Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
PNAS Nexus ; 3(3): pgae096, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38528952

ABSTRACT

Dysfunctional liver regeneration following surgical resection remains a major cause of postoperative mortality and has no therapeutic options. Without targeted therapies, the current treatment paradigm relies on supportive therapy until homeostasis can be achieved. Pharmacologic acceleration of regeneration represents an alternative therapeutic avenue. Therefore, we aimed to generate a small molecule inhibitor that could accelerate liver regeneration with an emphasis on diseased models, which represent a significant portion of patients who require surgical resection and are often not studied. Utilizing a clinically approved small molecule inhibitor as a parent compound, standard medicinal chemistry approaches were utilized to generate a small molecule inhibitor targeting serine/threonine kinase 4/3 (MST1/2) with reduced off-target effects. This compound, mCLC846, was then applied to preclinical models of murine partial hepatectomy, which included models of diet-induced metabolic dysfunction-associated steatohepatitis (MASH). mCLC846 demonstrated on target inhibition of MST1/2 and reduced epidermal growth factor receptor inhibition. The inhibitory effects resulted in restored pancreatic beta-cell function and survival under diabetogenic conditions. Liver-specific cell-line exposure resulted in Yes-associated protein activation. Oral delivery of mCLC846 perioperatively resulted in accelerated murine liver regeneration and improved survival in diet-induced MASH models. Bulk transcriptional analysis of regenerating liver remnants suggested that mCLC846 enhanced the normal regenerative pathways and induced them following liver resection. Overall, pharmacological acceleration of liver regeneration with mCLC846 was feasible, had an acceptable therapeutic index, and provided a survival benefit in models of diet-induced MASH.

2.
Redox Biol ; 69: 102994, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38128451

ABSTRACT

Progression of ß-cell loss in diabetes mellitus is significantly influenced by persistent hyperglycemia. At the cellular level, a number of signaling cascades affect the expression of apoptotic genes, ultimately resulting in ß-cell failure; these cascades have not been elucidated. Mitochondrial aldehyde dehydrogenase-2 (ALDH2) plays a central role in the detoxification of reactive aldehydes generated from endogenous and exogenous sources and protects against mitochondrial deterioration in cells. Here we report that under diabetogenic conditions, ALDH2 is strongly inactivated in ß-cells through CDK5-dependent glutathione antioxidant imbalance by glucose-6-phosphate dehydrogenase (G6PD) degradation. Intriguingly, CDK5 inhibition strengthens mitochondrial antioxidant defense through ALDH2 activation. Mitochondrial ALDH2 activation selectively preserves ß-cells against high-glucose-induced dysfunction by activating AMPK and Hydrogen Sulfide (H2S) signaling. This is associated with the stabilization and enhancement of the activity of G6PD by SIRT2, a cytoplasmic NAD+-dependent deacetylase, and is thereby linked to an elevation in the GSH/GSSG ratio, which leads to the inhibition of mitochondrial dysfunction under high-glucose conditions. Furthermore, treatment with NaHS, an H2S donor, selectively preserves ß-cell function by promoting ALDH2 activity, leading to the inhibition of lipid peroxidation by high-glucose concentrations. Collectively, our results provide the first direct evidence that ALDH2 activation enhances H2S-AMPK-G6PD signaling, leading to improved ß-cell function and survival under high-glucose conditions via the glutathione redox balance.


Subject(s)
Hydrogen Sulfide , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Hydrogen Sulfide/pharmacology , Antioxidants/pharmacology , Aldehyde Dehydrogenase/genetics , AMP-Activated Protein Kinases/metabolism , Glutathione/metabolism , Glucose/metabolism
3.
Front Endocrinol (Lausanne) ; 14: 1236574, 2023.
Article in English | MEDLINE | ID: mdl-38027145

