301
|
Adam Rindfleisch J. Prescribing Probiotics. Integr Med (Encinitas) 2012. [DOI: 10.1016/b978-1-4377-1793-8.00086-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
302
|
Remedio RN, Castellar A, Barbosa RA, Gomes RJ, Caetano FH. Morphological analysis of colon goblet cells and submucosa in type I diabetic rats submitted to physical training. Microsc Res Tech 2011; 75:821-8. [PMID: 22213277 DOI: 10.1002/jemt.22000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 11/15/2011] [Indexed: 12/27/2022]
Abstract
Colon layers, especially the submucosa, as well as the secretion of goblet cells are extremely important for the functioning and transit of substances in this organ. However, the damages arising from type I diabetes and the effects of physical training, which plays crucial role in the treatment of this disease, are not yet known in these regions. To analyze the changes in colon submucosa and goblet cells of diabetic rats, as well as the effects of physical training, Wistar rats were divided into four groups: sedentary control, trained control, sedentary diabetic (SD), and trained diabetic (TD). The training protocol consisted of swimming for 60 min a day, 5 days per week, during 8 weeks. Colon samples were collected, processed, and evaluated by histochemical and ultrastructural techniques. Goblet and submucosa cells did not show alterations in shape, size, protein and carbohydrate content, in all treatment groups. Decreased amount of collagen fibers, however, was observed in the submucosa and lamina propria of SD rats, but this alteration was recovered in TDs. The ultrastructural analysis, in turn, revealed greater quantity of Golgi apparatus cisterns in SDs, distinctly than TDs, which showed improvement in this diabetic condition. Thus, physical training was responsible for the recovery of some important diabetic alterations, possibly improving the motility of substances in the large intestine. Nevertheless, it cannot be considered alone in the treatment of this disease, requiring the combined practice of other methods.
Collapse
|
303
|
Krol S, Ellis-Behnke R, Marchetti P. Nanomedicine for treatment of diabetes in an aging population: state-of-the-art and future developments. Maturitas 2011; 73:61-7. [PMID: 22209199 DOI: 10.1016/j.maturitas.2011.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/01/2011] [Accepted: 12/03/2011] [Indexed: 01/08/2023]
Abstract
Nowadays diabetes, especially type 2 diabetes (which is strongly related to the Western diet and life-style), has developed worldwide into an epidemic disease. Nanomedicine aims to provide novel tools for diagnosis, therapy and point-of-care management of patients. Several nanotechnological approaches were developed to improve life quality for patients with insulin-dependent diabetes. They facilitate blood glucose management by non-invasive glucose measurement as well as insulin administration mainly by delivering the fragile protein as protected and targeted formulation via nasal or oral route. In the present review the oral or nasal insulin delivery by polymeric nanoparticles is discussed with focus on physiological change either related to the disease, diabetes or age-related metabolic variations influencing insulin release and bioavailability. One critical point is that new generations of targeted nanoparticle based drugs are developed and optimized for certain metabolic conditions. These conditions may change with age or disease. The influence of age-related factors such as immaturity in very young age, metabolic and physiologic changes in old age or insufficient animal models are still under-investigated not only in nanomedicine but also generally in pharmacology. Summarizing it can be noted that the bioavailability of insulin administered via routes others than subcutaneously is comparably low (max. 60%). Moreover factors like changed gut permeability as described for diabetes type 1 or other metabolic peculiarities such as insulin resistance in case of type 2 diabetes also play a role in affecting the development of novel nanoparticulated drug preparations and can be responsible for unsuccessful translation of promising animal results into human therapy. In future insulin nanoparticle development for diabetes must consider not only requirements imposed by the drug but also metabolic changes inflicted by disease or by age. Moreover new approaches are required for prevention of the disease.
Collapse
Affiliation(s)
- Silke Krol
- Fondazione IRCCS Istituto Neurologico Carlo Besta, IFOM-IEO-Campus, Milan, Italy.
| | | | | |
Collapse
|
304
|
Knip M, Virtanen SM, Becker D, Dupré J, Krischer JP, Åkerblom HK. Early feeding and risk of type 1 diabetes: experiences from the Trial to Reduce Insulin-dependent diabetes mellitus in the Genetically at Risk (TRIGR). Am J Clin Nutr 2011; 94:1814S-1820S. [PMID: 21653795 PMCID: PMC3364078 DOI: 10.3945/ajcn.110.000711] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Short-term breastfeeding and early exposure to complex dietary proteins, such as cow milk proteins and cereals, or to fruit, berries, and roots have been implicated as risk factors for β cell autoimmunity, clinical type 1 diabetes, or both. The Trial to Reduce Insulin-dependent diabetes mellitus in the Genetically at Risk (TRIGR) is an international, randomized, double-blind, controlled intervention trial designed to answer the question of whether weaning to an extensively hydrolyzed formula in infancy will decrease the risk of type 1 diabetes later in childhood. In our pilot study, weaning to a highly hydrolyzed formula decreased by ≈ 50% the cumulative incidence of one or more diabetes-associated autoantibodies by a mean age of 4.7 y. This finding was confirmed in a recent follow-up analysis to 10 y of age. Currently, the full-scale TRIGR takes place in 77 centers in 15 countries. The TRIGR initially recruited 5606 newborn infants with a family member affected by type 1 diabetes and enrolled 2159 eligible subjects who carried a risk-conferring HLA genotype. All recruited mothers were encouraged to breastfeed. The intervention lasted for 6-8 mo with a minimum study formula exposure time of 2 mo, and hydrolyzed casein and standard cow milk-based weaning formulas were compared. Eighty percent of the participants were exposed to the study formula. The overall retention rate over the first 5 y was 87%, and protocol compliance was 94%. The randomization code will be opened when the last recruited child turns 10 y of age (ie, in 2017).
Collapse
Affiliation(s)
- Mikael Knip
- Hospital for Children and Adolescents, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
305
|
Abstract
Diabetes mellitus diagnosed during the first 2 years of life differs from the disease in older children regarding its causes, clinical characteristics, treatment options and needs in terms of education and psychosocial support. Over the past decade, new genetic causes of neonatal diabetes mellitus have been elucidated, including monogenic β-cell defects and chromosome 6q24 abnormalities. In patients with KCNJ11 or ABCC8 mutations and diabetes mellitus, oral sulfonylurea offers an easy and effective treatment option. Type 1 diabetes mellitus in infants is characterized by a more rapid disease onset, poorer residual β-cell function and lower rate of partial remission than in older children. Insulin therapy in infants with type 1 diabetes mellitus or other monogenic causes of diabetes mellitus is a challenge, and novel data highlight the value of continuous subcutaneous insulin infusion in this very young patient population. Infants are entirely dependent on caregivers for insulin therapy, nutrition and glucose monitoring, which emphasizes the need for appropriate education and psychosocial support of parents. To achieve optimal long-term metabolic control with low rates of acute and chronic complications, continuous and structured diabetes care should be provided by a multidisciplinary health-care team.
Collapse
Affiliation(s)
- Beate Karges
- Division of Endocrinology and Diabetes, RWTH Aachen University, Pauwelsstraße 30, D-52074 Aachen, Germany.
| | | | | | | | | |
Collapse
|
306
|
Molecular characterization of stool microbiota in HIV-infected subjects by panbacterial and order-level 16S ribosomal DNA (rDNA) quantification and correlations with immune activation. J Acquir Immune Defic Syndr 2011; 57:363-70. [PMID: 21436711 DOI: 10.1097/qai.0b013e31821a603c] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The relationship between gut microbial community composition at the higher-taxonomic order level and local and systemic immunologic abnormalities in HIV disease may provide insight into how bacterial translocation impacts HIV disease. METHODS Antiretroviral-naive patients with HIV underwent upper endoscopy before and 9 months after starting antiretroviral treatment. Duodenal tissue was paraffin-embedded for immunohistochemical analysis and digested for fluorescence activated cell sorting for T-cell subsets and immune activation (CD38+/HLA-DR+) enumeration. Stool samples were provided from patients and control subjects for comparison. Metagenomic microbial DNA was extracted from feces for optimized 16S ribosomal RNA gene (rDNA) real-time quantitative polymerase chain reaction assays designed to quantify panbacterial loads and the relative abundances of proinflammatory Enterobacteriales order and the dominant Bacteroidales and Clostridiales orders. RESULTS Samples from 10 HIV subjects before initiating and from six subjects receiving antiretroviral treatment were available for analysis. There was a trend for a greater proportion of Enterobacteriales in HIV-positive subjects compared with control subjects (P = 0.099). There were significant negative correlations between total bacterial load and duodenal CD4 and CD8 T-cell activation levels (r = -0.74, P = 0.004 and r = -0.67, P = 0.013, respectively). The proportions of Enterobacteriales and Bacteroidales were significantly correlated with duodenal CD4 T-cell depletion and peripheral CD8 T-cell activation, respectively. CONCLUSIONS These data represent the first report of quantitative molecular and cellular correlations between total/universal and order-level gut bacterial populations and gastrointestinal-associated lymphoid tissue levels of immune activation in HIV-infected subjects. The correlations between lower overall 16S rDNA levels and tissue immune activation suggest that the gut microbiome may contribute to immune activation and influence HIV progression.
Collapse
|
307
|
Kolodziej LE, Lodolce JP, Chang JE, Schneider JR, Grimm WA, Bartulis SJ, Zhu X, Messer JS, Murphy SF, Reddy N, Turner JR, Boone DL. TNFAIP3 maintains intestinal barrier function and supports epithelial cell tight junctions. PLoS One 2011; 6:e26352. [PMID: 22031828 PMCID: PMC3198775 DOI: 10.1371/journal.pone.0026352] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 09/25/2011] [Indexed: 02/07/2023] Open
Abstract
Tight junctions between intestinal epithelial cells mediate the permeability of the intestinal barrier, and loss of intestinal barrier function mediated by TNF signaling is associated with the inflammatory pathophysiology observed in Crohn's disease and celiac disease. Thus, factors that modulate intestinal epithelial cell response to TNF may be critical for the maintenance of barrier function. TNF alpha-induced protein 3 (TNFAIP3) is a cytosolic protein that acts in a negative feedback loop to regulate cell signaling induced by Toll-like receptor ligands and TNF, suggesting that TNFAIP3 may play a role in regulating the intestinal barrier. To investigate the specific role of TNFAIP3 in intestinal barrier function we assessed barrier permeability in TNFAIP3−/− mice and LPS-treated villin-TNFAIP3 transgenic mice. TNFAIP3−/− mice had greater intestinal permeability compared to wild-type littermates, while villin-TNFAIP3 transgenic mice were protected from increases in permeability seen within LPS-treated wild-type littermates, indicating that barrier permeability is controlled by TNFAIP3. In cultured human intestinal epithelial cell lines, TNFAIP3 expression regulated both TNF-induced and myosin light chain kinase-regulated tight junction dynamics but did not affect myosin light chain kinase activity. Immunohistochemistry of mouse intestine revealed that TNFAIP3 expression inhibits LPS-induced loss of the tight junction protein occludin from the apical border of the intestinal epithelium. We also found that TNFAIP3 deubiquitinates polyubiquitinated occludin. These in vivo and in vitro studies support the role of TNFAIP3 in promoting intestinal epithelial barrier integrity and demonstrate its novel ability to maintain intestinal homeostasis through tight junction protein regulation.
