - Zengshuai Wu
- grid.64924.3d0000 0004 1760 5735State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, 1977 Xinzhu Road, Changchun, 130062 China
| | - Qiulei Yang
- grid.64924.3d0000 0004 1760 5735State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, 1977 Xinzhu Road, Changchun, 130062 China
| | - Qila Sa
- grid.64924.3d0000 0004 1760 5735State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, 1977 Xinzhu Road, Changchun, 130062 China
| | - Wenbo Ren
- grid.64924.3d0000 0004 1760 5735Department of First Hospital, Jilin University, 1 Xinmin Street, Changchun, 130021 China
| | - Tingting Wang
- grid.64924.3d0000 0004 1760 5735Department of First Hospital, Jilin University, 1 Xinmin Street, Changchun, 130021 China
| | - Zhengchao Ji
- grid.64924.3d0000 0004 1760 5735Department of First Hospital, Jilin University, 1 Xinmin Street, Changchun, 130021 China
| | - Na Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, 1977 Xinzhu Road, Changchun, 130062, China.
| | - Jing Huang
- Department of First Hospital, Jilin University, 1 Xinmin Street, Changchun, 130021, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, 1977 Xinzhu Road, Changchun, 130062, China.
| |
Collapse
|
9
|
Gluten-Free Diet in Co-Existent Celiac Disease and Type 1 Diabetes Mellitus: Is It Detrimental or Beneficial to Glycemic Control, Vascular Complications, and Quality of Life? Nutrients 2022; 15:nu15010199. [PMID: 36615856 PMCID: PMC9824312 DOI: 10.3390/nu15010199] [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: 12/12/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Celiac disease (CeD) is associated with type 1 diabetes mellitus (T1DM), and both have the same genetic background. Most patients with T1DM who develop CeD are either asymptomatic or have mild CeD-related gastrointestinal symptoms. Therefore, children affected by T1DM should undergo screening for asymptomatic CeD. The aim of this review is to highlight the influence of a gluten-free diet (GFD) on glycemic control, growth rate, microvascular complications, and quality of life in patients with T1DM and CeD. PubMed, Google Scholar, Web of Science, and Cochrane Central databases were searched. Reports reviewed were those published from 1969 to 2022 that focused on the interplay of T1DM and CeD and examined the effect of diet on glycemic control, growth rate, and quality of life. The most challenging aspect for a child with T1DM and CeD is that most GFD foods have a high glycemic index, while low glycemic index foods are recommended for T1DM. Interestingly, dietary therapy for CeD could improve the elevated HbA1c levels. Avoiding gluten added to a diabetic dietary regimen in T1DM patients might impose practical limitations and lead to important restrictions in the lifestyle of a young patient. Consequently, non-adherence to GFD in patients with T1DM and CeD is common. GFD in patients with T1DM and CeD seems to lower the incidence of micro- and macrovascular complications, but this requires further investigation. It seems that adherence to GFD in young patients with T1DM and CeD leads to regular growth and a stable body mass index without any negative effect on HbA1c or insulin requirements. Furthermore, the lipid profile and quality of life seem to have improved with the introduction of GFD.
Collapse
|
10
|
Szczuko M, Syrenicz A, Szymkowiak K, Przybylska A, Szczuko U, Pobłocki J, Kulpa D. Doubtful Justification of the Gluten-Free Diet in the Course of Hashimoto's Disease. Nutrients 2022; 14:nu14091727. [PMID: 35565695 PMCID: PMC9101474 DOI: 10.3390/nu14091727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
The popularization of the gluten-free diet brings with it a fashion for its use, which can harm the treatment of Hashimoto's disease. The few studies in this regard do not confirm positive changes resulting from a gluten-free diet. At the same time, the presence of other comorbid autoimmune diseases in this group of patients is increasing. This may have important implications for the interpretation of test results and the need for a gluten-free diet in some patients. In this review, the PubMed database was searched for links between a gluten-free diet, Hashimoto's disease, and autoimmune diseases. When analyzing the available literature, we found no basis for introducing a gluten-free diet for the standard management of Hashimoto patients. The recommended diet is instead an anti-inflammatory diet that levels the supply (to compensate for deficiencies) of vitamin D, iodine, and selenium, which are found in plant products rich in polyphenols, antioxidants, and omega-3 fatty acids, as illustrated in this article.
Collapse
Affiliation(s)
- Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (K.S.); (U.S.)
- Correspondence: ; Tel.: +48-91-441-4810
| | - Anhelli Syrenicz
- Department of Endocrinology, Metabolic Diseases and Internal Diseases, Pomeranian Medical University, 70-252 Szczecin, Poland; (A.S.); (J.P.)
| | - Katarzyna Szymkowiak
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (K.S.); (U.S.)
| | - Aleksandra Przybylska
- Department of Internal Medicine and Diabetology, Poznan University of Medical Sciences, 60-834 Poznań, Poland;
| | - Urszula Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (K.S.); (U.S.)
| | - Jakub Pobłocki
- Department of Endocrinology, Metabolic Diseases and Internal Diseases, Pomeranian Medical University, 70-252 Szczecin, Poland; (A.S.); (J.P.)
| | - Danuta Kulpa
- Department of Genetics, Plant Breeding and Biotechnology, Faculty of Environmental Management and Agriculture, West Pomeranian University of Technology, 71-434 Szczecin, Poland;
| |
Collapse
|
11
|
Kihl P, Krych L, Deng L, Hansen LH, Buschard K, Skov S, Nielsen DS, Kornerup Hansen A. Effect of gluten-free diet and antibiotics on murine gut microbiota and immune response to tetanus vaccination. PLoS One 2022; 17:e0266719. [PMID: 35417506 PMCID: PMC9007335 DOI: 10.1371/journal.pone.0266719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 03/25/2022] [Indexed: 12/25/2022] Open
Abstract
The purpose of this study was to compare the effect of a gluten-free diet and/or antibiotics on tetanus vaccine induced immunoglobulin G titers and immune cell levels in BALB/c mice. The gluten-free diet was associated with a reduced anti-tetanus IgG response, and it increased the relative abundance of the anti-inflammatory Bifidobacterium significantly in some of the mice. Antibiotics also led to gut microbiota changes and lower initial vaccine titer. After a second vaccination, neither gluten-free diet nor antibiotics reduced the titers. In the spleen, the gluten-free diet significantly increased regulatory T cell (Treg) fractions, CD4+ T cell activation, and tolerogenic dendritic cell fractions and activation, which extend the downregulating effect of the Treg. Therefore, the systemic effect of the gluten-free diet seems mainly tolerogenic. Antibiotics reduced the fractions of CD4+ T and B cells in the mesenteric lymph nodes. These results suggest that vaccine response in mice is under influence of their diet, the gut microbiota and the interplay between them. However, a gluten-free diet seems to work through mechanisms different from those induced by antibiotics. Therefore, diet should be considered when testing vaccines in mice and developing vaccines for humans.
