Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-39078946

ABSTRACT

CONTEXT: Diabetes is a heterogenic disease and distinct clusters have emerged, but the implications for diverse populations have remained understudied. OBJECTIVE: Apply cluster analysis to a diverse diabetes cohort in the U.S. Deep South. DESIGN: Retrospective hierarchical cluster analysis of electronic health records from 89,875 patients diagnosed with diabetes between January 1, 2010, and December 31, 2019, at the Kirklin Clinic of the University of Alabama at Birmingham, an ambulatory referral center. PATIENTS: Adult patients with ICD diabetes codes were selected based on available data for 6 established clustering parameters (GAD-autoantibody; HbA1c; BMI; Diagnosis age; HOMA2-B; HOMA2-IR); ∼42% were Black/African American. MAIN OUTCOME MEASURE(S): Diabetes subtypes and their associated characteristics in a diverse adult population based on clustering analysis. We hypothesized that racial background would affect the distribution of subtypes. Outcome and hypothesis were formulated prior to data collection. RESULTS: Diabetes cluster distribution was significantly different in Black/African Americans compared to Whites (P<0.001). Black/African Americans were more likely to have severe insulin deficient diabetes (SIDD) (OR 1.83, CI 1.36-2.45, P<0.001), associated with more serious metabolic perturbations and a higher risk for complications (OR 1.42, 95% CI 1.06-1.90, P=0.020). Surprisingly, Black/African Americans specifically had more severe impairment of beta cell function (HOMA2-B, C-peptide) (P<0.001), while not being more obese or insulin resistant. CONCLUSIONS: Racial background greatly influences diabetes cluster distribution and Black/African Americans are more frequently and more severely affected by SIDD. This may further help explain the disparity in outcomes and have implications for treatment choice.

2.
J Clin Transl Sci ; 7(1): e214, 2023.
Article in English | MEDLINE | ID: mdl-37900350

ABSTRACT

Knowledge graphs have become a common approach for knowledge representation. Yet, the application of graph methodology is elusive due to the sheer number and complexity of knowledge sources. In addition, semantic incompatibilities hinder efforts to harmonize and integrate across these diverse sources. As part of The Biomedical Translator Consortium, we have developed a knowledge graph-based question-answering system designed to augment human reasoning and accelerate translational scientific discovery: the Translator system. We have applied the Translator system to answer biomedical questions in the context of a broad array of diseases and syndromes, including Fanconi anemia, primary ciliary dyskinesia, multiple sclerosis, and others. A variety of collaborative approaches have been used to research and develop the Translator system. One recent approach involved the establishment of a monthly "Question-of-the-Month (QotM) Challenge" series. Herein, we describe the structure of the QotM Challenge; the six challenges that have been conducted to date on drug-induced liver injury, cannabidiol toxicity, coronavirus infection, diabetes, psoriatic arthritis, and ATP1A3-related phenotypes; the scientific insights that have been gleaned during the challenges; and the technical issues that were identified over the course of the challenges and that can now be addressed to foster further development of the prototype Translator system. We close with a discussion on Large Language Models such as ChatGPT and highlight differences between those models and the Translator system.

3.
Diabetes ; 72(10): 1460-1469, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37494660

ABSTRACT

Verapamil promotes functional ß-cell mass and improves glucose homeostasis in diabetic mice and humans with type 1 diabetes (T1D). Now, our global proteomics analysis of serum from subjects with T1D at baseline and after 1 year of receiving verapamil or placebo revealed IGF-I as a protein with significantly changed abundance over time. IGF-I, which promotes ß-cell survival and insulin secretion, decreased during disease progression, and this decline was blunted by verapamil. In addition, we found that verapamil reduces ß-cell expression of IGF-binding protein 3 (IGFBP3), whereas IGFBP3 was increased in human islets exposed to T1D-associated cytokines and in diabetic NOD mouse islets. IGFBP3 binds IGF-I and blocks its downstream signaling, which has been associated with increased ß-cell apoptosis and impaired glucose homeostasis. Consistent with the downregulation of IGFBP3, we have now discovered that verapamil increases ß-cell IGF-I signaling and phosphorylation/activation of the IGF-I receptor (IGF1R). Moreover, we found that thioredoxin-interacting protein (TXNIP), a proapoptotic factor downregulated by verapamil, promotes IGFBP3 expression and inhibits the phosphorylation/activation of IGF1R. Thus, our results reveal IGF-I signaling as yet another previously unappreciated pathway affected by verapamil and TXNIP that may contribute to the beneficial verapamil effects in the context of T1D. ARTICLE HIGHLIGHTS: Verapamil prevents the decline of IGF-I in subjects with type 1 diabetes (T1D). Verapamil decreases the expression of ß-cell IGF-binding protein 3 (IGFBP3), whereas IGFBP3 is increased in human and mouse islets under T1D conditions. Verapamil promotes ß-cell IGF-I signaling by increasing phosphorylation of IGF-I receptor and its downstream effector AKT. Thioredoxin-interacting protein (TXNIP) increases IGFBP3 expression and inhibits the phosphorylation/activation of IGF1R in ß-cells. Regulation of IGFBP3 and IGF-I signaling by verapamil and TXNIP may contribute to the beneficial verapamil effects in the context of T1D.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Humans , Mice , Animals , Diabetes Mellitus, Type 1/drug therapy , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Verapamil/pharmacology , Verapamil/therapeutic use , Insulin-Like Growth Factor Binding Protein 3/metabolism , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred NOD , Thioredoxins/metabolism , Glucose
4.
iScience ; 26(7): 107036, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37360692