Collapse
Affiliation(s)
- Lauren E. Kolodziej
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - James P. Lodolce
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jonathan E. Chang
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jeffrey R. Schneider
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Wesley A. Grimm
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Sarah J. Bartulis
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Xiaorong Zhu
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jeannette S. Messer
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Stephen F. Murphy
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Nishith Reddy
- Illinois Math and Science Academy, Aurora, Illinois, United States of America
| | - Jerrold R. Turner
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - David L. Boone
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
308
|
Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One 2011; 6:e25792. [PMID: 22043294 PMCID: PMC3197175 DOI: 10.1371/journal.pone.0025792] [Citation(s) in RCA: 582] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 09/11/2011] [Indexed: 12/12/2022] Open
Abstract
Recent studies have suggested a bacterial role in the development of autoimmune disorders including type 1 diabetes (T1D). Over 30 billion nucleotide bases of Illumina shotgun metagenomic data were analyzed from stool samples collected from four pairs of matched T1D case-control subjects collected at the time of the development of T1D associated autoimmunity (i.e., autoantibodies). From these, approximately one million open reading frames were predicted and compared to the SEED protein database. Of the 3,849 functions identified in these samples, 144 and 797 were statistically more prevalent in cases and controls, respectively. Genes involved in carbohydrate metabolism, adhesions, motility, phages, prophages, sulfur metabolism, and stress responses were more abundant in cases while genes with roles in DNA and protein metabolism, aerobic respiration, and amino acid synthesis were more common in controls. These data suggest that increased adhesion and flagella synthesis in autoimmune subjects may be involved in triggering a T1D associated autoimmune response. Extensive differences in metabolic potential indicate that autoimmune subjects have a functionally aberrant microbiome. Mining 16S rRNA data from these datasets showed a higher proportion of butyrate-producing and mucin-degrading bacteria in controls compared to cases, while those bacteria that produce short chain fatty acids other than butyrate were higher in cases. Thus, a key rate-limiting step in butyrate synthesis is more abundant in controls. These data suggest that a consortium of lactate- and butyrate-producing bacteria in a healthy gut induce a sufficient amount of mucin synthesis to maintain gut integrity. In contrast, non-butyrate-producing lactate-utilizing bacteria prevent optimal mucin synthesis, as identified in autoimmune subjects.
Collapse
|
309
|
Amir J, Waite M, Tobler J, Catalfamo DL, Koutouzis T, Katz J, Wallet SM. The role of hyperglycemia in mechanisms of exacerbated inflammatory responses within the oral cavity. Cell Immunol 2011; 272:45-52. [PMID: 21996642 PMCID: PMC3221011 DOI: 10.1016/j.cellimm.2011.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 08/11/2011] [Accepted: 09/19/2011] [Indexed: 02/06/2023]
Abstract
Immune modulating factors are necessary for pathogen clearance, but also contribute to host tissues damage, as those seen in periodontal diseases. Many of these responses can be exacerbated by host conditions including type 2 diabetes [T2D], where toll-like receptor 4 [TLR4] and the receptor for advanced glycated end products [RAGE] play a significant role. Here we investigate causality associated with the increase in inflammatory markers observed in periodontally diseased patients with T2D using multi-variant correlation analysis. Inflammation associated with periodontal diseases, characterized by elevated pro-inflammatory cytokines, innate immune receptor expression, and cellular infiltrate was exacerbated in patients with T2D. In addition, a feed forward loop regulated by poor glycemic control was associated with a loss of mucosal barrier integrity and accumulation of innate immune receptor ligands resulting in an exacerbation of ongoing inflammation, where RAGE and TLR4 cooperated to induce responses in oral epithelial cells, which were exacerbated by hyperglycemia.
Collapse
Affiliation(s)
- Jamie Amir
- Department of Periodontology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| | - Matthew Waite
- Department of Periodontology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| | - Jeffrey Tobler
- Department of Periodontology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| | - Dana L. Catalfamo
- Department of Periodontology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
- Department of Oral Biology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| | - Theofilos Koutouzis
- Department of Periodontology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| | - Joseph Katz
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| | - Shannon M. Wallet
- Department of Periodontology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
- Department of Oral Biology, University of Florida, College of Dentistry, BOX 100434, Gainesville, FL 32610, USA
| |
Collapse
|
310
|
Wasserfall C, Nead K, Mathews C, Atkinson MA. The threshold hypothesis: solving the equation of nurture vs nature in type 1 diabetes. Diabetologia 2011; 54:2232-6. [PMID: 21773685 PMCID: PMC7296551 DOI: 10.1007/s00125-011-2244-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 06/20/2011] [Indexed: 02/06/2023]
Abstract
For more than 40 years, the contributions of nurture (i.e. the environment) and nature (i.e. genetics) have been touted for their aetiological importance in type 1 diabetes. Disappointingly, knowledge gains in these areas, while individually successful, have to a large extent occurred in isolation from each other. One reason underlying this divide is the lack of a testable model that simultaneously considers the contributions of genetic and environmental determinants in the formation of this and potentially other disorders that are subject to these variables. To address this void, we have designed a model based on the hypothesis that the aetiological influences of genetics and environment, when evaluated as intersecting and reciprocal trend lines based on odds ratios, result in a method of concurrently evaluating both facets and defining the attributable risk of clinical onset of type 1 diabetes. The model, which we have elected to term the 'threshold hypothesis', also provides a novel means of conceptualising the complex interactions of nurture with nature in type 1 diabetes across various geographical populations.
Collapse
Affiliation(s)
- C Wasserfall
- Department of Pathology, University of Florida, College of Medicine, 1600 SW Archer Road, Gainesville, FL 32610-0275, USA
| | | | | | | |
Collapse
|
311
|
Vorobjova T, Uibo O, Ojakivi I, Teesalu K, Panarina M, Heilman K, Uibo R. Lower expression of tight junction protein 1 gene and increased FOXP3 expression in the small bowel mucosa in coeliac disease and associated type 1 diabetes mellitus. Int Arch Allergy Immunol 2011; 156:451-461. [PMID: 21832836 DOI: 10.1159/000324456] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 01/17/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The role of regulatory T cells expressing FOXP3 in the pathogenesis of coeliac disease (CD) and type 1 diabetes (T1D) has been reported. Recent data have placed special focus on the interplay between the intestinal barrier and immunoregulatory processes. We aimed to determine whether the expression of tight junction protein 1 (TJP1), which reflects small bowel mucosa permeability, is changed in CD and T1D. METHODS Transcription levels of TJP1 and FOXP3 genes were evaluated in the small bowel biopsies of 14 children with CD, 12 with CD and coexisting T1D and 40 controls using real-time PCR. Serum IgA and IgG to deamidated gliadin, bovine β-lactoglobulin, bovine α-casein and human tissue transglutaminase (tTG) were determined by ELISA. RESULTS The highest expression of FOXP3 mRNA was seen in patients with CD and T1D compared to patients with CD alone and controls (p = 0.02). In contrast, the lowest level of TJP1 mRNA expression was found in patients with CD and T1D (p = 0.01). The levels of IgA to deamidated gliadin and tTG were highest in patients with CD and T1D (p = 0.0001 and 0.01, respectively). The expression of FOXP3 mRNA correlated highly with the level of anti-gliadin IgA (p = 0.02) and anti-tTG IgA antibodies (p = 0.004). CONCLUSION The significant decline in TJP1 expression in CD patients, particularly in those with coexisting T1D, was accompanied by an increase in FOXP3 expression. This might reflect an attempt to maintain immune tolerance to counterbalance the loss of mucosal integrity in the small intestine in CD associated with T1D.
Collapse
Affiliation(s)
- Tamara Vorobjova
- Institute of General and Molecular Pathology, Department of Immunology, University of Tartu, Ravila 19, Tartu, Estonia.
| | | | | | | | | | | | | |
Collapse
|
312
|
Abstract
PURPOSE OF REVIEW Several studies indicate that factors affecting the gut are capable of modulating the development of autoimmune diabetes. This review discusses the recent research on these mechanisms, which may reveal novel pathogenic pathways and new possibilities for prevention of type 1 diabetes (T1D). RECENT FINDINGS The role of the gut as a regulator of T1D is mainly based on animal studies in which changes affecting the gut immune system have been shown to modulate the immune-mediated destruction of insulin-producing beta-cells. Dietary interventions, alterations in the intestinal microbiota and exposure to enteral pathogens regulate the development of autoimmune diabetes in animal models. In several studies, it has been demonstrated that these modulations affect the gut barrier mechanisms and intestinal immunity. Also, in humans, increased gut permeability and intestinal inflammation are associated with T1D. A recent report of dietary intervention study in infants at genetic risk of T1D showed that early diet could modulate the development of beta-cell autoimmunity in humans; weaning to hydrolyzed casein formula decreased the risk of beta-cell autoimmunity by age 10. SUMMARY The gut modulation affecting permeability, inflammation and microbiota is evidently associated with the regulation of the inflammation leading to beta-cell destruction. Although the mechanisms of action are not fully understood, the recent research points out the lines of approach for the prevention of T1D.
Collapse
Affiliation(s)
- Outi Vaarala
- Immune Response Unit, Department of Vaccination and Immune Protection, National Institute for Health and Welfare, Helsinki, Finland.
| |
Collapse
|
313
|
Conterno L, Fava F, Viola R, Tuohy KM. Obesity and the gut microbiota: does up-regulating colonic fermentation protect against obesity and metabolic disease? GENES & NUTRITION 2011; 6:241-60. [PMID: 21559992 PMCID: PMC3145060 DOI: 10.1007/s12263-011-0230-1] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 04/20/2011] [Indexed: 12/18/2022]
Abstract
Obesity is now considered a major public health concern globally as it predisposes to a number of chronic human diseases. Most developed countries have experienced a dramatic and significant rise in obesity since the 1980s, with obesity apparently accompanying, hand in hand, the adoption of "Western"-style diets and low-energy expenditure lifestyles around the world. Recent studies report an aberrant gut microbiota in obese subjects and that gut microbial metabolic activities, especially carbohydrate fermentation and bile acid metabolism, can impact on a number of mammalian physiological functions linked to obesity. The aim of this review is to present the evidence for a characteristic "obese-type" gut microbiota and to discuss studies linking microbial metabolic activities with mammalian regulation of lipid and glucose metabolism, thermogenesis, satiety, and chronic systemic inflammation. We focus in particular on short-chain fatty acids (SCFA) produced upon fiber fermentation in the colon. Although SCFA are reported to be elevated in the feces of obese individuals, they are also, in contradiction, identified as key metabolic regulators of the physiological checks and controls mammals rely upon to regulate energy metabolism. Most studies suggest that the gut microbiota differs in composition between lean and obese individuals and that diet, especially the high-fat low-fiber Western-style diet, dramatically impacts on the gut microbiota. There is currently no consensus as to whether the gut microbiota plays a causative role in obesity or is modulated in response to the obese state itself or the diet in obesity. Further studies, especially on the regulatory role of SCFA in human energy homeostasis, are needed to clarify the physiological consequences of an "obese-style" microbiota and any putative dietary modulation of associated disease risk.