Collapse
Affiliation(s)
- Pernille Kihl
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Ling Deng
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Lars H. Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Søren Skov
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Dennis S. Nielsen
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Axel Kornerup Hansen
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
12
|
Söderström H, Cervin M, Dereke J, Hillman M, Tiberg I, Norström F, Carlsson A. Does a gluten-free diet lead to better glycemic control in children with type 1 diabetes? Results from a feasibility study and recommendations for future trials. Contemp Clin Trials Commun 2022; 26:100893. [PMID: 35243123 PMCID: PMC8866053 DOI: 10.1016/j.conctc.2022.100893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 11/09/2021] [Accepted: 01/17/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Hanna Söderström
- Department of Clinical Sciences, Pediatrics, Skåne University Hospital Lund, Lund University, Lund, Sweden
- Corresponding author.
| | - Matti Cervin
- Department of Clinical Sciences, Child and Adolescent Psychiatry, Lund University, Lund, Sweden
| | - Jonatan Dereke
- Department of Clinical Sciences, Diabetes Research Laboratory, Lund University, Lund, Sweden
| | - Magnus Hillman
- Department of Health Sciences, Lund University, Lund, Sweden
| | - Iren Tiberg
- Department of Health Sciences, Lund University, Lund, Sweden
| | - Fredrik Norström
- Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden
| | - Annelie Carlsson
- Department of Clinical Sciences, Pediatrics, Skåne University Hospital Lund, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Comparative Study on the Exacerbating Effects of Casein-Rich vs. Gluten-Rich Diets on Biochemical-Induced Features in Rodent Model of Autism. J Mol Neurosci 2022; 72:359-371. [PMID: 35028884 DOI: 10.1007/s12031-021-01950-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 01/04/2023]
Abstract
In relation to dietary intervention in individuals with autism spectrum disorder (ASD), certain food constituents especially gluten and casein are recognized to be challenging and should be restricted. In this study, levels of glutathione S-transferase, glutathione, lipid peroxides, serotonin (5-HT), interleukin-6 (IL-6), glutamate, and gamma aminobutyric acid (GABA) were measured in the brain homogenates of ASD rodent model. Rats were treated either with single dose clindamycin (30 mg/kg) or with propionic acid (PPA) (250 mg/kg) for 3 days and then fed a standard diet, casein-rich diet (CRD), or gluten-rich diet (GRD). The obtained data demonstrates that clindamycin and PPA induced oxidative stress, which was slightly affected by CRD. A marked increase in the proinflammatory cytokine (IL-6) concentration found in clindamycin- and PPA-treated groups was lower in CRD fed rats. Both CRDs and GRDs produced similar trends in glutamate levels. 5-HT levels were higher in the clindamycin- and PPA-treated groups and increased with a GRD but were less affected by a CRD. CRD could be less deleterious compared to GRD. Although the underlying cause of gastrointestinal symptoms in patients with ASD is not exactly known, the most widely accepted one is the opioid theory which is related to GRD and CRD.
Collapse
|
14
|
Asri N, Nazemalhosseini Mojarad E, Mirjalali H, Mohebbi SR, Baghaei K, Rostami-Nejad M, Yadegar A, Rezaei-Tavirani M, Asadzadeh Aghdaei H, Rostami K, Masotti A. Toward finding the difference between untreated celiac disease and COVID-19 infected patients in terms of CD4, CD25 (IL-2 Rα), FOXP3 and IL-6 expressions as genes affecting immune homeostasis. BMC Gastroenterol 2021; 21:462. [PMID: 34895167 PMCID: PMC8665626 DOI: 10.1186/s12876-021-02056-1] [Citation(s) in RCA: 8] [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] [Received: 08/14/2021] [Accepted: 12/07/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is defined as an emerging infectious disease caused by severe acute respiratory syndrome coronavirus 2 and celiac disease (CD) is one of the autoimmune multiorgan diseases, which can be accompanied by an increased risk of viral infections. CD patients, especially untreated subjects, may be at greater risk of infections such as viral illnesses. Interleukin (IL)-6, CD4, CD25, and FOXP3 are known as genes affecting immune homeostasis and relate to the inflammation state. This study aimed to compare the expression levels of aforementioned genes in peripheral blood samples of CD and severe COVID-19 patients. METHODS Sixty newly diagnosed CD patients with median age (mean ± SD) of 35.40 ± 24.12 years; thirty confirmed severe COVID-19 patients with median age (mean ± SD) of 59.67 ± 17.22, and 60 healthy subjects with median age (mean ± SD) of 35.6 ± 13.02 years; were recruited from March to September 2020. Fresh whole blood samples were collected, total RNA was obtained and cDNA synthesis was carried out. RNA expression levels of IL-6, CD4, CD25, and FOXP3 genes were assessed using real-time quantitative RT-PCR according to the 2-∆∆Ct formula. Statistical analysis was performed using SPSS (V.21) and GraphPad, Prism (V.6). RESULTS While increased expression of CD4, CD25, and FOXP3 was observed in CD patients compared to the control group (p = 0.02, p = 0.03, and p < 0.0001 respectively) and COVID-19 patients group (p < 0.0001 for all of them), their expression levels in COVID-19 patients decreased compared to controls (p < 0.0001, p = 0.01, p = 0.007, respectively). Increased IL-6 expression was observed in both groups of patients compared to controls (p < 0.0001 for both of them). CONCLUSIONS Although untreated CD patients may be at greater risk of developing into severe COVID-19 if they are infected by SARS-CoV-2 virus (due to their high expression of IL-6), increased expression of anti-inflammatory markers in these patients may be beneficial for them with the ability of reducing the severity of COVID-19 disease, which needs to be proven in future studies involving celiac patients infected with COVID-19.
Collapse
Affiliation(s)
- Nastaran Asri
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Nazemalhosseini Mojarad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Mohebbi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rostami
- Department of Gastroenterology, MidCentral DHB, Palmerston North, New Zealand
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| |
Collapse
|
15
|
Gupta KB, Mantha AK, Dhiman M. Mitigation of Gliadin-Induced Inflammation and Cellular Damage by Curcumin in Human Intestinal Cell Lines. Inflammation 2021; 44:873-889. [PMID: 33394186 DOI: 10.1007/s10753-020-01383-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/08/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022]
Abstract
Wheat is a major diet from many years; apart from its nutritious value, the wheat protein gliadin is responsible for many inflammatory diseases like celiac disease (CD), and non-celiac gluten sensitivity (NCGS). In this study, the gliadin-induced inflammation and associated cellular damage along with the protective role of curcumin was evaluated using human intestinal cell lines (HCT-116 and HT-29) as a model. Cells were cultured and exposed to 160 μg/ml of gliadin, 100 μM H2O2, and 10 μM curcumin (3 h pretreatment) followed by the assessment of inflammation. Spectrophotometric methods, real-time-PCR, ELISA, Western blotting, and confocal microscopy techniques were used to assess inflammatory markers such as advanced oxidation protein products (AOPPs) level, activity of myeloperoxidase (MPO) and NADPH oxidase (NOX), cytokines, and cell damage markers. The results show that gliadin increases the AOPPs level and the activity of MPO and NOX expression. It enhances inflammation by increasing expression of pro-inflammatory cytokines, altered expression of anti-inflammatory, and regulatory cytokines. It exacerbates the cellular damage by increasing MMP-2 and 9 and decreasing integrin α and β expression. Gliadin promotes disease pathogenesis by inducing the inflammation and cellular damage which further alter the cellular homeostasis. The pretreatment of curcumin counteracts the adverse effect of gliadin and protect the cells via diminishing the inflammation and help the cell to regain the cellular morphology suggesting phytochemical-based remedial interventions against wheat allergies.
Collapse
Affiliation(s)
- Kunj Bihari Gupta
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Anil K Mantha
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
16
|
Haupt-Jorgensen M, Buschard K. Can a gluten-free diet be partly protective for COVID-19 infection? APMIS 2020; 128:558-559. [PMID: 32854147 PMCID: PMC7461366 DOI: 10.1111/apm.13075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/18/2020] [Indexed: 01/28/2023]
Affiliation(s)
| | - Karsten Buschard
- The Bartholin Institute, Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
17
|
Liu B, Che Y, Zhang M, Ren W, Xia X, Liu H, Huang T, Huang J, Lei L. IFN-γ Activates the TLR4-CCL5 Signaling Through Reducing Arginine Level, Leading to Enhanced Susceptibility of Bovine Mammary Epithelial Cells to Staphylococcus aureus. Inflammation 2020; 43:2209-2221. [PMID: 32725514 DOI: 10.1007/s10753-020-01288-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dairy cow mastitis is a common bacterial infectious disease which seriously threatens the development of the dairy cow industry. Previous studies have found that increased IFN-γ expression in dairy cows makes dairy cows more susceptible to mastitis, but the underlying mechanism is still not known. In this study, we utilized the in vitro bovine mammary epithelial cells (BMECs) model to explore the molecular mechanism via transcriptome sequencing technology, immunofluorescence, and Western blotting. It was found that IFN-γ promoted the adhesion and invasion of Staphylococcus aureus to BMECs through increasing the expression of TLR4-mediated CCL5 in BMECs. IFN-γ increased the activity of arginase II and reduced the level of arginine in cells, while the addition of arginine inhibited the expression of TLR4 and CCL5. An invasion experiment in mice further validated that IFN-γ treatment significantly increased the bacterial load in mammary glands and blood. However, the colonization and diffusion of S. aureus were interestingly decreased after Arg supplement. These data reveal that increased IFN-γ reduces arginine levels and activates TLR4-CCL5 signaling, leading to enhanced susceptibility of BMECs to S. aureus. Our findings are helpful to understand the pathogenesis of dairy cow mastitis and provide a theoretical basis for improvement of mastitis resistance in dairy cows.