ABSTRACT

Introduction: For more than a century, enteroviral infections have been associated with autoimmunity and type 1 diabetes (T1D). Uncontrolled viral response pathways repeatedly presented during childhood highly correlate with autoimmunity and T1D. Virus responses evoke chemokines and cytokines, the "cytokine storm" circulating through the body and attack cells especially vulnerable to inflammatory destruction. Intra-islet inflammation is a major trigger of ß-cell failure in both T1D and T2D. The genetic contribution of islet inflammation pathways is apparent in T1D, with several mutations in the interferon system. In contrast, in T2D, gene mutations are related to glucose homeostasis in ß cells and insulin-target tissue and rarely within viral response pathways. Therefore, the current study evaluated whether enteroviral RNA can be found in the pancreas from organ donors with T2D and its association with disease progression. Methods: Pancreases from well-characterized 29 organ donors with T2D and 15 age- and BMI-matched controls were obtained from the network for pancreatic organ donors with diabetes and were analyzed in duplicates. Single-molecule fluorescence in-situ hybridization analyses were performed using three probe sets to detect positive-strand enteroviral RNA; pancreas sections were co-stained by classical immunostaining for insulin and CD45. Results: There was no difference in the presence or localization of enteroviral RNA in control nondiabetic and T2D pancreases; viral infiltration showed large heterogeneity in both groups ranging from 0 to 94 virus+ cells scattered throughout the pancreas, most of them in the exocrine pancreas. Very rarely, a single virus+ cell was found within islets or co-stained with CD45+ immune cells. Only one single T2D donor presented an exceptionally high number of viruses, similarly as seen previously in T1D, which correlated with a highly reduced number of ß cells. Discussion: No association of enteroviral infection in the pancreas and T2D diabetes could be found. Despite great similarities in inflammatory markers in islets in T1D and T2D, long-term enteroviral infiltration is a distinct pathological feature of T1D-associated autoimmunity and in T1D pancreases.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Enterovirus Infections , Humans , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Enterovirus Infections/complications , Insulin/metabolism , Inflammation/complications , RNA
4.
PLoS One ; 18(3): e0282771, 2023.
Article in English | MEDLINE | ID: mdl-36862649

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0161834.].

5.
Stem Cell Res Ther ; 13(1): 348, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35883121

ABSTRACT

Bone marrow mesenchymal stromal cells (BM-MSCs) have anti-inflammatory and pro-survival properties. Naturally, they do not express human leukocyte antigen class II surface antigens and have immunosuppressive capabilities. Together with their relatively easy accessibility and expansion, they are an attractive tool for organ support in transplantation and regenerative therapy. Autologous BM-MSC transplantation alone or together with transplanted islets improves ß-cell function, graft survival, and glycemic control in diabetes. Albeit MSCs' capacity to transdifferentiate into ß-cell is limited, their protective effects are mediated mainly by paracrine mechanisms through BM-MSCs circulating through the body. Direct cell-cell contact and spontaneous fusion of BM-MSCs with injured cells, although at a very low rate, are further mechanisms of their supportive effect and for tissue regeneration. Diabetes is a disease of long-term chronic inflammation and cell therapy requires stable, highly functional cells. Several tools and protocols have been developed by mimicking natural fusion events to induce and accelerate fusion in vitro to promote ß-cell-specific gene expression in fused cells. BM-MSC-islet fusion before transplantation may be a strategy for long-term islet survival and improved function. This review discusses the cell-protective and anti-inflammatory characteristics of BM-MSCs to boost highly functional insulin-producing cells in vitro and in vivo, and the efficacy of their fusion with ß-cells as a path to promote ß-cell regeneration.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Bone Marrow , Bone Marrow Cells , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism
6.
PLoS One ; 17(7): e0271783, 2022.
Article in English | MEDLINE | ID: mdl-35834533

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0001397.].

7.
Front Endocrinol (Lausanne) ; 13: 830097, 2022.
Article in English | MEDLINE | ID: mdl-35370966

ABSTRACT

A critical decline of functional insulin-producing pancreatic ß-cells is the central pathologic element of both type 1 and type 2 diabetes. Mammalian Sterile 20-like kinase 1 (MST1) is a key mediator of ß-cell failure and the identification of neratinib as MST1 inhibitor with potent effects on ß-cell survival represents a promising approach for causative diabetes therapy. Here we report a case of robust glycemia and HbA1c normalization in a patient with breast cancer-T2D comorbidity under neratinib, a potent triple kinase inhibitor of HER2/EGFR and MST1. The patient, aged 62 years, was enrolled in the plasmaMATCH clinical trial and received 240 mg neratinib once daily. Neratinib therapy correlated with great improvement in glucose and HbA1c both to physiological levels during the whole treatment period (average reduction of random glucose from 13.6 ± 0.4 to 6.3 ± 0.5 mmol/l and of HbA1c from 82.2 ± 3.9 to 45.6 ± 4.2 mmol/mol before and during neratinib). 18 months later, when neratinib was withdrawn, random glucose rapidly raised together with high blood glucose fluctuations, which reflected in elevated HbA1c levels. This clinical case reports the combination of HER2/EGFR/MST1-inhibition by neratinib for the pharmacological intervention to effectively restore normoglycemia in a patient with poorly controlled T2D and suggests neratinib as potent therapeutic regimen for the cancer-diabetes comorbidity.