Collapse
Affiliation(s)
- Lorenza Conterno
- Nutrition and Nutrigenomics Group, Research and Innovation Centre, FEM-IASMA, 38010, S. Michele a.A, Trento, Italy,
| | | | | | | |
Collapse
|
314
|
Virtanen SM, Takkinen HM, Nevalainen J, Kronberg-Kippilä C, Salmenhaara M, Uusitalo L, Kenward MG, Erkkola M, Veijola R, Simell O, Ilonen J, Knip M. Early introduction of root vegetables in infancy associated with advanced ß-cell autoimmunity in young children with human leukocyte antigen-conferred susceptibility to Type 1 diabetes. Diabet Med 2011; 28:965-71. [PMID: 21418094 DOI: 10.1111/j.1464-5491.2011.03294.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS Early introduction of supplementary foods has been implicated to play a role in the development of ß-cell autoimmunity. We set out to study the effects of breastfeeding and age at introduction of supplementary foods on the development of ß-cell autoimmunity. METHODS A prospective birth cohort of 6069 infants with HLA-DQB-conferred susceptibility to Type 1 diabetes was recruited between 1996 and 2004. Antibodies against islet cells, insulin, glutamate dehydroxylase and islet antigen 2 were measured at 3- to 12-month intervals. The families recorded at home the age at introduction of new foods and, for each visit, completed a structured dietary questionnaire. The endpoint was repeated positivity for islet cell antibodies plus at least one other antibody and/or clinical Type 1 diabetes (n = 265). RESULTS Early introduction of root vegetables (by the age of 4 months) was related to increased risk of developing positivity for the endpoint [hazard ratio (95% CI) for the earliest third 1.75 (1.11-2.75) and for the middle third 1.79 (1.22-2.62) compared with the last third (> 4 months), likelihood ratio test P = 0.006], independently of the introduction of other foods and of several putative socio-demographic and perinatal confounding factors. Introducing wheat, rye, oats and/or barley cereals (P = 0.013) and egg (P = 0.031) early was related to an increased risk of the endpoint, but only during the first 3 years of life. CONCLUSIONS Early introduction of root vegetables during infancy is independently associated with increased risk of ß-cell autoimmunity among Finnish children with increased genetic susceptibility to Type 1 diabetes.
Collapse
Affiliation(s)
- S M Virtanen
- Department of Lifestyle and Participation, Nutrition Unit, National Institute for Health and Welfare, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
315
|
Badami E, Sorini C, Coccia M, Usuelli V, Molteni L, Bolla AM, Scavini M, Mariani A, King C, Bosi E, Falcone M. Defective differentiation of regulatory FoxP3+ T cells by small-intestinal dendritic cells in patients with type 1 diabetes. Diabetes 2011; 60:2120-4. [PMID: 21646390 PMCID: PMC3142071 DOI: 10.2337/db10-1201] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The gut environment modulates the pathogenesis of type 1 diabetes (T1D), but how it affects autoimmunity toward pancreatic β-cells, a self-tissue located outside the intestine, is still unclear. In the small intestine, lamina propria dendritic cells (LPDCs) induce peripheral differentiation of FoxP3(+) regulatory T (Treg) cells. We tested the hypothesis that the intestinal milieu impinges on human T1D by affecting differentiation of FoxP3(+) Treg cells. RESEARCH DESIGN AND METHODS We collected duodenal biopsies of 10 T1D patients, 16 healthy subjects, and 20 celiac individuals and performed a fluorescent-activated cell sorter analysis to measure percentages of various immune cell subsets, including CD4(+) and CD8(+) T cells, NK cells, γδ T cells, CD103(+)CD11c(+) LPDCs, and CD4(+)CD25(+)FoxP3(+)CD127(-) Treg cells. In parallel, we assessed the tolerogenic function (i.e., capacity to induce differentiation of FoxP3(+) Treg cells) by LPDCs of T1D patients and control subjects. RESULTS Our analysis revealed a significant reduction in the percentage of intestinal CD4(+)CD25(+)FoxP3(+)CD127(-) Treg cells in T1D patients compared with healthy subjects (P = 0.03) and celiac individuals (P = 0.003). In addition, we found that LPDCs from T1D patients completely lacked their tolerogenic function; they were unable to convert CD4(+)CD25(-) T cells into CD4(+)CD25(+)FoxP3(+)CD127(-) Treg cells. CONCLUSIONS Our data indicate that T1D patients have a reduced number of intestinal FoxP3(+) Treg cells as a result of their defective differentiation in the gut. These findings suggest that intestinal immune regulation is not only calibrated to tolerate commensal bacteria and food components but also is instrumental in maintaining immune tolerance toward pancreatic β-cells and preventing T1D.
Collapse
Affiliation(s)
- Ester Badami
- Experimental Diabetes Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Sorini
- Experimental Diabetes Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Margherita Coccia
- Experimental Diabetes Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Vera Usuelli
- Experimental Diabetes Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Laura Molteni
- Department of Internal Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Mario Bolla
- Department of Internal Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Marina Scavini
- Department of Internal Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Mariani
- Gastroenterology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Cecile King
- Department of Immunology, Garvan Institute for Medical Research, Darlinhurst, New South Wales, Australia
| | - Emanuele Bosi
- Department of Internal Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Marika Falcone
- Experimental Diabetes Unit, Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Corresponding author: Marika Falcone,
| |
Collapse
|
316
|
Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci U S A 2011; 108:11548-53. [PMID: 21709219 DOI: 10.1073/pnas.1108924108] [Citation(s) in RCA: 338] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vertebrates typically harbor a rich gastrointestinal microbiota, which has coevolved with the host over millennia and is essential for several host physiological functions, in particular maturation of the immune system. Recent studies have highlighted the importance of a single bacterial species, segmented filamentous bacteria (SFB), in inducing a robust T-helper cell type 17 (Th17) population in the small-intestinal lamina propria (SI-LP) of the mouse gut. Consequently, SFB can promote IL-17-dependent immune and autoimmune responses, gut-associated as well as systemic, including inflammatory arthritis and experimental autoimmune encephalomyelitis. Here, we exploit the incomplete penetrance of SFB colonization of NOD mice in our animal facility to explore its impact on the incidence and course of type 1 diabetes in this prototypical, spontaneous model. There was a strong cosegregation of SFB positivity and diabetes protection in females, but not in males, which remained relatively disease-free regardless of the SFB status. In contrast, insulitis did not depend on SFB colonization. SFB-positive, but not SFB-negative, females had a substantial population of Th17 cells in the SI-LP, which was the only significant, repeatable difference in the examined T-cell compartments of the gut, pancreas, or systemic lymphoid tissues. Th17-signature transcripts dominated the very limited SFB-induced molecular changes detected in SI-LP CD4(+) T cells. Thus, a single bacterium, and the gut immune system alterations associated with it, can either promote or protect from autoimmunity in predisposed mouse models, probably reflecting their variable dependence on different Th subsets.
Collapse
|
317
|
Alam C, Bittoun E, Bhagwat D, Valkonen S, Saari A, Jaakkola U, Eerola E, Huovinen P, Hänninen A. Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice. Diabetologia 2011; 54:1398-406. [PMID: 21380595 DOI: 10.1007/s00125-011-2097-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 02/01/2011] [Indexed: 01/24/2023]
Abstract
AIMS/HYPOTHESIS Microbial factors influence the development of diabetes in NOD mice. Studies in germ-free animals have revealed important roles of microbiota in the regulation of Th17 and forkhead box P3 (FOXP3)(+) T regulatory (Treg) activation in the intestine. However, the effects of intestinal microbiota in immune regulation and diabetes development in NOD mice are still poorly understood. METHODS A colony of germ-free NOD mice was established to evaluate the effects of intestinal microbiota on regulatory immunity in the gut, and on the development of insulitis and diabetes in NOD mice. RESULTS Diabetes developed in roughly equal numbers in germ-free and specific pathogen-free NOD mice. Insulitis was accentuated in germ-free NOD mice; yet insulin preservation was unaltered. Germ-free NOD mice showed increased levels of Il17 (also known as Il17a) mRNA in the colon, and of Th17 and Th1 cells in the mesenteric and pancreatic lymph nodes, while Foxp3 mRNA and FOXP3(+) Tregs were reduced. In the islet infiltrates, FOXP3(+)CD4(+) T cells were slightly increased in germ-free mice. B cells appeared less activated in the peritoneum and were less abundant in islet infiltrates. CONCLUSIONS/INTERPRETATION These results indicate that lack of intestinal microbiota promotes an imbalance between Th1, Th17 and Treg differentiation in the intestine. This imbalance is associated with accelerated insulitis, but intact recruitment of FOXP3(+) Tregs into islets, suggesting: (1) a microbial dependence of local induction of Treg in the gut and draining lymph nodes; but (2) a potentially compensatory function of naturally occurring Tregs in the islets, which may help control diabetogenic T cells.
Collapse
Affiliation(s)
- C Alam
- Department of Medical Microbiology and Immunology, Kiinamyllynkatu 13, University of Turku, 20520 Turku, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
318
|
Ponsonby AL, Pezic A, Cochrane J, Cameron FJ, Pascoe M, Kemp A, Dwyer T. Infant anthropometry, early life infection, and subsequent risk of type 1 diabetes mellitus: a prospective birth cohort study. Pediatr Diabetes 2011; 12:313-21. [PMID: 21615650 DOI: 10.1111/j.1399-5448.2010.00693.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Higher birthweight is associated with increased type 1 diabetes mellitus (T1DM) risk, but the contribution of higher adiposity or lean mass is unclear. In this Tasmanian infant cohort, early upper respiratory infection has been associated with higher asthma risk. PATIENTS AND METHODS Eligible infants represented one-fifth of live births in Tasmania, 1988-1995. Hospital interview data (day 6) were obtained on 96.3% (10 628/11 040), home (5 wk) visit data (38 d) on 92.9% (9876/10 628) of those, then a phone (12 wk) interview (87 d). Tricep and subscapular skinfold measures and upper arm circumference were recorded at the first two interviews. T1DM cases (n = 26) arising from the age of 16 or under in Tasmania from 1988 to 2006 were ascertained. RESULTS Higher birthweight [adjusted odds ratio (AOR) 2.82 (95% CI 1.31-6.09)], lean mid-upper arm circumference [AOR 1.76 (95% CI 1.16-2.66)], not skinfold measures, were associated with T1DM risk. Children with an early upper respiratory tract infection by 5-wk visit [AOR 2.74 (95% CI 1.19-6.32)] or ear infection by 12-wk interview [AOR 3.44 (95% CI 1.00-11.79)] were also at higher risk. Putative markers of altered microbial exposure such as resident density were not associated with T1DM risk but the effect of increasing birth order on T1DM risk differed for older (AOR 0.41, p = 0.02) than young mother (AOR 2.45, p = 0.01); difference in effect, p = 0.001. CONCLUSION In this cohort, early upper respiratory tract infection was associated with T1DM risk, as had been previously found for asthma, consistent with immunoinflammatory upregulation. Using the detailed anthropometric measures available, the link between higher birthweight and T1DM did not appear to reflect increased adiposity.