Collapse
Affiliation(s)
- Baijun Liu
- College of Veterinary Medicine, Jilin University, Changchun, Xi'an Road 5333, China
| | - Yanyi Che
- College of Veterinary Medicine, Jilin University, Changchun, Xi'an Road 5333, China
| | - Meina Zhang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Wenbo Ren
- The First Hospital, Jilin University, Xinmin street 71, Changchun, China
| | - Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hongtao Liu
- College of Veterinary Medicine, Jilin University, Changchun, Xi'an Road 5333, China
| | - Tinghao Huang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Jing Huang
- The First Hospital, Jilin University, Xinmin street 71, Changchun, China.
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun, Xi'an Road 5333, China. .,College of Animal Sciences, Yangtze University, Jingzhou, China.
| |
Collapse
|
18
|
Neuman V, Pruhova S, Kulich M, Kolouskova S, Vosahlo J, Romanova M, Petruzelkova L, Obermannova B, Funda DP, Cinek O, Sumnik Z. Gluten-free diet in children with recent-onset type 1 diabetes: A 12-month intervention trial. Diabetes Obes Metab 2020; 22:866-872. [PMID: 31984648 DOI: 10.1111/dom.13974] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/17/2022]
Abstract
AIM To test whether a gluten-free diet (GFD) is associated with the deceleration of the decline in beta-cell capacity in non-coeliac children with recently diagnosed type 1 diabetes. METHODS Forty-five children (aged 10.2 ± 3.3 years) were recruited into a self-selected intervention trial: 26 started with a GFD within a median of 38 days postonset, whereas 19 remained on a standard diet. The main outcomes were the decline in C-peptide area under the curve (AUC) in mixed-meal tolerance tests (MMTTs) at 6 and 12 months relative to 1 month after diabetes onset and the difference in insulin dose, insulin dose-adjusted A1c (IDAA1c) and HbA1c assessed every 3 months. The adherence to the GFD was verified by immunoreactive gluten in the stool and by food questionnaires at every visit. Quality of life (QoL) questionnaires were administered to the participants at the end of the intervention at 12 months. The data were analysed as per protocol (in 39 subjects who duly completed the whole follow-up: 20 in the GFD group, 19 in the control group) by linear and longitudinal regression models adjusted for sex, age and baseline variables. RESULTS At 12 months, the difference in C-peptide AUC between subjects in the GFD group and controls was 205 pmol/L (95% CI -223 to 633; P = 0.34) in a model adjusted for age, sex and body weight, and for baseline insulin dose, MMTT C-peptide AUC and HbA1c assessed at 1 month after diagnosis. In a longitudinal analysis of all three time points adjusted for age, sex and body weight, C-peptide declined more slowly in the GFD group than in controls, with the difference in trends being 409 pmol/L/year (P = 0.04). The GFD group had a marginally lower insulin dose (by 0.15 U/kg/day; P = 0.07), a lower IDAA1c (by 1.37; P = 0.01) and a lower mean HbA1c (by 0.7% [7.8 mmol/mol]; P = 0.02) than those of the controls at 12 months. There was no appreciable difference between the groups in daily carbohydrate intake (P = 0.49) or in the QoL reported by the patients (P = 0.70) and their parents/caregivers (P = 0.59). CONCLUSIONS A GFD maintained over the first year after type 1 diabetes diagnosis was associated with better HbA1c and a prolonged partial remission period. There was a hint of slower C-peptide decline but the association was not strong enough to make definite conclusions.
Collapse
Affiliation(s)
- Vit Neuman
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Stepanka Pruhova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Michal Kulich
- Department of Probability and Mathematical Statistics, Faculty of Mathematics and Physics, Charles University, Prague, Czech Republic
| | - Stanislava Kolouskova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jan Vosahlo
- Department of Pediatrics, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Martina Romanova
- Department of Pediatrics, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Lenka Petruzelkova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Barbora Obermannova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - David P Funda
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Cinek
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Zdenek Sumnik
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
19
|
Pei J, Wei S, Pei Y, Wu H, Wang D. Role of Dietary Gluten in Development of Celiac Disease and Type I Diabetes: Management Beyond Gluten-Free Diet. Curr Med Chem 2019; 27:3555-3576. [PMID: 30963964 DOI: 10.2174/0929867326666190409120716] [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: 09/03/2018] [Revised: 03/25/2019] [Accepted: 04/03/2019] [Indexed: 11/22/2022]
Abstract
Gluten triggers Celiac Disease (CD) and type I diabetes in genetically predisposed population of human leukocyte antigen DQ2/DQ8+ and associates with disorders such as schizophrenia and autism. Application of a strict gluten-free diet is the only well-established treatment for patients with CD, whereas the treatment for patients with celiac type I diabetes may be depend on the timing and frequency of the diet. The application of a gluten-free diet in patients with CD may contribute to the development of metabolic syndrome and nonalcoholic fatty liver disease and may also lead to a high glycemic index, low fiber diet and micronutrient deficiencies. The alteration of copper bioavailability (deficient, excess or aberrant coordination) may contribute to the onset and progress of related pathologies. Therefore, nutrient intake of patients on a gluten-free diet should be the focus of future researches. Other gluten-based therapies have been rising with interest such as enzymatic pretreatment of gluten, oral enzyme supplements to digest dietary gluten, gluten removal by breeding wheat varieties with reduced or deleted gluten toxicity, the development of polymeric binders to suppress gluten induced pathology.
Collapse
Affiliation(s)
- Jinli Pei
- Hainan Province Key Laboratory for Sustainable Utilization of Tropical Bioresources, Hainan University, Hainan, 570228, China.,Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Hainan 570228, China
| | - Shuangshuang Wei
- Hainan Province Key Laboratory for Sustainable Utilization of Tropical Bioresources, Hainan University, Hainan, 570228, China.,Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Hainan 570228, China
| | - Yechun Pei
- Hainan Province Key Laboratory for Sustainable Utilization of Tropical Bioresources, Hainan University, Hainan, 570228, China.,Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Hainan 570228, China
| | - Hao Wu
- Hainan Province Key Laboratory for Sustainable Utilization of Tropical Bioresources, Hainan University, Hainan, 570228, China.,Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Hainan 570228, China
| | - Dayong Wang
- Hainan Province Key Laboratory for Sustainable Utilization of Tropical Bioresources, Hainan University, Hainan, 570228, China.,Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Hainan 570228, China
| |
Collapse
|
20
|
Possible Prevention of Diabetes with a Gluten-Free Diet. Nutrients 2018; 10:nu10111746. [PMID: 30428550 PMCID: PMC6266002 DOI: 10.3390/nu10111746] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Gluten seems a potentially important determinant in type 1 diabetes (T1D) and type 2 diabetes (T2D). Intake of gluten, a major component of wheat, rye, and barley, affects the microbiota and increases the intestinal permeability. Moreover, studies have demonstrated that gluten peptides, after crossing the intestinal barrier, lead to a more inflammatory milieu. Gluten peptides enter the pancreas where they affect the morphology and might induce beta-cell stress by enhancing glucose- and palmitate-stimulated insulin secretion. Interestingly, animal studies and a human study have demonstrated that a gluten-free (GF) diet during pregnancy reduces the risk of T1D. Evidence regarding the role of a GF diet in T2D is less clear. Some studies have linked intake of a GF diet to reduced obesity and T2D and suggested a role in reducing leptin- and insulin-resistance and increasing beta-cell volume. The current knowledge indicates that gluten, among many environmental factors, may be an aetiopathogenic factors for development of T1D and T2D. However, human intervention trials are needed to confirm this and the proposed mechanisms.