Subject(s)
Breast Neoplasms , Diabetes Mellitus, Type 2 , Animals , Breast Neoplasms/complications , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Female , Glycemic Control , Humans , Mammals , Middle Aged , Quinolines , Receptor, ErbB-2/therapeutic use
8.
Nutr Diabetes ; 12(1): 7, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136036

ABSTRACT

The pro-apoptotic kinase Mammalian Sterile 20-like kinase 1 (MST1), an integral component of the Hippo pathway, is a key regulator of organ size, stress response, and tissue homeostasis; its aberrant hyperactivation is linked to multiple pathological disorders including diabetes. Here we show that MST1 deletion in mice resulted in improved glucose tolerance and insulin secretion, and restored pancreatic ß-cell mass as a result of improved ß-cell survival and proliferation in the combined high fat/high sucrose and streptozotocin (HFS/STZ) model of ß-cell destruction and diabetes. Importantly, the glucose-lowering effects in the MST1-knockout (KO) mice could be accounted to the enhanced ß-cell mass and improved insulin secretion without changes in insulin sensitivity. Metabolic and morphological data suggest that normalization of blood glucose and insulin secretion, islet architecture, and ß-cell mass by MST1 deletion in response to diabetes-induced injury occurs as a result of improved ß-cell survival and proliferation establishing MST1 as potent regulator of physiological ß-cell turnover.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Animals , Diabetes Mellitus/metabolism , Insulin , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mammals/metabolism , Mice , Mice, Knockout , Streptozocin/metabolism
10.
Cell Metab ; 34(2): 189-191, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108508

ABSTRACT

In this issue of Cell Metabolism, Cheng et al. identify olfactory receptor Olfr109 in ß cells with increased expression in islets from mouse models of obesity and type 1 and type 2 diabetes. Binding of a small insulin fragment to Olfr109 fosters islet inflammation, ß cell failure, and diabetes progression.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Animals , Insulin , Mice , Smell
11.
Cell Rep Med ; 2(8): 100371, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34467248

ABSTRACT

Enteroviral infections have been associated with autoimmunity and type 1 diabetes (T1D), but reliable methods to ascertain localization of single infected cells in the pancreas were missing. Using a single-molecule-based fluorescent in situ hybridization (smFISH) method, we detected increased virus infection in pancreases from organ donors with T1D and with disease-associated autoantibodies (AAb+). Although virus-positive ß cells are found at higher frequency in T1D pancreases, compared to control donors, but are scarce, most virus-positive cells are scattered in the exocrine pancreas. Augmented CD45+ lymphocytes in T1D pancreases show virus positivity or localization in close proximity to virus-positive cells. Many more infected cells were also found in spleens from T1D donors. The overall increased proportion of virus-positive cells in the pancreas of AAb+ and T1D organ donors suggests that enteroviruses are associated with immune cell infiltration, autoimmunity, and ß cell destruction in both preclinical and diagnosed T1D.


Subject(s)
Autoantibodies/immunology , Diabetes Mellitus, Type 1/immunology , Enterovirus/genetics , Pancreas/virology , RNA, Viral/metabolism , Tissue Donors , Humans , Lymphocytes/immunology , Spleen/immunology
12.
Autophagy ; 17(12): 4494-4496, 2021 12.
Article in English | MEDLINE | ID: mdl-34470573

ABSTRACT

A progressive decline in the macroautophagic/autophagic flux is a hallmark of pancreatic ß-cell failure in type 2 diabetes (T2D) but the responsible intrinsic factors and underlying molecular mechanisms are incompletely understood. A stress-sensitive multicomponent cellular loop of the Hippo pathway kinase LATS2 (large tumor suppressor 2), MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) and autophagy regulates ß-cell survival and metabolic adaptation. Chronic metabolic stress leads to LATS2 hyperactivation which then induces MTORC1, subsequently impairing the cellular autophagic flux and consequently triggering ß-cell death. Reciprocally, under physiological conditions, autophagy controls ß-cell survival by lysosomal degradation of LATS2. These signaling cross-talks and the interaction between autophagy and LATS2 are important for the regulation of ß-cell turnover and functional compensation under metabolic stress.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Autophagy/physiology , Diabetes Mellitus, Type 2/metabolism , Hippo Signaling Pathway , Humans , Insulin-Secreting Cells/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins/metabolism
13.
Nat Commun ; 12(1): 4928, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389720