Collapse
Affiliation(s)
- Anne-Louise Ponsonby
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
319
|
Abstract
Coeliac disease (CD) is a systemic immune-mediated disorder elicited by gluten in genetically susceptible individuals. The common factor for all patients with CD is the presence of a variable combination of gluten-dependent clinical manifestations, specific autoantibodies (anti-tissue transglutaminase/anti-endomysium), HLA-DQ2 and/or DQ8 haplotypes and different degrees of enteropathy. Recently, gluten sensitivity has received much interest, although the limits and possible overlap between gluten sensitivity and CD remain poorly defined. At present, a number of morphological, functional and immunological disorders that are lacking one or more of the key CD criteria (enteropathy, associated HLA haplotypes and presence of anti-transglutaminase two antibodies) but respond to gluten exclusion are included under the umbrella of gluten sensitivity. The possible immunological mechanisms underlying these conditions are discussed. Emphasis is given to specific autoantibodies as markers of the coeliac spectrum and to the hypothesis that innate epithelial stress can exist independently from adaptive intestinal immunity in gluten sensitivity.
Collapse
Affiliation(s)
- R Troncone
- Department of Pediatrics and European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy.
| | | |
Collapse
|
320
|
A cluster of coregulated genes determines TGF-beta-induced regulatory T-cell (Treg) dysfunction in NOD mice. Proc Natl Acad Sci U S A 2011; 108:8737-42. [PMID: 21543717 DOI: 10.1073/pnas.1105364108] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Foxp3(+) regulatory T cells (Tregs) originate in the thymus, but the Treg phenotype can also be induced in peripheral lymphoid organs or in vitro by stimulation of conventional CD4(+) T cells with IL-2 and TGF-β. There have been divergent reports on the suppressive capacity of these TGF-Treg cells. We find that TGF-Tregs derived from diabetes-prone NOD mice, although expressing normal Foxp3 levels, are uniquely defective in suppressive activity, whereas TGF-Tregs from control strains (B6g7) or ex vivo Tregs from NOD mice all function normally. Most Treg-typical transcripts were shared by NOD or B6g7 TGF-Tregs, except for a small group of differentially expressed genes, including genes relevant for suppressive activity (Lrrc32, Ctla4, and Cd73). Many of these transcripts form a coregulated cluster in a broader analysis of T-cell differentiation. The defect does not map to idd3 or idd5 regions. Whereas Treg cells from NOD mice are normal in spleen and lymph nodes, the NOD defect is observed in locations that have been tied to pathogenesis of diabetes (small intestine lamina propria and pancreatic lymph node). Thus, a genetic defect uniquely affects a specific Treg subpopulation in NOD mice, in a manner consistent with a role in determining diabetes susceptibility.
Collapse
|
321
|
Bech-Nielsen GV, Hansen CHF, Hufeldt MR, Nielsen DS, Aasted B, Vogensen FK, Midtvedt T, Hansen AK. Manipulation of the gut microbiota in C57BL/6 mice changes glucose tolerance without affecting weight development and gut mucosal immunity. Res Vet Sci 2011; 92:501-8. [PMID: 21543097 DOI: 10.1016/j.rvsc.2011.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 04/05/2011] [Accepted: 04/06/2011] [Indexed: 02/06/2023]
Abstract
Inflammatory diseases such as type 2 diabetes (T2D) in humans and mice are under the influence of the composition of the gut microbiota (GM). It was previously demonstrated that treating Lep(ob) mice with antibiotics improved glucose tolerance. However, wild type C57BL/6J mice may also exhibit plasma glucose intolerance reminiscent of human T2D. We hypothesized that antibiotic treatment in C57BL/6 mice would have an impact on glucose tolerance without affecting weight and gut immunology. When compared to mice treated with erythromycin or the controls, treatment for five weeks with ampicillin improved glucose tolerance without significantly affecting the weight or the number of gut mucosal regulatory T cells, tolerogenic dendritic cells or T helper cells type 1. 16S rRNA gene based denaturing gradient gel electrophoresis profiles clearly clustered according to treatment and showed that antibiotic treatment reduced GM diversity. It is concluded that antibiotic treatment changes glucose metabolism as well as the composition of the GM in C57BL/6 mice, and that this does not seem to be correlated to weight development in the mice.
Collapse
Affiliation(s)
- Gunilla Veslemøy Bech-Nielsen
- Department of Veterinary Disease Biology, Faculty of Life Sciences, University of Copenhagen, Grønnegårdsvej 18, DK-1870 Frederiksberg, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
322
|
Giongo A, Atkinson MA, Triplett EW. Microbiology of Type 1 diabetes: possible implications for management of the disease. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/dmt.11.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
323
|
Buschard K, Hansen AK, Jensen K, Lindenbergh-Kortleve DJ, de Ruiter LF, Krohn TC, Hufeldt MR, Vogensen FK, Aasted B, Osterbye T, Roep BO, de Haar C, Nieuwenhuis EE. Alcohol facilitates CD1d loading, subsequent activation of NKT cells, and reduces the incidence of diabetes in NOD mice. PLoS One 2011; 6:e17931. [PMID: 21483778 PMCID: PMC3069977 DOI: 10.1371/journal.pone.0017931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 02/20/2011] [Indexed: 01/13/2023] Open
Abstract
Background Ethanol (‘alcohol’) is a partly hydrophobic detergent that may affect the accessibility of glycolipids thereby influencing immunological effects of these molecules. Methods The study included cellular in vitro tests using α-galactosylceramide (αGalCer), and in vivo NOD mice experiments detecting diabetes incidence and performing behavioural and bacterial analyses. Results Alcohol in concentrations from 0.6% to 2.5% increased IL-2 production from NKT cells stimulated with αGalCer by 60% (p<0.05). CD1d expressed on HeLa cells contained significantly increasing amounts of αGalCer with increasing concentrations of alcohol, suggesting that alcohol facilitated the passive loading of αGalCer to CD1d. NOD mice were found to tolerate 5% ethanol in their drinking water without signs of impairment in liver function. Giving this treatment, the diabetes incidence declined significantly. Higher numbers of CD3+CD49b+ NKT cells were found in spleen and liver of the alcohol treated compared to the control mice (p<0.05), whereas the amount of CD4+Foxp3+ regulator T cells did not differ. Increased concentrations of IFN-γ were detected in 24-hour blood samples of alcohol treated mice. Behavioural studies showed no change in attitude of the ethanol-consuming mice, and bacterial composition of caecum samples was not affected by alcohol, disqualifying these as protective mechanisms. Conclusion Alcohol facilitates the uptake of glycolipids and the stimulation of NKT cells, which are known to counteract Type 1 diabetes development. We propose that this is the acting mechanism by which treatment with alcohol reduces the incidence of diabetes in NOD mice. This is corroborated by epidemiology showing beneficial effect of alcohol to reduce the severity of atherosclerosis and related diseases.
Collapse
|
324
|
Uibo R, Panarina M, Teesalu K, Talja I, Sepp E, Utt M, Mikelsaar M, Heilman K, Uibo O, Vorobjova T. Celiac disease in patients with type 1 diabetes: a condition with distinct changes in intestinal immunity? Cell Mol Immunol 2011; 8:150-156. [PMID: 21317917 PMCID: PMC4003136 DOI: 10.1038/cmi.2010.66] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 12/09/2010] [Indexed: 02/06/2023] Open
Abstract
Two common chronic childhood diseases-celiac disease (CD) and type 1 diabetes (T1D)-result from complex pathological mechanisms where genetic susceptibility, environmental exposure, alterations in intestinal permeability and immune responses play central roles. In this study, we investigated whether these characteristics were universal for CD independently of T1D association. For this purpose, we studied 36 children with normal small-bowel mucosa and 26 children with active CD, including 12 patients with T1D. In samples from the small-bowel mucosa, we detected the lowest expression of tight junction protein 1 (TJP1) mRNA in CD patients with T1D, indicating an increase in intestinal permeability. Furthermore, these samples displayed the highest expression of forkhead box P3 (FoxP3) mRNA, a marker for regulatory T cells, as compared with other patient groups. At the same time, serum levels of IgA antibodies specific for the CD-related antigens deamidated gliadin and tissue transglutaminase (tTG) were the highest in CD patients with T1D. In contrast, no significant differences were found in IgA or IgG antibodies specific for bovine beta-lactoglobulin or Bifidobacterium adolescentis DSM 20083-derived proteins. There were also no differences in the transamidating activity of serum autoantibodies between patients and control individuals. Our results show that patients with T1D and newly detected CD exhibit severely altered intestinal permeability, strong local immune activation and increased immunoregulatory mechanisms in the small bowel. Further study is required to determine whether these extreme changes in this CD subgroup are due to some specific environmental factors (virus infections), unknown genetic effects or autoimmune reactions to antigenic targets in intracellular tight junctions.
Collapse
Affiliation(s)
- Raivo Uibo
- Department of Immunology, University of Tartu, Tartu, Estonia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Bashiri H, Keshavarz A, Madani H, Hooshmandi A, Bazargan-Hejazi S, Ahmadi A. Celiac disease in type-I diabetes mellitus: coexisting phenomenon. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2011; 16 Suppl 1:S401-6. [PMID: 22247725 PMCID: PMC3252773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/15/2011] [Indexed: 11/13/2022]
Abstract
BACKGROUND This study aimed to determine the prevalence of celiac disease in type I diabetic patients and to compare the symptoms and complications of celiac in patients with diabetes and celiac with patients with diabetes only. METHODS A total of 241 type I diabetic patients age ≥ 18 who needed insulin intake were recruited from diabetic patients attending the Diabetic Research Center in Kermanshah, Iran. Sample was screened for celiac disease by drawing 5cc blood for complete blood count (CBC), and anti-endomysial antibody test (AEA). Patients then were classified based on immunofluorescent method for the presence of AEA. Those with AEA positive underwent biopsy. The biopsy tissues were classified based on Marsh classification. RESULTS Twenty one patients tested positive for celiac disease based on AEA test (8.7%) and 20 (8.3%) tested positive based on the biopsy. Prevalence of celiac among diabetic patients in comparison to normal population was 8.3% vs. 0.6%; and 70% were in the stages III and IV. Weight loss was significantly more prevalent among the celiac patients, who were 4 times more likely to lose weight. Other parameters such as anemia, mucocutaneous and cutaneous hemorrhage, milk intolerance, related oral aphthous, diarrhea and steatorrhea, alopecia, dermatitis herpetiform and alopecia were higher in celiac patients but not high enough to be statistically significant. CONCLUSIONS There is a need to improve screening identification and treatment of celiac among all diabetic patients type I, especially in cases with uncontrolled diabetic or weight loss.