Collapse
|
21
|
Antvorskov JC, Halldorsson TI, Josefsen K, Svensson J, Granström C, Roep BO, Olesen TH, Hrolfsdottir L, Buschard K, Olsen SF. Association between maternal gluten intake and type 1 diabetes in offspring: national prospective cohort study in Denmark. BMJ 2018; 362:k3547. [PMID: 30232082 PMCID: PMC6283375 DOI: 10.1136/bmj.k3547] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To examine the association between prenatal gluten exposure and offspring risk of type 1 diabetes in humans. DESIGN National prospective cohort study. SETTING National health information registries in Denmark. PARTICIPANTS Pregnant Danish women enrolled into the Danish National Birth Cohort, between January 1996 and October 2002, MAIN OUTCOME MEASURES: Maternal gluten intake, based on maternal consumption of gluten containing foods, was reported in a 360 item food frequency questionnaire at week 25 of pregnancy. Information on type 1 diabetes occurrence in the participants' children, from 1 January 1996 to 31 May 2016, were obtained through registry linkage to the Danish Registry of Childhood and Adolescent Diabetes. RESULTS The study comprised 101 042 pregnancies in 91 745 women, of whom 70 188 filled out the food frequency questionnaire. After correcting for multiple pregnancies, pregnancies ending in abortions, stillbirths, lack of information regarding the pregnancy, and pregnancies with implausibly high or low energy intake, 67 565 pregnancies (63 529 women) were included. The average gluten intake was 13.0 g/day, ranging from less than 7 g/day to more than 20 g/day. The incidence of type 1 diabetes among children in the cohort was 0.37% (n=247) with a mean follow-up period of 15.6 years (standard deviation 1.4). Risk of type 1 diabetes in offspring increased proportionally with maternal gluten intake during pregnancy (adjusted hazard ratio 1.31 (95% confidence interval 1.001 to 1.72) per 10 g/day increase of gluten). Women with the highest gluten intake versus those with the lowest gluten intake (≥20 v <7 g/day) had double the risk of type 1 diabetes development in their offspring (adjusted hazard ratio 2.00 (95% confidence interval 1.02 to 4.00)). CONCLUSIONS High gluten intake by mothers during pregnancy could increase the risk of their children developing type 1 diabetes. However, confirmation of these findings are warranted, preferably in an intervention setting.
Collapse
Affiliation(s)
- Julie C Antvorskov
- Bartholin Institute, Rigshospitalet, Ole Måløes Vej 5, 2200 Copenhagen K, Denmark
| | - Thorhallur I Halldorsson
- Centre for Foetal Programming, Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark
- Unit for Nutrition Research, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Food Science and Nutrition, University of Iceland, Reykjavik, Iceland
| | - Knud Josefsen
- Bartholin Institute, Rigshospitalet, Ole Måløes Vej 5, 2200 Copenhagen K, Denmark
| | - Jannet Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Charlotta Granström
- Centre for Foetal Programming, Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark
| | - Bart O Roep
- Department of Diabetes Immunology, Diabetes and Metabolism Research Institute at the Beckman Diabetes Research Institute, City of Hope, Duarte, CA, USA
- Departments of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands
| | - Trine H Olesen
- Centre for Foetal Programming, Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark
| | - Laufey Hrolfsdottir
- Department of Education, Science, and Quality, Akureyri Hospital, Akureyri, Iceland
| | - Karsten Buschard
- Bartholin Institute, Rigshospitalet, Ole Måløes Vej 5, 2200 Copenhagen K, Denmark
| | - Sjudur F Olsen
- Centre for Foetal Programming, Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
22
|
Kihl P, Krych L, Buschard K, Wesley JD, Kot W, Hansen AK, Nielsen DS, von Herrath MG. Oral insulin does not alter gut microbiota composition of NOD mice. Diabetes Metab Res Rev 2018; 34:e3010. [PMID: 29637693 DOI: 10.1002/dmrr.3010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/08/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Oral insulin as a preventive strategy and/or treatment of type 1 diabetes has been the target of much research. Producing oral insulins is a complex and challenging task, with numerous pitfalls, due to physiological, physical, and biochemical barriers. Our aim was to determine the impact of oral insulin on the delicate gut microbiota composition. METHODS Female nonobese diabetic mice were given oral porcine insulin 2 times a week from 5 weeks of age for 4 weeks, and then subsequently once a week for 21 weeks, or until euthanized. The mice were divided into groups on a gluten-reduced diet or a standard diet. Gut microbiota composition was analysed based on faecal samples, and the type 1 diabetes incidence of the mice was monitored. RESULTS We observed no influence of the oral porcine insulin on the gut microbiota composition of mice on a gluten-reduced or a standard diet at 9 weeks of age. Also, the administration of oral insulin did not influence the incidence of type 1 diabetes at 30 weeks of age. CONCLUSIONS Oral porcine insulin does not alter the gut microbiota composition of nonobese diabetic mice on either a gluten-reduced diet or standard diet. Also, the oral porcine insulin did not influence the incidence of type 1 diabetes in the groups.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/microbiology
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/microbiology
- Diabetes Mellitus, Type 1/pathology
- Dysbiosis/immunology
- Dysbiosis/pathology
- Feces/microbiology
- Female
- Gastrointestinal Microbiome/drug effects
- Insulin, Regular, Pork/administration & dosage
- Insulin, Regular, Pork/adverse effects
- Mice
- Mice, Inbred NOD
- Swine
Collapse
Affiliation(s)
- Pernille Kihl
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Johnna D Wesley
- Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Witold Kot
- Department of Environmental Science, Aarhus University, Aarhus, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dennis S Nielsen
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
23
|
Haupt-Jorgensen M, Larsen J, Josefsen K, Jørgensen TZ, Antvorskov JC, Hansen AK, Buschard K. Gluten-free diet during pregnancy alleviates signs of diabetes and celiac disease in NOD mouse offspring. Diabetes Metab Res Rev 2018; 34:e2987. [PMID: 29392873 DOI: 10.1002/dmrr.2987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/08/2018] [Accepted: 01/22/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Gluten-free (GF) diet during pregnancy ameliorates autoimmune diabetes in nonobese diabetic (NOD) mouse offspring. Due to comorbidity of celiac disease in type 1 diabetes, we hypothesized that GF diet in utero alleviates the humoral and histopathological signs of celiac disease in NOD mice. We aimed to establish the mechanisms behind the diabetes-protective effect of GF diet in utero. METHODS Breeding pairs of NOD mice were fed a GF or gluten-containing standard (STD) diet until parturition. The offspring were nursed by mothers on STD diet and continued on this diet until ages 4 and 13 weeks. Analyses of serum antitissue transglutaminase (anti-tTG) intestine and islet histology, islet transglutaminase (TG) activity, and cytokine expression in T cells from lymphoid organs were performed. RESULTS GF versus STD diet in utero led to reduced serum anti-tTG titre and increased villus-to-crypt ratio at both ages. Insulitis along with systemic and local inflammation were decreased, but islet TG activity was unchanged in 13-week-old GF mice. These mice had unchanged beta-cell volumes, but increased islet numbers throughout the prediabetic period. CONCLUSIONS Collectively, GF diet administered during pregnancy improves signs of celiac disease and autoimmune diabetes in the offspring. The diabetes-ameliorative effect of GF diet in utero is followed by dampening of inflammation, unchanged beta-cell volume, but increased islet numbers.