ABSTRACT

Human and mouse genetics have delivered numerous diabetogenic loci, but it is mainly through the use of animal models that the pathophysiological basis for their contribution to diabetes has been investigated. More than 20 years ago, we serendipidously identified a mouse strain that could serve as a model of obesity-prone type 2 diabetes, the BTBR (Black and Tan Brachyury) mouse (BTBR T+ Itpr3tf/J, 2018) carrying the Lepob mutation. We went on to discover that the BTBR-Lepob mouse is an excellent model of diabetic nephropathy and is now widely used by nephrologists in academia and the pharmaceutical industry. In this review, we describe the motivation for developing this animal model, the many genes identified and the insights about diabetes and diabetes complications derived from >100 studies conducted in this remarkable animal model.

5.
Endocrinology ; 163(11)2022 10 11.
Article in English | MEDLINE | ID: mdl-35957590

ABSTRACT

Thioredoxin-interacting protein (TXNIP) has emerged as a key factor in pancreatic beta cell biology, and its upregulation by glucose and diabetes contributes to the impairment in functional beta cell mass and glucose homeostasis. In addition, beta cell deletion of TXNIP protects against diabetes in different mouse models. However, while TXNIP is ubiquitously expressed, its role in pancreatic alpha cells has remained elusive. We generated an alpha cell TXNIP knockout (aTKO) mouse and assessed the effects on glucose homeostasis. While no significant changes were observed on regular chow, after a 30-week high-fat diet, aTKO animals showed improvement in glucose tolerance and lower blood glucose levels compared to their control littermates. Moreover, in the context of streptozotocin (STZ)-induced diabetes, aTKO mice showed significantly lower blood glucose levels compared to controls. While serum insulin levels were reduced in both control and aTKO mice, STZ-induced diabetes significantly increased glucagon levels in control mice, but this effect was blunted in aTKO mice. Moreover, glucagon secretion from aTKO islets was >2-fold lower than from control islets, while insulin secretion was unchanged in aTKO islets. At the same time, no change in alpha cell or beta cell numbers or mass was observed, and glucagon and insulin expression and content were comparable in isolated islets from aTKO and control mice. Thus together the current studies suggest that downregulation of alpha cell TXNIP is associated with reduced glucagon secretion and that this may contribute to the glucose-lowering effects observed in diabetic aTKO mice.


Subject(s)
Diabetes Mellitus, Experimental , Glucagon-Secreting Cells , Hyperglycemia , Insulin-Secreting Cells , Pancreatic Diseases , Animals , Blood Glucose/metabolism , Carrier Proteins , Diabetes Mellitus, Experimental/metabolism , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , Streptozocin , Thioredoxins
6.
Diabetes ; 71(10): 2123-2135, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35877180

ABSTRACT

Long-term glucagon receptor (GCGR) agonism is associated with hyperglycemia and glucose intolerance, while acute GCGR agonism enhances whole-body insulin sensitivity and hepatic AKTSer473 phosphorylation. These divergent effects establish a critical gap in knowledge surrounding GCGR action. mTOR complex 2 (mTORC2) is composed of seven proteins, including RICTOR, which dictates substrate binding and allows for targeting of AKTSer473. We used a liver-specific Rictor knockout mouse (RictorΔLiver) to investigate whether mTORC2 is necessary for insulin receptor (INSR) and GCGR cross talk. RictorΔLiver mice were characterized by impaired AKT signaling and glucose intolerance. Intriguingly, RictorΔLiver mice were also resistant to GCGR-stimulated hyperglycemia. Consistent with our prior report, GCGR agonism increased glucose infusion rate and suppressed hepatic glucose production during hyperinsulinemic-euglycemic clamp of control animals. However, these benefits to insulin sensitivity were ablated in RictorΔLiver mice. We observed diminished AKTSer473 and GSK3α/ßSer21/9 phosphorylation in RictorΔLiver mice, whereas phosphorylation of AKTThr308 was unaltered in livers from clamped mice. These signaling effects were replicated in primary hepatocytes isolated from RictorΔLiver and littermate control mice, confirming cell-autonomous cross talk between GCGR and INSR pathways. In summary, our study reveals the necessity of RICTOR, and thus mTORC2, in GCGR-mediated enhancement of liver and whole-body insulin action.