Collapse
Affiliation(s)
- Homayoon Bashiri
- Department of Internal Medicine, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran., Corresponding Author E-mail:
| | - Aliasghar Keshavarz
- Department of Internal Medicine, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamid Madani
- Department of Pathology, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmadreza Hooshmandi
- Department of Internal Medicine, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Alireza Ahmadi
- Department of Anesthesiology, Critical Care and Pain Management, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran., Department of Public Health Sciences, Division of Social Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
326
|
Stene LC, Rønningen KS, Undlien DE, Joner G. Does the relative risk for type 1 diabetes conferred by HLA-DQ, INS, and PTPN22 polymorphisms vary with maternal age, birth weight, or cesarean section? Pediatr Diabetes 2011; 12:91-4. [PMID: 21352425 DOI: 10.1111/j.1399-5448.2010.00669.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Maternal age at birth, birth weight, and cesarean section has been associated with a weak but significant increase in risk of type 1 diabetes. The objective was to assess whether the relative risk for type 1 diabetes conferred by established susceptibility loci human leukocyte antigen (HLA)-DQ, INS, and PTPN22 differed depending on these perinatal factors. METHODS We employed a case-control study with 456 cases of type 1 diabetes diagnosed before 15 yr of age and 1377 population-based control children. HLA genotypes were divided into high to moderate risk (DQ8/DQ2, DQ8/DQ8, DQ8/X, DQ2/DQ2) vs. all other genotypes. Case-only analysis using logistic regression was used to test for significant interaction. RESULTS There was no significant difference in the relative risks conferred by HLA-DQ, INS, or PTPN22 by maternal age, birth weight, or mode of delivery, except the relative risk conferred by PTPN22 which was 2.11 [95% confidence interval (CI): 1.64-2.72] for those born vaginally and 0.99 (95% CI: 0.50-1.99) for those born by cesarean section [p(interaction) = 0.028]. CONCLUSION The relative risks conferred by the three established susceptibility genes investigated here were independent of the perinatal factors, apart from a possible interaction between PTPN22 and mode of delivery.
Collapse
Affiliation(s)
- Lars C Stene
- Division of Epidemiology, Norwegian Institute of Public Health, Oslo, Norway.
| | | | | | | |
Collapse
|
327
|
Lau K, Benitez P, Ardissone A, Wilson TD, Collins EL, Lorca G, Li N, Sankar D, Wasserfall C, Neu J, Atkinson MA, Shatz D, Triplett EW, Larkin J. Inhibition of Type 1 Diabetes Correlated to a Lactobacillus johnsonii N6.2-Mediated Th17 Bias. THE JOURNAL OF IMMUNOLOGY 2011; 186:3538-46. [DOI: 10.4049/jimmunol.1001864] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
328
|
Vehik K, Dabelea D. The changing epidemiology of type 1 diabetes: why is it going through the roof? Diabetes Metab Res Rev 2011; 27:3-13. [PMID: 21218503 DOI: 10.1002/dmrr.1141] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/12/2010] [Accepted: 09/29/2010] [Indexed: 12/23/2022]
Abstract
The incidence of type 1 diabetes is increasing and this may double the burden of disease in our youngest by 2020. The recent increase in incidence is mostly happening in the very young and those with moderate genetic susceptibility. Many environmental factors have been implicated, but no major determinants have been clearly identified, and the mechanisms of involvement remain elusive. This review summarizes current research efforts directed at understanding the possible reasons for this increase, including the role of viruses, gut microbiota, early life feeding patterns, perinatal factors and childhood growth patterns. It also provides a road map for future research directions.
Collapse
Affiliation(s)
- Kendra Vehik
- Department of Pediatrics, College of Medicine, University of South Florida, Pediatrics Epidemiology Center, Tampa, FL 33612, USA.
| | | |
Collapse
|
329
|
Composto G, Gonzalez D, Bucknum A, Silberman D, Taylor J, Kozlowski M, Bloomfield T, Bartlett T, Riggs J. Peritoneal T lymphocyte regulation by macrophages. Immunobiology 2011; 216:256-64. [PMID: 20488579 PMCID: PMC2935942 DOI: 10.1016/j.imbio.2010.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Revised: 04/03/2010] [Accepted: 04/04/2010] [Indexed: 10/19/2022]
Abstract
The T cell composition of the peritoneal cavity (PerC) in naïve BALB/c, C57BL/6, DBA/2J, and B-1 B cell-defective BALB.xid mice was investigated. The BALB.xid PerC T cell pool had a high CD4:CD8 T cell ratio relative to the other strains whose ratios were similar to those found in their lymph node and spleen. All mice had significant representation of T cells with an activated (CD25(+), GITR(hi), CD44(hi), CD45RB(lo), CD62L(lo)) phenotype and low numbers of Foxp3(+) T(reg) cells in their PerC. Despite a phenotype indicative of activation, peritoneal T cell responses to CD3 ligation were very low for C57BL/6 and BALB.xid, but not BALB/c, mice. Enzyme inhibition and cytokine neutralization studies revealed active suppression of the T cell response mediated by the macrophages that represent a significant portion of PerC leucocytes. Driven by IFNγ to express iNOS, macrophages suppressed T cell activation in vitro by arginine catabolism. Although BALB/c T cells were also in a macrophage-dense environment their limited IFNγ production failed to trigger suppression. This difference between BALB/c and BALB.xid PerC T cells suggests a role for xid in shaping macrophage-mediated immune regulation.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Communication/immunology
- Cells, Cultured
- Forkhead Transcription Factors/biosynthesis
- Glucocorticoid-Induced TNFR-Related Protein
- Immune Tolerance
- Immunologic Memory
- Interleukin-2 Receptor alpha Subunit/biosynthesis
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Mice
- Mice, Inbred Strains
- Mice, Mutant Strains
- Receptors, Nerve Growth Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/biosynthesis
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- G Composto
- Department of Biology, Rider University, Lawrenceville, NJ 08648-3099, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Van Belle TL, Coppieters KT, Von Herrath MG. Type 1 Diabetes: Etiology, Immunology, and Therapeutic Strategies. Physiol Rev 2011; 91:79-118. [DOI: 10.1152/physrev.00003.2010] [Citation(s) in RCA: 673] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease in which destruction or damaging of the beta-cells in the islets of Langerhans results in insulin deficiency and hyperglycemia. We only know for sure that autoimmunity is the predominant effector mechanism of T1D, but may not be its primary cause. T1D precipitates in genetically susceptible individuals, very likely as a result of an environmental trigger. Current genetic data point towards the following genes as susceptibility genes: HLA, insulin, PTPN22, IL2Ra, and CTLA4. Epidemiological and other studies suggest a triggering role for enteroviruses, while other microorganisms might provide protection. Efficacious prevention of T1D will require detection of the earliest events in the process. So far, autoantibodies are most widely used as serum biomarker, but T-cell readouts and metabolome studies might strengthen and bring forward diagnosis. Current preventive clinical trials mostly focus on environmental triggers. Therapeutic trials test the efficacy of antigen-specific and antigen-nonspecific immune interventions, but also include restoration of the affected beta-cell mass by islet transplantation, neogenesis and regeneration, and combinations thereof. In this comprehensive review, we explain the genetic, environmental, and immunological data underlying the prevention and intervention strategies to constrain T1D.
Collapse
Affiliation(s)
- Tom L. Van Belle
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Ken T. Coppieters
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Matthias G. Von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| |
Collapse
|
331
|
Contribution of the intestinal microbiota to human health: from birth to 100 years of age. Curr Top Microbiol Immunol 2011; 358:323-46. [PMID: 22094893 DOI: 10.1007/82_2011_189] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Our intestinal tract is colonized since birth by multiple microbial species that show a characteristic succession in time. Notably the establishment of the microbiota in early life is important as it appears to impact later health. While apparently stable in healthy adults, the intestinal microbiota is changing significantly during aging. After 100 years of symbiosis marked changes have been observed that may relate to an increased level of intestinal inflammation. There is considerable interest in the microbiota in health and disease as it may provide functional biomarkers, the possibility to differentiate subjects, and avenues for interventions. This chapter reviews the present state of the art on the research to investigate the contribution of the intestinal microbiota to human health. Specific attention will be given to the healthy microbiota and aberrations due to disturbances such as celiac disease, irritable bowel syndrome, inflammatory bowel disease, obesity and diabetes, and non-alcoholic fatty liver disease.
Collapse
|
332
|
Godzien J, Ciborowski M, Angulo S, Ruperez FJ, Paz Martínez M, Señorans FJ, Cifuentes A, Ibañez E, Barbas C. Metabolomic Approach with LC-QTOF to Study the Effect of a Nutraceutical Treatment on Urine of Diabetic Rats. J Proteome Res 2010; 10:837-44. [DOI: 10.1021/pr100993x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Joanna Godzien
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Michal Ciborowski
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Santiago Angulo
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Francisco J. Ruperez
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - M Paz Martínez
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Francisco J. Señorans
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Alejandro Cifuentes
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Elena Ibañez
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| | - Coral Barbas
- CEMBIO (Center for Metabolomics and Bioanalysis) Pharmacy Faculty, Campus Monteprincipe, San Pablo-CEU University, 28668 Boadilla del Monte, Madrid, Spain, Department of Molecular Biology, Faculty of Mathematics and Natural Sciences, The John Paul II Catholic University of Lublin, Krasnicka 102, 20-718 Lublin, Poland, Department of Physical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland, Sección Departamental Ciencias de la Alimentación, Universidad Autónoma de Madrid
| |
Collapse
|
333
|
Anderson RC, Cookson AL, McNabb WC, Park Z, McCann MJ, Kelly WJ, Roy NC. Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol 2010; 10:316. [PMID: 21143932 PMCID: PMC3004893 DOI: 10.1186/1471-2180-10-316] [Citation(s) in RCA: 325] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 12/09/2010] [Indexed: 02/07/2023] Open
Abstract
Background Intestinal barrier function is important for preserving health, as a compromised barrier allows antigen entry and can induce inflammatory diseases. Probiotic bacteria can play a role in enhancing intestinal barrier function; however, the mechanisms are not fully understood. Existing studies have focused on the ability of probiotics to prevent alterations to tight junctions in disease models, and have been restricted to a few tight junction bridging proteins. No studies have previously investigated the effect of probiotic bacteria on healthy intestinal epithelial cell genes involved in the whole tight junction signalling pathway, including those encoding for bridging, plaque and dual location tight junction proteins. Alteration of tight junction signalling in healthy humans is a potential mechanism that could lead to the strengthening of the intestinal barrier, resulting in limiting the ability of antigens to enter the body and potentially triggering undesirable immune responses. Results The effect of Lactobacillus plantarum MB452 on tight junction integrity was determined by measuring trans-epithelial electrical resistance (TEER) across Caco-2 cell layers. L. plantarum MB452 caused a dose-dependent TEER increase across Caco-2 cell monolayers compared to control medium. Gene expression was compared in Caco-2 cells untreated or treated with L. plantarum MB452 for 10 hours. Caco-2 cell RNA was hybridised to human oligonucleotide arrays. Data was analysed using linear models and differently expressed genes were examined using pathway analysis tools. Nineteen tight junction-related genes had altered expression levels in response to L. plantarum MB452 (modified-P < 0.05, fold-change > 1.2), including those encoding occludin and its associated plaque proteins that anchor it to the cytoskeleton. L. plantarum MB452 also caused changes in tubulin and proteasome gene expression levels which may be linked to intestinal barrier function. Caco-2 tight junctions were visualised by fluorescent microscopy of immuno-stained occludin, zona occludens (ZO)-1, ZO-2 and cingulin. Caco-2 cells treated with L. plantarum MB452 had higher intensity fluorescence of each of the four tight junction proteins compared to untreated controls. Conclusions This research indicates that enhancing the expression of genes involved in tight junction signalling is a possible mechanism by which L. plantarum MB452 improves intestinal barrier function.