Collapse
Affiliation(s)
| | - Jesper Larsen
- The Bartholin Institute, Rigshospitalet, Copenhagen, Denmark
| | - Knud Josefsen
- The Bartholin Institute, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Axel K Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | |
Collapse
|
24
|
Lerner A, Shoenfeld Y, Matthias T. Adverse effects of gluten ingestion and advantages of gluten withdrawal in nonceliac autoimmune disease. Nutr Rev 2017; 75:1046-1058. [PMID: 29202198 DOI: 10.1093/nutrit/nux054] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
In light of the coincident surge in overall gluten intake and the incidence of autoimmune diseases, the possible biological adverse effects of gluten were explored. PubMed, MEDLINE, and the Cochrane Library databases were screened for reports published between 1964 and 2016 regarding the adverse effects of gluten as well as the effects of a gluten-free diet on autoimmune diseases. In vitro and in vivo studies describing gluten intake in animal models or cell lines and gluten-free diets in human autoimmune diseases were reviewed. Multiple detrimental aspects of gluten affect human health, including gluten-dependent digestive and extradigestive manifestations mediated by potentially immunological or toxic reactions that induce gastrointestinal inadequacy. Gluten affects the microbiome and increases intestinal permeability. It boosts oxidative stress and affects epigenetic behavior. It is also immunogenic, cytotoxic, and proinflammatory. Gluten intake increases apoptosis and decreases cell viability and differentiation. In certain nonceliac autoimmune diseases, gluten-free diets may help curtail the adverse effects of gluten. Additional in vivo studies are needed to unravel the puzzle of gluten effects in humans and to explore the potential beneficial effects of gluten-free diets in autoimmune diseases.
Collapse
Affiliation(s)
- Aaron Lerner
- B. Rappaport School of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- AESKU.KIPP Institute, Wendelsheim, Germany
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center-Tel Hashomer, Ramat Gan, Israel, and the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | |
Collapse
|
25
|
Gianchecchi E, Fierabracci A. On the pathogenesis of insulin-dependent diabetes mellitus: the role of microbiota. Immunol Res 2017; 65:242-256. [PMID: 27421719 DOI: 10.1007/s12026-016-8832-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder characterized by the selective destruction of insulin-producing β cells as result of a complex interplay between genetic, stochastic and environmental factors in genetically susceptible individuals. An increasing amount of experimental data from animal models and humans has supported the role played by imbalanced gut microbiome in T1D pathogenesis. The commensal intestinal microbiota is fundamental for several physiologic mechanisms, including the establishment of immune homeostasis. Alterations in its composition have been correlated to changes in the gut immune system, including defective tolerance to food antigens, intestinal inflammation and enhanced gut permeability. Early findings reported differences in the intestinal microbiome of subjects affected by prediabetes or overt disease compared to healthy individuals. The present review focuses on microbiota-host homeostasis, its alterations, factors that influence microbiome composition and discusses their putative correlation with T1D development. Further studies are necessary to clarify the role played by microbiota modifications in the processes that cause enhanced permeability and the autoimmune mechanisms responsible for T1D onset.
Collapse
Affiliation(s)
- Elena Gianchecchi
- Vismederi Srl, Siena, Italy
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale S. Paolo 15, 00146, Rome, Italy
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale S. Paolo 15, 00146, Rome, Italy.
| |
Collapse
|
26
|
Svensson J, Sildorf SM, Pipper CB, Kyvsgaard JN, Bøjstrup J, Pociot FM, Mortensen HB, Buschard K. Potential beneficial effects of a gluten-free diet in newly diagnosed children with type 1 diabetes: a pilot study. SPRINGERPLUS 2016; 5:994. [PMID: 27398272 PMCID: PMC4936999 DOI: 10.1186/s40064-016-2641-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/21/2016] [Indexed: 12/22/2022]
Abstract
AIM Gluten-free diet has shown promising effects in preventing type 1 diabetes (T1D) in animals as well as beneficial effects on the immune system. Gluten-free diet at diabetes onset may alter the natural course and outcome of autoimmune diseases such as T1D. METHODS In a 12-month study, 15 children newly diagnosed with T1D were instructed to follow a gluten-free diet. Questionnaires were used to evaluate adherence to the gluten-free diet. Partial remission (PR) was defined by insulin dose-adjusted A1c (IDAA1c) ≤9 or stimulated C-peptide (SCP) >300 pmol/L measured 90 min after a liquid mixed meal at the inclusion, six and 12 months after onset. The intervention group was compared with two previous cohorts. Linear mixed models were used to estimate differences between cohorts. RESULTS After 6 months, more children on a gluten-free diet tended to have SCP values above 300 pmol/L compared to the European cohort (p = 0.08). The adherence to a gluten-free diet decreased during the 12-month study period. After 1 year there was no difference in SCP levels or percentage in remission according to SCP (p > 0.1). Three times as many children were still in PR based on IDAA1c (p < 0.05). Twelve months after onset HbA1c were 21 % lower and IDAA1c >1 unit lower in the cohort on a gluten-free diet compared to the two previous cohorts (p < 0.001). CONCLUSION Gluten-free diet is feasible in highly motivated families and is associated with a significantly better outcome as assessed by HbA1c and IDAA1c. This finding needs confirmation in a randomized trial including screening for quality of life. (Clinicaltrials.gov number NCT02284815).
Collapse
Affiliation(s)
- Jannet Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Stine Møller Sildorf
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Christian B Pipper
- Section of Biostatistics, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade, 1014 Copenhagen, Denmark
| | - Julie N Kyvsgaard
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Julie Bøjstrup
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Flemming M Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Henrik B Mortensen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Children and Adolescents, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Karsten Buschard
- The Bartholin Institute, Rigshospitalet, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
27
|
Xia XJ, Gao YY, Zhang J, Wang L, Zhao S, Che YY, Ao CJ, Yang HJ, Wang JQ, Lei LC. Autophagy mediated by arginine depletion activation of the nutrient sensor GCN2 contributes to interferon-γ-induced malignant transformation of primary bovine mammary epithelial cells. Cell Death Discov 2016; 2:15065. [PMID: 27551491 PMCID: PMC4979444 DOI: 10.1038/cddiscovery.2015.65] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/19/2015] [Accepted: 11/22/2015] [Indexed: 12/26/2022] Open
Abstract
Autophagy has been linked to the regulation of both the prevention and progression of cancer. IFN-γ has been shown to induce autophagy in multiple cell lines in vitro. However, whether IFN-γ can induce autophagy and whether autophagy promotes malignant transformation in healthy lactating bovine mammary epithelial cells (BMECs) remain unclear. Here, we provide the first evidence of the correlation between IFN-γ treatment, autophagy and malignant transformation and of the mechanism underlying IFN-γ-induced autophagy and subsequent malignant transformation in primary BMECs. IFN-γ levels were significantly increased in cattle that received normal long-term dietary corn straw (CS) roughage supplementation. In addition, an increase in autophagy was clearly observed in the BMECs from the mammary tissue of cows expressing high levels of IFN-γ. In vitro, autophagy was clearly induced in primary BMECs by IFN-γ within 24 h. This induced autophagy could subsequently promote dramatic primary BMEC transformation. Furthermore, we found that IFN-γ promoted arginine depletion, activated the general control nonderepressible-2 kinase (GCN2) signalling pathway and resulted in an increase in autophagic flux and the amount of autophagy in BMECs. Overall, our findings are the first to demonstrate that arginine depletion and kinase GCN2 expression mediate IFN-γ-induced autophagy that may promote malignant progression and that immunometabolism, autophagy and cancer are strongly correlated. These results suggest new directions and paths for preventing and treating breast cancer in relation to diet.