ABSTRACT

Diabetes results from a decline in functional pancreatic ß-cells, but the molecular mechanisms underlying the pathological ß-cell failure are poorly understood. Here we report that large-tumor suppressor 2 (LATS2), a core component of the Hippo signaling pathway, is activated under diabetic conditions and induces ß-cell apoptosis and impaired function. LATS2 deficiency in ß-cells and primary isolated human islets as well as ß-cell specific LATS2 ablation in mice improves ß-cell viability, insulin secretion and ß-cell mass and ameliorates diabetes development. LATS2 activates mechanistic target of rapamycin complex 1 (mTORC1), a physiological suppressor of autophagy, in ß-cells and genetic and pharmacological inhibition of mTORC1 counteracts the pro-apoptotic action of activated LATS2. We further show a direct interplay between Hippo and autophagy, in which LATS2 is an autophagy substrate. On the other hand, LATS2 regulates ß-cell apoptosis triggered by impaired autophagy suggesting an existence of a stress-sensitive multicomponent cellular loop coordinating ß-cell compensation and survival. Our data reveal an important role for LATS2 in pancreatic ß-cell turnover and suggest LATS2 as a potential therapeutic target to improve pancreatic ß-cell survival and function in diabetes.


Subject(s)
Autophagy , Diabetes Mellitus/metabolism , Insulin-Secreting Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , Cells, Cultured , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Humans , Insulin-Secreting Cells/cytology , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , RNA Interference , Rats , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics
14.
Cell Rep ; 36(5): 109490, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348155

ABSTRACT

Pancreatic ß-cell failure is the key pathogenic element of the complex metabolic deterioration in type 2 diabetes (T2D); its underlying pathomechanism is still elusive. Here, we identify pleckstrin homology domain leucine-rich repeat protein phosphatases 1 and 2 (PHLPP1/2) as phosphatases whose upregulation leads to ß-cell failure in diabetes. PHLPP levels are highly elevated in metabolically stressed human and rodent diabetic ß-cells. Sustained hyper-activation of mechanistic target of rapamycin complex 1 (mTORC1) is the primary mechanism of the PHLPP upregulation linking chronic metabolic stress to ultimate ß-cell death. PHLPPs directly dephosphorylate and regulate activities of ß-cell survival-dependent kinases AKT and MST1, constituting a regulatory triangle loop to control ß-cell apoptosis. Genetic inhibition of PHLPPs markedly improves ß-cell survival and function in experimental models of diabetes in vitro, in vivo, and in primary human T2D islets. Our study presents PHLPPs as targets for functional regenerative therapy of pancreatic ß cells in diabetes.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/enzymology , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Apoptosis , Cell Survival , Diet, High-Fat , Female , Gene Deletion , Hepatocyte Growth Factor/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/pathology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , Models, Biological , Protein Biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stress, Physiological , Up-Regulation
15.
Trends Endocrinol Metab ; 32(11): 842-845, 2021 11.
Article in English | MEDLINE | ID: mdl-34373155

ABSTRACT

The widespread extrapulmonary complications of coronavirus disease 2019 (COVID-19) have gained momentum; the pancreas is another major target for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we take a closer look into potential pathological interactions. We provide an overview of the current knowledge and understanding of SARS-CoV-2 infection of the pancreas with a special focus on pancreatic islets and propose direct, indirect, and systemic mechanisms for pancreas injury as result of the COVID-19-diabetes fatal bidirectional relationship.


Subject(s)
COVID-19/metabolism , Diabetes Mellitus/metabolism , Insulin-Secreting Cells/metabolism , Acinar Cells/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Glucagon-Secreting Cells/metabolism , Humans , Islets of Langerhans/metabolism , Pancreas/metabolism , Receptors, Coronavirus/metabolism , SARS-CoV-2/metabolism , Serine Endopeptidases/metabolism , Viral Tropism
16.
Biochem Biophys Res Commun ; 568: 158-166, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34217973

ABSTRACT

The lactate dehydrogenase isoform A (LDHA) is a key metabolic enzyme that preferentially catalyzes the conversion of pyruvate to lactate. Whereas LDHA is highly expressed in many tissues, its expression is turned off in the differentiated adult ß-cell within the pancreatic islets. The repression of LDHA under normal physiological condition and its inappropriate upregulation under a diabetogenic environment is well-documented in rodent islets/ß-cells but little is known about LDHA expression in human islet cells and whether its abundance is altered under diabetic conditions. Analysis of public single-cell RNA-seq (sc-RNA seq) data as well as cell type-specific immunolabeling of human pancreatic islets showed that LDHA was mainly localized in human α-cells while it is expressed at a very low level in ß-cells. Furthermore, LDHA, both at mRNA and protein, as well as lactate production is upregulated in human pancreatic islets exposed to chronic high glucose treatment. Microscopic analysis of stressed human islets and autopsy pancreases from individuals with type 2 diabetes (T2D) showed LDHA upregulation mainly in human α-cells. Pharmacological inhibition of LDHA in isolated human islets enhanced insulin secretion under physiological conditions but did not significantly correct the deregulated secretion of insulin or glucagon under diabetic conditions.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucagon-Secreting Cells/metabolism , L-Lactate Dehydrogenase/genetics , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/cytology , Glucose/metabolism , Humans , Insulin Secretion , L-Lactate Dehydrogenase/analysis , L-Lactate Dehydrogenase/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Up-Regulation
17.
Front Endocrinol (Lausanne) ; 12: 697120, 2021.
Article in English | MEDLINE | ID: mdl-34290670