Collapse
Affiliation(s)
- Rachel C Anderson
- AgriFoods & Health Section, Food & Textiles Group, AgResearch Grasslands, Private Bag 11008, Palmerston North 4442, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
334
|
Visser JTJ, Lammers K, Hoogendijk A, Boer MW, Brugman S, Beijer-Liefers S, Zandvoort A, Harmsen H, Welling G, Stellaard F, Bos NA, Fasano A, Rozing J. Restoration of impaired intestinal barrier function by the hydrolysed casein diet contributes to the prevention of type 1 diabetes in the diabetes-prone BioBreeding rat. Diabetologia 2010; 53:2621-8. [PMID: 20853098 PMCID: PMC2974912 DOI: 10.1007/s00125-010-1903-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 07/23/2010] [Indexed: 01/19/2023]
Abstract
AIMS/HYPOTHESIS Impaired intestinal barrier function is observed in type 1 diabetes patients and animal models of the disease. Exposure to diabetogenic antigens from the intestinal milieu due to a compromised intestinal barrier is considered essential for induction of the autoimmune process leading to type 1 diabetes. Since a hydrolysed casein (HC) diet prevents autoimmune diabetes onset in diabetes-prone (DP)-BioBreeding (BB) rats, we studied the role of the HC diet on intestinal barrier function and, therefore, prevention of autoimmune diabetes onset in this animal model. METHODS DP-BB rats were fed the HC diet from weaning onwards and monitored for autoimmune diabetes development. Intestinal permeability was assessed in vivo by lactulose-mannitol test and ex vivo by measuring transepithelial electrical resistance (TEER). Levels of serum zonulin, a physiological tight junction modulator, were measured by ELISA. Ileal mRNA expression of Myo9b, Cldn1, Cldn2 and Ocln (which encode the tight junction-related proteins myosin IXb, claudin-1, claudin-2 and occludin) and Il-10, Tgf-ß (also known as Il10 and Tgfb, respectively, which encode regulatory cytokines) was analysed by quantitative PCR. RESULTS The HC diet reduced autoimmune diabetes by 50% in DP-BB rats. In DP-BB rats, prediabetic gut permeability negatively correlated with the moment of autoimmune diabetes onset. The improved intestinal barrier function that was induced by HC diet in DP-BB rats was visualised by decreasing lactulose:mannitol ratio, decreasing serum zonulin levels and increasing ileal TEER. The HC diet modified ileal mRNA expression of Myo9b, and Cldn1 and Cldn2, but left Ocln expression unaltered. CONCLUSIONS/INTERPRETATION Improved intestinal barrier function might be an important intermediate in the prevention of autoimmune diabetes by the HC diet in DP-BB rats. Effects on tight junctions, ileal cytokines and zonulin production might be important mechanisms for this effect.
Collapse
Affiliation(s)
- J T J Visser
- Department of Cell Biology, Section Immunology, University Medical Center Groningen, University of Groningen, PO Box 196, 9700 AD Groningen, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
335
|
Hufeldt MR, Nielsen DS, Vogensen FK, Midtvedt T, Hansen AK. Variation in the gut microbiota of laboratory mice is related to both genetic and environmental factors. Comp Med 2010; 60:336-347. [PMID: 21262117 PMCID: PMC2958200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 01/16/2010] [Accepted: 03/21/2010] [Indexed: 05/30/2023]
Abstract
During recent years, the composition of the gut microbiota (GM) has received increasing attention as a factor in the development of experimental inflammatory disease in animal models. Because increased variation in the GM might lead to increased variation in disease parameters, determining and reducing GM variation between laboratory animals may provide more consistent models. Both genetic and environmental aspects influence the composition of the GM and may vary between laboratory animal breeding centers and within an individual breeding center. This study investigated the variation in cecal microbiota in 8-wk-old NMRI and C57BL/6 mice by using denaturing gradient gel electrophoresis to profile PCR-derived amplicons from bacterial 16S rRNA genes. Comparison of the cecal microbiotas revealed that the similarity index of the inbred C57BL/6Sca strain was 10% higher than that of the outbred Sca:NMRI stock. Comparing C57BL/6 mice from 2 vendors revealed significant differences in the microbial profile, whereas the profiles of C57BL/6Sca mice raised in separate rooms within the same breeding center were not significantly different. Furthermore, housing in individually ventilated cages did not lead to intercage variation. These results show that denaturing gradient gel electrophoresis is a simple tool that can be used to characterize the gut microbiota of mice. Including such characterizations in future quality-control programs may increase the reproducibility of mouse studies.
Collapse
Affiliation(s)
- Majbritt Ravn Hufeldt
- Centre for Applied Laboratory Animal Research, Department of Veterinary Disease Biology
- Department of Food Science, Faculty of Life Sciences, University of Copenhagen, Frederiksberg, Denmark
- Centre for Applied Laboratory Animal Research, Scanbur A/S, Karlslunde, Denmark
| | - Dennis S Nielsen
- Department of Food Science, Faculty of Life Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Finn Kvist Vogensen
- Department of Food Science, Faculty of Life Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Tore Midtvedt
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Axel Kornerup Hansen
- Centre for Applied Laboratory Animal Research, Department of Veterinary Disease Biology
| |
Collapse
|
336
|
Kaldunski M, Jia S, Geoffrey R, Basken J, Prosser S, Kansra S, Mordes JP, Lernmark Å, Wang X, Hessner MJ. Identification of a serum-induced transcriptional signature associated with type 1 diabetes in the BioBreeding rat. Diabetes 2010; 59:2375-85. [PMID: 20682698 PMCID: PMC3279523 DOI: 10.2337/db10-0372] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Inflammatory mediators associated with type 1 diabetes are dilute and difficult to measure in the periphery, necessitating development of more sensitive and informative biomarkers for studying diabetogenic mechanisms, assessing preonset risk, and monitoring therapeutic interventions. RESEARCH DESIGN AND METHODS We previously utilized a novel bioassay in which human type 1 diabetes sera were used to induce a disease-specific transcriptional signature in unrelated, healthy peripheral blood mononuclear cells (PBMCs). Here, we apply this strategy to investigate the inflammatory state associated with type 1 diabetes in biobreeding (BB) rats. RESULTS Consistent with their common susceptibility, sera of both spontaneously diabetic BB DRlyp/lyp and diabetes inducible BB DR+/+ rats induced transcription of cytokines, immune receptors, and signaling molecules in PBMCs of healthy donor rats compared with control sera. Like the human type 1 diabetes signature, the DRlyp/lyp signature, which is associated with progression to diabetes, was differentiated from that of the DR+/+ by induction of many interleukin (IL)-1-regulated genes. Supplementing cultures with an IL-1 receptor antagonist (IL-1Ra) modulated the DRlyp/lyp signature (P < 10(-6)), while administration of IL-1Ra to DRlyp/lyp rats delayed onset (P = 0.007), and sera of treated animals did not induce the characteristic signature. Consistent with the presence of immunoregulatory cells in DR+/+ rats was induction of a signature possessing negative regulators of transcription and inflammation. CONCLUSIONS Paralleling our human studies, serum signatures in BB rats reflect processes associated with progression to type 1 diabetes. Furthermore, these studies support the potential utility of this approach to detect changes in the inflammatory state during therapeutic intervention.
Collapse
Affiliation(s)
- Mary Kaldunski
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
| | - Shuang Jia
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
| | - Rhonda Geoffrey
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
| | - Joel Basken
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
| | - Simon Prosser
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
| | - Sanjay Kansra
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
| | - John P. Mordes
- Division of Endocrinology and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Åke Lernmark
- Robert H. Williams Laboratory, Department of Medicine, University of Washington, Seattle, Washington
| | - Xujing Wang
- Department of Physics and the Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Martin J. Hessner
- Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin, the Children's Research Institute of Children's Hospital of Wisconsin, and the Human and Molecular Genetics Center, Milwaukee, Wisconsin
- Corresponding author: Martin J. Hessner,
| |
Collapse
|
337
|
Abstract
Type 1 diabetes is a chronic autoimmune disease characterized by a preclinical period of autoimmunity. It is well accepted that both genetic and environmental factors contribute to disease risk. Given that type 1 diabetes, and its preclinical autoimmunity, appear early in life, infant and childhood diet have been implicated as potential initiating exposures in the etiology of the disease. Several publications in the past year have provided further evidence for existing hypotheses regarding the roles of wheat, cow's milk, omega-3 fatty acids, and the maternal diet during pregnancy. However, inconsistencies in findings between studies suggest the need for collaboration and standardization of study methods to move forward in research in this area. One such example of this is the TEDDY (The Environmental Determinants of Diabetes in the Young) study, which is an international, multicenter birth cohort study with standardized recruitment, dietary collection methodologies, and analytic approaches.
Collapse
Affiliation(s)
- Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, 13001 East 17th Place, Aurora, CO 80045, USA.
| |
Collapse
|
338
|
Alam C, Valkonen S, Palagani V, Jalava J, Eerola E, Hänninen A. Inflammatory tendencies and overproduction of IL-17 in the colon of young NOD mice are counteracted with diet change. Diabetes 2010; 59:2237-46. [PMID: 20547977 PMCID: PMC2927946 DOI: 10.2337/db10-0147] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Dietary factors influence diabetes development in the NOD mouse. Diet affects the composition of microbiota in the distal intestine, which may subsequently influence intestinal immune homeostasis. However, the specific effects of antidiabetogenic diets on gut immunity and the explicit associations between intestinal immune disruption and type 1 diabetes onset remain unclear. RESEARCH DESIGN AND METHODS Gut microbiota of NOD mice fed a conventional diet or ProSobee formula were compared using gas chromatography. Colonic lamina propria immune cells were characterized in terms of activation markers, cytokine mRNA and Th17 and Foxp3(+) T-cell numbers, using real-time PCR and flow cytometry. Activation of diabetogenic CD4 T-cells by purified B-cells was assessed in both groups. Immune tolerance to autologous commensal bacteria was evaluated in vitro using thymidine-incorporation tests. RESULTS Young NOD mice showed a disturbed tolerance to autologous commensal bacteria. Increased numbers of activated CD4 T-cells and (CD11b(+)CD11c(+)) dendritic cells and elevated levels of Th17 cells and IL23 mRNA were moreover observed in colon lamina propria. These phenomena were abolished when mice were fed an antidiabetogenic diet. The antidiabetogenic diet also altered the expression levels of costimulatory molecules and the capacity of peritoneal B-cells to induce insulin-specific CD4 T-cell proliferation. CONCLUSIONS Young NOD mice show signs of subclinical colitis, but the symptoms are alleviated by a diet change to an antidiabetogenic diet. Disrupted immune tolerance in the distal intestine may influence peritoneal cell pools and B-cell-mediated activation of diabetogenic T-cells.