Collapse
Affiliation(s)
- X-J Xia
- College of Veterinary Medicine, Jilin University , Changchun, PR China
| | - Y-Y Gao
- College of Animal Science, Jilin University , Changchun, PR China
| | - J Zhang
- Changchun University of Chinese Medicine , Changchun, PR China
| | - L Wang
- College of Animal Science, Henan Institute of Science and Technology , Xinxiang, PR China
| | - S Zhao
- College of Animal Science, Jilin University , Changchun, PR China
| | - Y-Y Che
- College of Veterinary Medicine, Jilin University , Changchun, PR China
| | - C-J Ao
- College of Animal Science, Inner Mongolian Agricultural University , Hohhot, PR China
| | - H-J Yang
- College of Animal Science and Technology, China Agricultural University , Beijing, PR China
| | - J-Q Wang
- Institute of Animal Science, Chinese Academy of Agricultural Science , Beijing, PR China
| | - L-C Lei
- College of Veterinary Medicine, Jilin University , Changchun, PR China
| |
Collapse
|
28
|
Bruun SW, Josefsen K, Tanassi JT, Marek A, Pedersen MHF, Sidenius U, Haupt-Jorgensen M, Antvorskov JC, Larsen J, Heegaard NH, Buschard K. Large Gliadin Peptides Detected in the Pancreas of NOD and Healthy Mice following Oral Administration. J Diabetes Res 2016; 2016:2424306. [PMID: 27795959 PMCID: PMC5067331 DOI: 10.1155/2016/2424306] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022] Open
Abstract
Gluten promotes type 1 diabetes in nonobese diabetic (NOD) mice and likely also in humans. In NOD mice and in non-diabetes-prone mice, it induces inflammation in the pancreatic lymph nodes, suggesting that gluten can initiate inflammation locally. Further, gliadin fragments stimulate insulin secretion from beta cells directly. We hypothesized that gluten fragments may cross the intestinal barrier to be distributed to organs other than the gut. If present in pancreas, gliadin could interact directly with the immune system and the beta cells to initiate diabetes development. We orally and intravenously administered 33-mer and 19-mer gliadin peptide to NOD, BALB/c, and C57BL/6 mice and found that the peptides readily crossed the intestinal barrier in all strains. Several degradation products were found in the pancreas by mass spectroscopy. Notably, the exocrine pancreas incorporated large amounts of radioactive label shortly after administration of the peptides. The study demonstrates that, even in normal animals, large gliadin fragments can reach the pancreas. If applicable to humans, the increased gut permeability in prediabetes and type 1 diabetes patients could expose beta cells directly to gliadin fragments. Here they could initiate inflammation and induce beta cell stress and thus contribute to the development of type 1 diabetes.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Chromatography, Liquid
- Diabetes Mellitus, Type 1/immunology
- Electrophoresis, Polyacrylamide Gel
- Gliadin/immunology
- Gliadin/pharmacokinetics
- Inflammation
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Intestinal Mucosa/metabolism
- Male
- Mass Spectrometry
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Pancreas/metabolism
- Pancreas, Exocrine/metabolism
- Peptide Fragments/pharmacokinetics
- Permeability
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
| | - Knud Josefsen
- The Bartholin Institute, Rigshospitalet, Copenhagen N, Denmark
- *Knud Josefsen:
| | - Julia T. Tanassi
- Clinical Biochemistry, Immunology & Genetics, Statens Serum Institut, Copenhagen S, Denmark
| | - Aleš Marek
- The Hevesy Laboratory, DTU Nutech, Technical University of Denmark, Roskilde, Denmark
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague 6, Czech Republic
| | - Martin H. F. Pedersen
- The Hevesy Laboratory, DTU Nutech, Technical University of Denmark, Roskilde, Denmark
| | - Ulrik Sidenius
- Enzyme Purification and Characterization, Novozymes A/S, Bagsværd, Denmark
| | | | | | - Jesper Larsen
- The Bartholin Institute, Rigshospitalet, Copenhagen N, Denmark
| | - Niels H. Heegaard
- Clinical Biochemistry, Immunology & Genetics, Statens Serum Institut, Copenhagen S, Denmark
| | | |
Collapse
|
29
|
Antvorskov JC, Josefsen K, Haupt-Jorgensen M, Fundova P, Funda DP, Buschard K. Gluten-Free Diet Only during Pregnancy Efficiently Prevents Diabetes in NOD Mouse Offspring. J Diabetes Res 2016; 2016:3047574. [PMID: 27642610 PMCID: PMC5014974 DOI: 10.1155/2016/3047574] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/05/2016] [Accepted: 07/10/2016] [Indexed: 11/17/2022] Open
Abstract
Studies have documented that the pathogenesis of autoimmune diabetes is influenced by the intake of gluten. Aims. To investigate the importance of gluten exposure during pregnancy and the subsequent development of autoimmune diabetes in offspring. Methods. Nonobese diabetic mice were divided into 7 groups to receive combinations of gluten-free and standard diet before, during, or after pregnancy. Diabetes incidence in offspring was followed in each group (n = 16-27) for 310 days. Insulitis score and intestinal expression of T-cell transcription factors (RT-QPCR) were evaluated in animals from the different diet groups. Results. If mothers were fed a gluten-free diet only during pregnancy, the development of autoimmune diabetes in offspring was almost completely prevented with an incidence reduction from 62.5% in gluten-consuming mice to 8.3% (p < 0.0001) in the gluten-free group. The islets of Langerhans were less infiltrated (p < 0.001) and the intestinal expression of RORγt (Th17) (p < 0.0001) reduced in mice whose mothers were Gluten-free during pregnancy. Conclusion. A gluten-free diet exclusively during pregnancy efficiently prevents autoimmune diabetes development in offspring and reduces insulitis and intestinal expression of RORγt (Th17).
Collapse
Affiliation(s)
- Julie C. Antvorskov
- The Bartholin Institute, Rigshospitalet, 2200 Copenhagen, Denmark
- *Julie C. Antvorskov:
| | - Knud Josefsen
- The Bartholin Institute, Rigshospitalet, 2200 Copenhagen, Denmark
| | | | - Petra Fundova
- The Bartholin Institute, Rigshospitalet, 2200 Copenhagen, Denmark
- Laboratory of Specific Cellular Immunity, Institute of Microbiology ASCR, 54922 Prague, Czech Republic
| | - David P. Funda
- The Bartholin Institute, Rigshospitalet, 2200 Copenhagen, Denmark
- Laboratory of Specific Cellular Immunity, Institute of Microbiology ASCR, 54922 Prague, Czech Republic
| | - Karsten Buschard
- The Bartholin Institute, Rigshospitalet, 2200 Copenhagen, Denmark
| |
Collapse
|
30
|
Han YY, Forno E, Brehm JM, Acosta-Pérez E, Alvarez M, Colón-Semidey A, Rivera-Soto W, Campos H, Litonjua AA, Alcorn JF, Canino G, Celedón JC. Diet, interleukin-17, and childhood asthma in Puerto Ricans. Ann Allergy Asthma Immunol 2015; 115:288-293.e1. [PMID: 26319606 PMCID: PMC4721241 DOI: 10.1016/j.anai.2015.07.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/03/2015] [Accepted: 07/08/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Dietary patterns might influence the pathogenesis of asthma in Puerto Ricans, the ethnic group most affected by this disease in the United States. OBJECTIVE To examine the association among diet, T-helper cell type 17 cytokines, and asthma in Puerto Rican children. METHODS As part of a case-control study of 678 Puerto Rican children 6 to 14 years old in San Juan, participants completed a 75-item questionnaire on the child's food consumption in the prior week. Foods were aggregated into 7 groups: fruits, vegetables, grains, protein foods, dairy, fats, and sweets. Logistic regression was used to evaluate consumption frequency of each group, plasma T-helper cell type 17 cytokine levels, and asthma. Based on this analysis, a food score (range -2 [unhealthy diet: high consumption of dairy products and sweets, low consumption of vegetables and grains] to +2 [healthy diet: high consumption of grains and vegetables, low consumption of dairy and sweets]) was created to identify dietary patterns. RESULTS High consumption of grains was associated with lower odds of asthma (adjusted odds ratio [aOR] 0.52, 95% confidence interval [CI] 0.33-0.82), whereas frequent consumption of dairy products (aOR 1.93, 95% CI 1.32-2.84) or sweets (aOR 1.82, 95% CI 1.08-2.72) was associated with higher odds of asthma. A healthier diet (each 1-point increment in food score) was associated with lower levels of interleukin-17F (β = -1.48 pg/mL, 95% CI -1.78 to -1.20) and with 36% decreased odds of asthma (aOR 0.64, 95% CI 0.53-0.77). CONCLUSION A healthy diet, with frequent consumption of vegetables and grains and low consumption of dairy products and sweets, was associated with lower levels of interleulin-17F and decreased odds of childhood asthma in Puerto Ricans.