ABSTRACT

Glucagon-like peptide-1 (GLP-1) shows robust protective effects on ß-cell survival and function and GLP-1 based therapies are successfully applied for type-2 diabetes (T2D) and obesity. Another cleavage product of pro-glucagon, Glucagon-like peptide-2 (GLP-2; both GLP-1 and GLP-2 are inactivated by DPP-4) has received little attention in its action inside pancreatic islets. In this study, we investigated GLP-2 production, GLP-2 receptor (GLP-2R) expression and the effect of GLP-2R activation in human islets. Isolated human islets from non-diabetic donors were exposed to diabetogenic conditions: high glucose, palmitate, cytokine mix (IL-1ß/IFN-γ) or Lipopolysaccharide (LPS) in the presence or absence of the DPP4-inhibitor linagliptin, the TLR4 inhibitor TAK-242, the GLP-2R agonist teduglutide and/or its antagonist GLP-2(3-33). Human islets under control conditions secreted active GLP-2 (full-length, non-cleaved by DPP4) into the culture media, which was increased by combined high glucose/palmitate, the cytokine mix and LPS and highly potentiated by linagliptin. Low but reproducible GLP-2R mRNA expression was found in all analyzed human islet isolations from 10 donors, which was reduced by pro-inflammatory stimuli: the cytokine mix and LPS. GLP-2R activation by teduglutide neither affected acute or glucose stimulated insulin secretion nor insulin content. Also, teduglutide had no effect on high glucose/palmitate- or LPS-induced dysfunction in cultured human islets but dampened LPS-induced macrophage-dependent IL1B and IL10 expression, while its antagonist GLP-2(3-33) abolished such reduction. In contrast, the expression of islet macrophage-independent cytokines IL6, IL8 and TNF was not affected by teduglutide. Medium conditioned by teduglutide-exposed human islets attenuated M1-like polarization of human monocyte-derived macrophages, evidenced by a lower mRNA expression of pro-inflammatory cytokines, compared to vehicle treated islets, and a reduced production of itaconate and succinate, marker metabolites of pro-inflammatory macrophages. Our results reveal intra-islet production of GLP-2 and GLP-2R expression in human islets. Despite no impact on ß-cell function, local GLP-2R activation reduced islet inflammation which might be mediated by a crosstalk between endocrine cells and macrophages.


Subject(s)
Glucagon-Like Peptide 2/metabolism , Inflammation , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Macrophages/physiology , Cell Communication/drug effects , Cell Communication/physiology , Cells, Cultured , Female , Glucagon-Like Peptide-2 Receptor/genetics , Glucagon-Like Peptide-2 Receptor/metabolism , Homeostasis/drug effects , Humans , Immune System/drug effects , Immune System/physiology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Lipopolysaccharides , Macrophages/drug effects , Male , Pancreatitis/immunology , Pancreatitis/metabolism , Pancreatitis/pathology
18.
Nat Metab ; 3(3): 295-296, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33758425
20.
Microorganisms ; 8(7)2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32650582

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune disorder that results from the selective destruction of insulin-producing ß-cells in the pancreas. Up to now, the mechanisms triggering the initiation and progression of the disease are, in their complexity, not fully understood and imply the disruption of several tolerance networks. Viral infection is one of the environmental factors triggering diabetes, which is initially based on the observation that the disease's incidence follows a periodic pattern within the population. Moreover, the strong correlation of genetic susceptibility is a prerequisite for enteroviral infection associated islet autoimmunity. Epidemiological data and clinical findings indicate enteroviral infections, mainly of the coxsackie B virus family, as potential pathogenic mechanisms to trigger the autoimmune reaction towards ß-cells, resulting in the boost of inflammation following ß-cell destruction and the onset of T1D. This review discusses previously identified virus-associated genetics and pathways of ß-cell destruction. Is it the virus itself which leads to ß-cell destruction and T1D progression? Or is it genetic, so that the virus may activate auto-immunity and ß-cell destruction only in genetically predisposed individuals?

SELECTION OF CITATIONS
SEARCH DETAIL
...