Collapse
Affiliation(s)
- Catharina Alam
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Suvi Valkonen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Vindhya Palagani
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Jari Jalava
- Antimicrobial Research Laboratory, National Institute for Health and Welfare (THL), Turku, Finland
| | - Erkki Eerola
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Arno Hänninen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
- Corresponding author: Arno Hänninen,
| |
Collapse
|
339
|
Neu J, Lorca G, Kingma SDK, Triplett EW. The intestinal microbiome: relationship to type 1 diabetes. Endocrinol Metab Clin North Am 2010; 39:563-71. [PMID: 20723820 DOI: 10.1016/j.ecl.2010.05.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This article discusses recent evidence that associates the developing intestinal microbiome to the pathogenesis of autoimmune T1D. It attempts to identify avenues that should be pursued that relate this new evidence to interventions that eventually could result in prevention.
Collapse
Affiliation(s)
- Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL 326101, USA.
| | | | | | | |
Collapse
|
340
|
Hufeldt MR, Nielsen DS, Vogensen FK, Midtvedt T, Hansen AK. Family relationship of female breeders reduce the systematic inter-individual variation in the gut microbiota of inbred laboratory mice. Lab Anim 2010; 44:283-9. [PMID: 20713427 DOI: 10.1258/la.2010.010058] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The gut microbiota (GM) may influence disease expression in several animal models for inflammatory diseases. It may therefore seem reasonable to pursue reduction in the number of animals used for individual studies by reducing the variation in the GM. Previous studies have shown that the composition of the GM is related to genetics to a certain extent. We hypothesized that the GM similarity in a group of mice born by mothers not being sisters would be lower than that in a group born by mothers being sisters. The lower similarity could lead to clustering of the GM of mice born by non-sisters according to their mothers, while such clustering would not be visible if the mothers were sisters. We used 16S rRNA gene (V3 region) polymerase chain reaction-derived amplicon profiling by denaturing gradient gel electrophoresis (DGGE) to study the GM composition in caecum samples of 33 eight-week-old C57BL/6Sca mice from a breeding set-up with dam breeders that were sisters, as well as caecum samples of 35 eight-week-old C57BL/6Sca mice from a breeding set-up with dam breeders that were not sisters. Principal component analysis revealed a significant difference between the litters from the breeding set-up with dam breeders that were not sisters, whereas no significant difference between the litters based on the breeding set-up with dam breeders that were sisters was observed. The results obtained indicate that the systematic variation in the GM of inbred mice can be reduced by increasing the family relatedness of the breeding pairs.
Collapse
Affiliation(s)
- M R Hufeldt
- Department of Veterinary Disease Biology, Faculty of Life Sciences, Centre for Applied Laboratory Animal Research, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | | | | | | | | |
Collapse
|
341
|
Abstract
BACKGROUND Recombinant vaccinia virus (rVV) strains expressing the immunomodulatory cholera toxin B subunit (CTB) fused to the autoantigen glutamic acid decarboxylase (GAD) or the immunosuppressive cytokine interleukin-10 (IL-10) were independently able to generate only low levels of immune suppression of type 1 diabetes mellitus (T1DM). Here we suggest that a vaccinia virus (VV)-mediated combination of CTB::GAD fusion and IL-10 proteins promises a effective and durable immunotherapeutic strategy for T1DM. METHODS To explore this hypothesis, a CTB::GAD fusion gene was co-delivered with a gene encoding IL-10 by rVV infection (rVV-CTB::GAD + rVV-IL10) into 5-7-week-old non-obese diabetic (NOD) mice. The mice were assessed for vaccine protection against development of hyperglycemia from 12 to 64 weeks of age by assessment of pancreatic inflammation (insulitis) and splenocyte-secreted interferon-gamma and IL-10 cytokine levels. RESULTS By 36 weeks of age, from 54% to 80% of the mice in the negative control animal groups (either mock-infected or inoculated with unrelated plasmid or VV) had developed hyperglycemia. Similarly, no statistically significant improvement in protection against diabetes onset was achieved by inoculation with VV expressing CTB::GAD or IL-10 independently. Surprisingly, only 20% of mice co-inoculated with rVV-CTB::GAD + rVV-IL10 developed hyperglycemia by 28 weeks of age. Other treatment groups developed hyperglycemia by 32-36 weeks. After 36 weeks, diabetes incidence no longer increased in any groups until the end of experiment at 64 weeks of age. Histological analysis of pancreatic tissues of hyperglycemic mice revealed high levels of intra-islet insulitis. Analysis of insulitis at termination of the experiment showed that euglycemic mice co-inoculated with VV expressing CTB::GAD and IL-10 had more effectively reduced inflammation in comparison with the other groups. CONCLUSIONS A combinatorial vaccination strategy based on VV co-delivery of genes encoding the immunoenhanced autoantigen CTB::GAD and the anti-inflammatory cytokine IL-10 can maintain effective and durable euglycemia and immunological homeostasis in NOD mice with prediabetes.
Collapse
Affiliation(s)
- Béla Dénes
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
- Department of Immunology, Central Veterinary Institute, Budapest, Hungary
| | - István Fodor
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
| | - William H.R. Langridge
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
| |
Collapse
|
342
|
Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyöty H, Veijola R, Simell T, Simell O, Neu J, Wasserfall CH, Schatz D, Atkinson MA, Triplett EW. Toward defining the autoimmune microbiome for type 1 diabetes. ISME JOURNAL 2010; 5:82-91. [PMID: 20613793 DOI: 10.1038/ismej.2010.92] [Citation(s) in RCA: 600] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Several studies have shown that gut bacteria have a role in diabetes in murine models. Specific bacteria have been correlated with the onset of diabetes in a rat model. However, it is unknown whether human intestinal microbes have a role in the development of autoimmunity that often leads to type 1 diabetes (T1D), an autoimmune disorder in which insulin-secreting pancreatic islet cells are destroyed. High-throughput, culture-independent approaches identified bacteria that correlate with the development of T1D-associated autoimmunity in young children who are at high genetic risk for this disorder. The level of bacterial diversity diminishes overtime in these autoimmune subjects relative to that of age-matched, genotype-matched, nonautoimmune individuals. A single species, Bacteroides ovatus, comprised nearly 24% of the total increase in the phylum Bacteroidetes in cases compared with controls. Conversely, another species in controls, represented by the human firmicute strain CO19, represented nearly 20% of the increase in Firmicutes compared with cases overtime. Three lines of evidence are presented that support the notion that, as healthy infants approach the toddler stage, their microbiomes become healthier and more stable, whereas, children who are destined for autoimmunity develop a microbiome that is less diverse and stable. Hence, the autoimmune microbiome for T1D may be distinctly different from that found in healthy children. These data also suggest bacterial markers for the early diagnosis of T1D. In addition, bacteria that negatively correlated with the autoimmune state may prove to be useful in the prevention of autoimmunity development in high-risk children.
Collapse
Affiliation(s)
- Adriana Giongo
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611-0700, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Kim SJ, Nian C, McIntosh CH. Sitagliptin (MK0431) inhibition of dipeptidyl peptidase IV decreases nonobese diabetic mouse CD4+ T-cell migration through incretin-dependent and -independent pathways. Diabetes 2010; 59:1739-50. [PMID: 20368408 PMCID: PMC2889774 DOI: 10.2337/db09-1618] [Citation(s) in RCA: 41] [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] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Treatment of NOD mice with the dipeptidyl peptidase-IV (DPP-IV) inhibitor sitagliptin preserved islet transplants through a pathway involving modulation of splenic CD4(+) T-cell migration. In the current study, effects of sitagliptin on migration of additional subsets of CD4(+) T-cells were examined and underlying molecular mechanisms were further defined. RESEARCH DESIGN AND METHODS Effects of sitagliptin on migration of NOD mouse splenic, thymic, and lymph node CD4(+) T-cells were determined. Signaling modules involved in DPP-IV-, Sitagliptin- and incretin-mediated modulation of CD4(+) T-cell migration were studied using Western blot and Rac1 and nuclear factor-kappaB (NF-kappaB) activity assays. RESULTS Migration of splenic and lymph node CD4(+) T-cells of diabetic NOD mice was reduced by sitagliptin treatment. In vitro treatment of splenic, but not thymic or lymph node CD4(+) T-cells, from nondiabetic NOD mice with soluble (s) DPP-IV increased migration. Sitagliptin abolished sDPP-IV effects on splenic CD4(+) T-cell migration, whereas incretins decreased migration of lymph node, but not splenic, CD4(+) T-cells. Splenic CD4(+) T-cells demonstrating increased in vitro migration in response to sDPP-IV and lymph node CD4(+) T-cells that were nonresponsive to incretins selectively infiltrated islets of NOD mice, after injection. Sitagliptin decreases migration of splenic CD4(+) T-cells through a pathway involving Rac1/vasodilator-stimulated phosphoprotein, whereas its inhibitory effects on the migration of lymph node CD4(+) T-cells involve incretin-activation of the NF-kappaB pathway. CONCLUSIONS Benefits of sitagliptin treatment in diabetic NOD mice may be mediated through selective effects on subpopulations of T-cells that are related to autoimmunity.
Collapse
Affiliation(s)
- Su-Jin Kim
- From the Department of Cellular and Physiological Sciences and the Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cuilan Nian
- From the Department of Cellular and Physiological Sciences and the Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher H.S. McIntosh
- From the Department of Cellular and Physiological Sciences and the Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Corresponding author: C.H.S. McIntosh,
| |
Collapse
|
344
|
Sud S, Marcon M, Assor E, Palmert MR, Daneman D, Mahmud FH. Celiac disease and pediatric type 1 diabetes: diagnostic and treatment dilemmas. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2010; 2010:161285. [PMID: 20652072 PMCID: PMC2905696 DOI: 10.1155/2010/161285] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 04/01/2010] [Indexed: 02/08/2023]
Abstract
Despite the advent of sensitive and specific serologic testing, routine screening for celiac disease (CD) in diabetic populations may not be universal practice, and many clinicians struggle to find the optimal approach to managing CD in pediatric Type 1 diabetes (T1D) patients. While some clinicians advocate screening for CD in all patients with T1D, others are unsure whether this is warranted. The diagnosis of patients who present with symptomatic CD, including malabsorption and obvious pathology upon biopsy, remains straightforward, with improvements noted on a gluten-free diet. Many patients identified by screening, however, tend to be asymptomatic. Evidence is inconclusive as to whether the benefits of screening and potentially treating asymptomatic individuals outweigh the harms of managing a population already burdened with a serious illness. This review focuses on current knowledge of CD in children and youth with T1D, highlighting important elements of the disease's pathophysiology, epidemiology, clinical presentation, and diagnostic challenges.