Collapse
Affiliation(s)
- Yueh-Ying Han
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA
| | - John M. Brehm
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA
| | - Edna Acosta-Pérez
- Behavioral Sciences Research Institute and Department of Pediatrics, University of Puerto Rico, San Juan, PR
| | - María Alvarez
- Behavioral Sciences Research Institute and Department of Pediatrics, University of Puerto Rico, San Juan, PR
| | - Angel Colón-Semidey
- Behavioral Sciences Research Institute and Department of Pediatrics, University of Puerto Rico, San Juan, PR
| | | | | | - Augusto A. Litonjua
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - John F. Alcorn
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA
| | - Glorisa Canino
- Behavioral Sciences Research Institute and Department of Pediatrics, University of Puerto Rico, San Juan, PR
| | - Juan C. Celedón
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
31
|
Larsen J, Weile C, Antvorskov JC, Engkilde K, Nielsen SMB, Josefsen K, Buschard K. Effect of dietary gluten on dendritic cells and innate immune subsets in BALB/c and NOD mice. PLoS One 2015; 10:e0118618. [PMID: 25738288 PMCID: PMC4349814 DOI: 10.1371/journal.pone.0118618] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/21/2015] [Indexed: 12/13/2022] Open
Abstract
The innate immune system is known to play an important role in oral tolerance to dietary antigens. This is important in development of celiac disease (CD) but may also be important in type 1 diabetes (T1D), and could potentially explain the reduced incidence of T1D in mice receiving a gluten-free (GF) diet. The direct in vivo effect of gluten on innate cells, and particularly dendritic cells (DC) is not sufficiently clarified. Therefore, we wished to investigate the innate cell populations of spontaneous diabetic NOD mice and healthy BALB/c mice kept on a GF or a standard (STD) gluten containing diet. We studied, by flow cytometry and reverse transcription-quantitative polymerase chain reaction (qRT-PCR), if dietary gluten induces changes in the activation of DCs and distribution of selected innate cells in lymphoid, pancreatic and intestinal tissues in BALB/c and NOD mice. We found that a GF diet increased the percentage of macrophages in BALB/c spleen and of CD11c+ DCs in BALB/c and NOD spleen. Strictly gluten-free (SGF) diet increased the percentage of CD103+ DCs in BALB/c mice and decreased percentages of CD11b+ DCs in mesenteric and pancreatic lymph nodes in BALB/c mice. SGF diet in BALB/c mice also decreased DC expression of CD40, CCR7 and MHC-II in pancreatic lymph nodes. In conclusion, GF diet changes the composition of the innate immune system in BALB/c and NOD mice and increases expression of DC activation markers in NOD mice. These results contribute to the explanation of the low diabetes incidence in GF NOD mice. This mechanism may be important in development of type 1 diabetes, celiac disease and non-celiac gluten sensitivity.
Collapse
Affiliation(s)
- Jesper Larsen
- The Bartholin Institute, Rigshospitalet, 2100, Copenhagen, Denmark
- * E-mail:
| | - Christian Weile
- The Bartholin Institute, Rigshospitalet, 2100, Copenhagen, Denmark
| | | | - Kåre Engkilde
- The Bartholin Institute, Rigshospitalet, 2100, Copenhagen, Denmark
| | | | - Knud Josefsen
- The Bartholin Institute, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Karsten Buschard
- The Bartholin Institute, Rigshospitalet, 2100, Copenhagen, Denmark
| |
Collapse
|
32
|
Adlercreutz EH, Weile C, Larsen J, Engkilde K, Agardh D, Buschard K, Antvorskov JC. A gluten-free diet lowers NKG2D and ligand expression in BALB/c and non-obese diabetic (NOD) mice. Clin Exp Immunol 2014; 177:391-403. [PMID: 24673402 PMCID: PMC4226590 DOI: 10.1111/cei.12340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/22/2022] Open
Abstract
The interplay between diet and immune parameters which could affect type 1 diabetes (T1D) pathogenesis is not sufficiently clarified. Intestinal up-regulation of the activating receptor natural killer group 2D (NKG2D) (CD314) and its ligands is a hallmark of coeliac disease. However, the direct effect of gluten on NKG2D expression is not known. We studied, by fluorescence activated cell sorter (lymphoid tissues) and reverse transcription–quantitative polymerase chain reaction (intestine and pancreatic islets), if a gluten-free diet (GF diet) from 4 weeks of age or a gluten-free diet introduced in breeding pairs (SGF diet), induced changes in NKG2D expression on DX5+(CD49b) natural killer (NK) cells, CD8+ T cells and in intestinal and islet levels of NKG2D and ligands in BALB/c and non-obese diabetic (NOD) mice. Gluten-free NOD mice had lower insulitis (P < 0·0001); reduced expression of NKG2D on DX5+ NK cells in spleen and auricular lymph nodes (P < 0·05); and on CD8+ T cells in pancreas-associated lymph nodes (P = 0·04). Moreover, the level of CD71 on DX5+ NK cells and CD8+ T cells (P < 0·005) was markedly reduced. GF and SGF mice had reduced expression of NKG2D and DX5 mRNA in intestine (P < 0·05). Differences in intestinal mRNA expression were found in mice at 8, 13 and 20 weeks. Intestinal expression of NKG2D ligands was reduced in SGF mice with lower expression of all ligands. In isolated islets, a SGF diet induced a higher expression of specific NKG2D ligands. Our data show that a gluten-free diet reduces the level of NKG2D and the expression of NKG2D ligands. These immunological changes may contribute to the lower T1D incidence associated with a gluten-free diet.
Collapse
Affiliation(s)
- E H Adlercreutz
- Diabetes and Celiac Disease Unit, Lund University, Malmö, Sweden
| | | | | | | | | | | | | |
Collapse
|
33
|
Larsen J, Dall M, Antvorskov JC, Weile C, Engkilde K, Josefsen K, Buschard K. Dietary gluten increases natural killer cell cytotoxicity and cytokine secretion. Eur J Immunol 2014; 44:3056-67. [PMID: 25043259 DOI: 10.1002/eji.201344264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 05/23/2014] [Accepted: 07/04/2014] [Indexed: 12/14/2022]
Abstract
Dietary gluten influences the development of type 1 diabetes in nonobese diabetic (NOD) mice and biobreeding rats, and has been shown to influence a wide range of immunological factors in the pancreas and gut. In the present study, the effects of gluten on NK cells were studied in vitro and in vivo. We demonstrated that gliadin increased direct cytotoxicity and IFN-γ secretion from murine splenocytes and NK cells toward the pancreatic beta-cell line MIN6 cells. Additionally, stimulation of MIN6 cells led to a significantly increased proportion of degranulating C57BL/6 CD107a(+) NK cells. Stimulation of C57BL/6 pancreatic islets with gliadin significantly increased secretion of IL-6 more than ninefold. In vivo, the gluten-containing diet led to a higher expression of NKG2D and CD71 on NKp46(+) cells in all lymphoid organs in BALB/c and NOD mice compared with the gluten-free diet. Collectively, our data suggest that dietary gluten increases murine NK-cell activity against pancreatic beta cells. This mechanism may contribute to development of type 1 diabetes and explain the higher disease incidence associated with gluten intake in NOD mice.