Collapse
Affiliation(s)
- Shama Sud
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Margaret Marcon
- Division of Gastroenterology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Esther Assor
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Mark R. Palmert
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Denis Daneman
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Farid H. Mahmud
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| |
Collapse
|
345
|
Brugman S, Nieuwenhuis EES. Mucosal control of the intestinal microbial community. J Mol Med (Berl) 2010; 88:881-8. [PMID: 20523962 DOI: 10.1007/s00109-010-0639-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 05/18/2010] [Accepted: 05/25/2010] [Indexed: 12/11/2022]
Abstract
Although the knowledge of the effects of bacterial colonization on the immune system is rapidly expanding, surprisingly little is known about the immunological mechanisms that shape the intestinal microbial community. Specifically, the complexity of the intestinal microbiota and what constitutes a "healthy" microbial composition has only recently been addressed, facilitated by large-scale metagenomic screens. Containment of such a vast number of different microbes requires tight regulation at the mucosal surface. While beneficial relationships must not be compromised, invading pathogenic bacteria must be dealt with in order to maintain homeostasis. In this review, we will address the latest insights into the role of the mucosal immune system in the control of the microbiota.
Collapse
Affiliation(s)
- Sylvia Brugman
- Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | | |
Collapse
|
346
|
Prangli AL, Utt M, Talja I, Sepp E, Mikelsaar M, Rajasalu T, Uibo O, Tillmann V, Uibo R. Antigenic proteins of Lactobacillus acidophilus that are recognised by serum IgG antibodies in children with type 1 diabetes and coeliac disease. Pediatr Allergy Immunol 2010; 21:e772-9. [PMID: 19573144 DOI: 10.1111/j.1399-3038.2009.00904.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Immune responses to lactobacilli have been so far insufficiently investigated in patients with autoimmune diseases. We used whole-cell lysate of an indigenous Lactobacillus acidophilus strain isolated from an Estonian child to study serum IgG antibodies in children groups with type 1 diabetes [insulin dependent diabetes mellitus (IDDM)] (n = 21, age 4-18 yr) and with acute coeliac disease (CD) (n = 20, age 0.6-15 yr) and to compare the results with the controls (n = 24, age 2-17 yr). We found that our developed 1-D immunoblot assay readily enables to reveal antibodies against 28 L. acidophilus antigenic proteins in patients' and controls' sera. As verified by immunoproteomics analysis with 2-D and LC ESI-MS/MS the antigens of L. acidophilus were mainly common cytoplasmic proteins GroEL (HSP60), enolase, transcription factor EF-Ts and EF-Tu. However, in addition we identified formyl-CoA transferase being target for antibodies in every tested IDDM patients' serum. We have characterized for the first time the antigenic profile of L. acidophilus whole-cell lysate using sera from children with IDDM, CD, and controls. The different prevalence of reactions against tested antigens in patients and controls sera may indicate significant differences in immune system and commensal bacteria cross-talk in these groups.
Collapse
|
347
|
Association analysis of myosin IXB and type 1 diabetes. Hum Immunol 2010; 71:598-601. [DOI: 10.1016/j.humimm.2010.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 03/07/2010] [Accepted: 03/10/2010] [Indexed: 11/21/2022]
|
348
|
Sakly W, Mankaï A, Sakly N, Thabet Y, Achour A, Ghedira-Besbes L, Jeddi M, Ghedira I. Anti-Saccharomyces cerevisiae antibodies are frequent in type 1 diabetes. Endocr Pathol 2010; 21:108-14. [PMID: 20387011 DOI: 10.1007/s12022-010-9118-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Anti-Saccharomyces cerevisiae antibodies (ASCA) have been described in many autoimmune diseases in which there is an increased intestinal permeability. Also in type 1 diabetes (T1D), there is an increased intestinal permeability. Since no data are available about ASCA in T1D, we evaluated, retrospectively, the frequency of ASCA in this disease. ASCA, IgG, and IgA, were determined by ELISA in sera of 224 T1D patients in which coeliac disease has been excluded and 157 healthy control group. The frequency of ASCA (IgG or IgA) was significantly higher in T1D patients than in the control group (24.5% vs. 2.5%, p < 10(-7)). The same observation was found in children and in adult patients when we compare them to healthy children and blood donors group respectively. Compared to children, adult patients with T1D showed significantly higher frequencies of ASCA of any isotype (38% vs. 13.7%, p < 10(-4)), both ASCA IgG and IgA (12% vs. 1.6%, p = 0.002), ASCA IgG (35% vs. 9.8%, p < 10(-5)) and ASCA IgA (15% vs. 5.6%, p = 0.001). The frequency of ASCA was statistically higher in females of all T1D than in males (30.8% vs.17.7%, p = 0.03), in girls than in boys (22% vs.6.2%, p = 0.017), and significantly higher in men than in boys (35.7% vs. 6.2%, p < 10(-4)). The frequency of ASCA IgG was significantly higher than that of ASCA IgA in all T1D patients (21% vs. 9.8%, p < 0.002), in all females (26.5% vs. 10.2%, p < 0.002), in women (37.9% vs. 12%, p < 0.001). The frequency of ASCA was significantly higher in all long-term T1D than in an inaugural T1D (29% vs. 14.5%, p = 0.019). The same observation was found in adults (45.8% vs. 17.8%, p = 0.01). In long-term T1D patients, ASCA were significantly more frequent in adults than children (45.8% vs. 14.5%, p < 10(-4)). The frequency of ASCA IgG was significantly higher in long-term T1D than in an inaugural T1D (25.2% vs. 11.6%, p = 0.03). Patients with T1D had a high frequency of ASCA.
Collapse
Affiliation(s)
- Wahiba Sakly
- Research unit (03UR/07-02), Faculty of Pharmacy, Rue Avicenne, 5000, Monastir, Tunisia
| | | | | | | | | | | | | | | |
Collapse
|
349
|
Valladares R, Sankar D, Li N, Williams E, Lai KK, Abdelgeliel AS, Gonzalez CF, Wasserfall CH, Larkin J, Schatz D, Atkinson MA, Triplett EW, Neu J, Lorca GL. Lactobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats. PLoS One 2010; 5:e10507. [PMID: 20463897 PMCID: PMC2865539 DOI: 10.1371/journal.pone.0010507] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 04/12/2010] [Indexed: 11/24/2022] Open
Abstract
Background The intestinal epithelium is a barrier that composes one of the most immunologically active surfaces of the body due to constant exposure to microorganisms as well as an infinite diversity of food antigens. Disruption of intestinal barrier function and aberrant mucosal immune activation have been implicated in a variety of diseases within and outside of the gastrointestinal tract. With this model in mind, recent studies have shown a link between diet, composition of intestinal microbiota, and type 1 diabetes pathogenesis. In the BioBreeding rat model of type 1 diabetes, comparison of the intestinal microbial composition of diabetes prone and diabetes resistant animals found Lactobacillus species were negatively correlated with type 1 diabetes development. Two species, Lactobacillus johnsonii and L. reuteri, were isolated from diabetes resistant rats. In this study diabetes prone rats were administered pure cultures of L. johnsonii or L. reuteri isolated from diabetes resistant rats to determine the effect on type 1 diabetes development. Methodology/Principal Findings Results Rats administered L. johnsonii, but not L. reuteri, post-weaning developed type 1 diabetes at a protracted rate. Analysis of the intestinal ileum showed administration of L. johnsonii induced changes in the native microbiota, host mucosal proteins, and host oxidative stress response. A decreased oxidative intestinal environment was evidenced by decreased expression of several oxidative response proteins in the intestinal mucosa (Gpx1, GR, Cat). In L. johnsonii fed animals low levels of the pro-inflammatory cytokine IFNγ were correlated with low levels of iNOS and high levels of Cox2. The administration of L. johnsonii also resulted in higher levels of the tight junction protein claudin. Conclusions It was determined that the administration of L. johnsonii isolated from BioBreeding diabetes resistant rats delays or inhibits the onset of type 1 diabetes in BioBreeding diabetes prone rats. Taken collectively, these data suggest that the gut and the gut microbiota are potential agents of influence in type 1 diabetes development. These data also support therapeutic efforts that seek to modify gut microbiota as a means to modulate development of this disorder.
Collapse
Affiliation(s)
- Ricardo Valladares
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Dhyana Sankar
- Department of Pediatrics, University of Florida, Gainesville, Florida, United States of America
| | - Nan Li
- Department of Pediatrics, University of Florida, Gainesville, Florida, United States of America
| | - Emily Williams
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Kin-Kwan Lai
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Asmaa Sayed Abdelgeliel
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Claudio F. Gonzalez
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Desmond Schatz
- Department of Pediatrics, University of Florida, Gainesville, Florida, United States of America
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Eric W. Triplett
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, Florida, United States of America
| | - Graciela L. Lorca
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
350
|
Lee AS, Gibson DL, Zhang Y, Sham HP, Vallance BA, Dutz JP. Gut barrier disruption by an enteric bacterial pathogen accelerates insulitis in NOD mice. Diabetologia 2010; 53:741-8. [PMID: 20012858 DOI: 10.1007/s00125-009-1626-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Accepted: 11/06/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS Increased exposure to enteric microbes as a result of intestinal barrier disruption is thought to contribute to the development of several intestinal inflammatory diseases; however, it less clear whether such exposure modulates the development of extra-intestinal inflammatory and autoimmune diseases. The goal of this study was to examine the potential role of pathogenic enteric microbes and intestinal barrier dysfunction in the pathogenesis of type 1 diabetes. METHODS Using NOD mice, we assessed: (1) intrinsic barrier function in mice at different ages by measuring serum levels of FITC-labelled dextran; and (2) the impact on insulitis development of infection by strains of an enteric bacterial pathogen (Citrobacter rodentium) either capable (wild-type) or incapable (lacking Escherichia coli secreted protein F virulence factor owing to deletion of the gene [DeltaespF]) of causing intestinal epithelial barrier disruption. RESULTS Here we demonstrate that prediabetic (12-week-old) NOD mice display increased intestinal permeability compared with non-obese diabetes-resistant and C57BL/6 mice. We also found that young (4-week-old) NOD mice infected with wild-type C. rodentium exhibited accelerated development of insulitis in concert with infection-induced barrier disruption. In contrast, insulitis development was not altered in NOD mice infected with the non-barrier-disrupting DeltaespF strain. Moreover, C. rodentium-infected NOD mice demonstrated increased activation and proliferation of pancreatic-draining lymph node T cells, including diabetogenic CD8(+) T cells, compared with uninfected NOD mice. CONCLUSIONS/INTERPRETATION This is the first demonstration that a loss of intestinal barrier integrity caused by an enteric bacterial pathogen results in the activation of diabetogenic CD8(+) T cells and modulates insulitis.
Collapse
Affiliation(s)
- A S Lee
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | |
Collapse
|