Collapse
Affiliation(s)
- Jesper Larsen
- The Bartholin Institute, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
34
|
Antvorskov JC, Josefsen K, Engkilde K, Funda DP, Buschard K. Dietary gluten and the development of type 1 diabetes. Diabetologia 2014; 57:1770-80. [PMID: 24871322 PMCID: PMC4119241 DOI: 10.1007/s00125-014-3265-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 04/09/2014] [Indexed: 01/10/2023]
Abstract
Gluten proteins differ from other cereal proteins as they are partly resistant to enzymatic processing in the intestine, resulting in a continuous exposure of the proteins to the intestinal immune system. In addition to being a disease-initiating factor in coeliac disease (CD), gluten intake might affect type 1 diabetes development. Studies in animal models of type 1 diabetes have documented that the pathogenesis is influenced by diet. Thus, a gluten-free diet largely prevents diabetes in NOD mice while a cereal-based diet promotes diabetes development. In infants, amount, timing and mode of introduction have been shown to affect the diabetogenic potential of gluten, and some studies now suggest that a gluten-free diet may preserve beta cell function. Other studies have not found this effect. There is evidence that the intestinal immune system plays a primary role in the pathogenesis of type 1 diabetes, as diabetogenic T cells are initially primed in the gut, islet-infiltrating T cells express gut-associated homing receptors, and mesenteric lymphocytes transfer diabetes from NOD mice to NOD/severe combined immunodeficiency (SCID) mice. Thus, gluten may affect diabetes development by influencing proportional changes in immune cell populations or by modifying the cytokine/chemokine pattern towards an inflammatory profile. This supports an important role for gluten intake in the pathogenesis of type 1 diabetes and further studies should be initiated to clarify whether a gluten-free diet could prevent disease in susceptible individuals or be used with newly diagnosed patients to stop disease progression.
Collapse
Affiliation(s)
- Julie C Antvorskov
- The Bartholin Institute, Rigshospitalet, Ole Maaløes Vej 5, section 3733, Copenhagen, Denmark,
| | | | | | | | | |
Collapse
|
35
|
Hansen CHF, Krych L, Buschard K, Metzdorff SB, Nellemann C, Hansen LH, Nielsen DS, Frøkiær H, Skov S, Hansen AK. A maternal gluten-free diet reduces inflammation and diabetes incidence in the offspring of NOD mice. Diabetes 2014; 63:2821-32. [PMID: 24696449 DOI: 10.2337/db13-1612] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Early-life interventions in the intestinal environment have previously been shown to influence diabetes incidence. We therefore hypothesized that a gluten-free (GF) diet, known to decrease the incidence of type 1 diabetes, would protect against the development of diabetes when fed only during the pregnancy and lactation period. Pregnant nonobese diabetic (NOD) mice were fed a GF or standard diet until all pups were weaned to a standard diet. The early-life GF environment dramatically decreased the incidence of diabetes and insulitis. Gut microbiota analysis by 16S rRNA gene sequencing revealed a pronounced difference between both mothers and their offspring on different diets, characterized by increased numbers of Akkermansia, Proteobacteria, and TM7 in the GF diet group. In addition, pancreatic forkhead box P3 regulatory T cells were increased in GF-fed offspring, as were M2 macrophage gene markers and tight junction-related genes in the gut, while intestinal gene expression of proinflammatory cytokines was reduced. An increased proportion of T cells in the pancreas expressing the mucosal integrin α4β7 suggests that the mechanism involves increased trafficking of gut-primed immune cells to the pancreas. In conclusion, a GF diet during fetal and early postnatal life reduces the incidence of diabetes. The mechanism may involve changes in gut microbiota and shifts to a less proinflammatory immunological milieu in the gut and pancreas.
Collapse
Affiliation(s)
- Camilla Hartmann Friis Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | | | - Stine B Metzdorff
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Christine Nellemann
- Division of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - Lars H Hansen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Dennis S Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Hanne Frøkiær
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Søren Skov
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Axel K Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
36
|
Franzén S, Friederich-Persson M, Fasching A, Hansell P, Nangaku M, Palm F. Differences in susceptibility to develop parameters of diabetic nephropathy in four mouse strains with type 1 diabetes. Am J Physiol Renal Physiol 2014; 306:F1171-8. [DOI: 10.1152/ajprenal.00595.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
One-third of diabetes mellitus patients develop diabetic nephropathy, and with underlying mechanisms unknown it is imperative that diabetic animal models resemble human disease. The present study investigated the susceptibility to develop diabetic nephropathy in four commonly used and commercially available mouse strains with type 1 diabetes to determine the suitability of each strain. Type 1 diabetes was induced in C57Bl/6, NMRI, BALB/c, and 129Sv mice by alloxan, and conscious glomerular filtration rate, proteinuria, and oxidative stress levels were measured in control and diabetic animals at baseline and after 5 and 10 wk. Histological alterations were analyzed using periodic acid-Schiff staining. Diabetic C57Bl/6 displayed increased glomerular filtration rate, i.e., hyperfiltration, whereas all other parameters remained unchanged. Diabetic NMRI developed the most pronounced hyperfiltration as well as increased oxidative stress and proteinuria but without glomerular damage. Diabetic BALB/c did not develop hyperfiltration but presented with pronounced proteinuria, increased oxidative stress, and glomerular damage. Diabetic 129Sv displayed proteinuria and increased oxidative stress without glomerular hyperfiltration or damage. However, all strains displayed intrastrain correlation between oxidative stress and proteinuria. In conclusion, diabetic C57Bl/6 and NMRI both developed glomerular hyperfiltration but neither presented with histological damage, although NMRI developed low-degree proteinuria. Thus these strains may be suitable when investigating the mechanism causing hyperfiltration. Neither BALB/c nor 129Sv developed hyperfiltration although both developed pronounced proteinuria. However, only BALB/c developed detectable histological damage. Thus BALB/c may be suitable when studying the roles of proteinuria and histological alterations for the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Stephanie Franzén
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden
| | | | - Angelica Fasching
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Peter Hansell
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo, Japan
| | - Fredrik Palm
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| |
Collapse
|
37
|
Li M, Song LJ, Qin XY. Advances in the cellular immunological pathogenesis of type 1 diabetes. J Cell Mol Med 2014; 18:749-58. [PMID: 24629100 PMCID: PMC4119381 DOI: 10.1111/jcmm.12270] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/30/2014] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of insulin-producing pancreatic β cells. In recent years, the incidence of type 1 diabetes continues to increase. It is supposed that genetic, environmental and immune factors participate in the damage of pancreatic β cells. Both the immune regulation and the immune response are involved in the pathogenesis of type 1 diabetes, in which cellular immunity plays a significant role. For the infiltration of CD4(+) and CD8(+) T lymphocyte, B lymphocytes, natural killer cells, dendritic cells and other immune cells take part in the damage of pancreatic β cells, which ultimately lead to type 1 diabetes. This review outlines the cellular immunological mechanism of type 1 diabetes, with a particular emphasis to T lymphocyte and natural killer cells, and provides the effective immune therapy in T1D, which is approached at three stages. However, future studies will be directed at searching for an effective, safe and long-lasting strategy to enhance the regulation of a diabetogenic immune system with limited toxicity and without global immunosuppression.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Lu-Jun Song
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Xin-Yu Qin
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
38
|
Nielsen DS, Krych Ł, Buschard K, Hansen CHF, Hansen AK. Beyond genetics. Influence of dietary factors and gut microbiota on type 1 diabetes. FEBS Lett 2014; 588:4234-43. [PMID: 24746688 DOI: 10.1016/j.febslet.2014.04.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/31/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease ultimately leading to destruction of insulin secreting β-cells in the pancreas. Genetic susceptibility plays an important role in T1D etiology, but even mono-zygotic twins only have a concordance rate of around 50%, underlining that other factors than purely genetic are involved in disease development. Here we review the influence of dietary and environmental factors on T1D development in humans as well as animal models. Even though data are still inconclusive, there are strong indications that gut microbiota dysbiosis plays an important role in T1D development and evidence from animal models suggests that gut microbiota manipulation might prove valuable in future prevention of T1D in genetically susceptible individuals.
Collapse
Affiliation(s)
- Dennis S Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg C, Denmark.
| | - Łukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
| | | | - Camilla H F Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1871 Frederiksberg C, Denmark
| | - Axel K Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1871 Frederiksberg C, Denmark
| |
Collapse
|