Subject(s)
Glucose Intolerance , Hyperglycemia , Insulin Resistance , Animals , Glucose/metabolism , Glucose Intolerance/metabolism , Homeostasis , Hyperglycemia/metabolism , Insulin/metabolism , Insulin/pharmacology , Insulin, Regular, Human , Liver/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Receptor, Insulin/metabolism , Receptors, Glucagon/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Nat Commun ; 13(1): 1159, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35241690

ABSTRACT

Currently, no oral medications are available for type 1 diabetes (T1D). While our recent randomized placebo-controlled T1D trial revealed that oral verapamil had short-term beneficial effects, their duration and underlying mechanisms remained elusive. Now, our global T1D serum proteomics analysis identified chromogranin A (CHGA), a T1D-autoantigen, as the top protein altered by verapamil and as a potential therapeutic marker and revealed that verapamil normalizes serum CHGA levels and reverses T1D-induced elevations in circulating proinflammatory T-follicular-helper cell markers. RNA-sequencing further confirmed that verapamil regulates the thioredoxin system and promotes an anti-oxidative, anti-apoptotic and immunomodulatory gene expression profile in human islets. Moreover, continuous use of oral verapamil delayed T1D progression, promoted endogenous beta-cell function and lowered insulin requirements and serum CHGA levels for at least 2 years and these benefits were lost upon discontinuation. Thus, the current studies provide crucial mechanistic and clinical insight into the beneficial effects of verapamil in T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Immunologic Factors/therapeutic use , Immunotherapy , Insulin , Verapamil/pharmacology , Verapamil/therapeutic use
8.
Endocrinology ; 163(5)2022 05 01.
Article in English | MEDLINE | ID: mdl-35290443

ABSTRACT

Endoplasmic reticulum (ER) stress contributes to pancreatic beta-cell apoptosis in diabetes, but the factors involved are still not fully elucidated. Growth differentiation factor 15 (GDF15) is a stress response gene and has been reported to be increased and play an important role in various diseases. However, the role of GDF15 in beta cells in the context of ER stress and diabetes is still unclear. In this study, we have discovered that GDF15 promotes ER stress-induced beta-cell apoptosis and that downregulation of GDF15 has beneficial effects on beta-cell survival in diabetes. Specifically, we found that GDF15 is induced by ER stress in beta cells and human islets, and that the transcription factor C/EBPß is involved in this process. Interestingly, ER stress-induced apoptosis was significantly reduced in INS-1 cells with Gdf15 knockdown and in isolated Gdf15 knockout mouse islets. In vivo, we found that Gdf15 deletion attenuates streptozotocin-induced diabetes by preserving beta cells and insulin levels. Moreover, deletion of Gdf15 significantly delayed diabetes development in spontaneous ER stress-prone Akita mice. Thus, our findings suggest that GDF15 contributes to ER stress-induced beta-cell apoptosis and that inhibition of GDF15 may represent a novel strategy to promote beta-cell survival and treat diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Animals , Apoptosis , Diabetes Mellitus, Experimental/genetics , Endoplasmic Reticulum Stress , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/pharmacology , Mice
9.
Islets ; 14(1): 58-68, 2022 12 31.
Article in English | MEDLINE | ID: mdl-34968409

ABSTRACT

Excess nutrients and proinflammatory cytokines impart stresses on pancreatic islet ß-cells that, if unchecked, can lead to cellular dysfunction and/or death. Among these stress-induced effects is loss of key ß-cell transcriptional regulator mRNA and protein levels required for ß-cell function. Previously, our lab and others reported that LIM-domain complexes comprised the LDB1 transcriptional co-regulator and Islet-1 (ISL1) transcription factor are required for islet ß-cell development, maturation, and function. The LDB1:ISL1 complex directly occupies and regulates key ß-cell genes, including MafA, Pdx1, and Slc2a2, to maintain ß-cell identity and function. Given the importance of LDB1:ISL1 complexes, we hypothesized that LDB1 and/or ISL1 levels, like other transcriptional regulators, are sensitive to ß-cell nutrient and cytokine stresses, likely contributing to ß-cell (dys)function under various stimuli. We tested this by treating ß-cell lines or primary mouse islets with elevating glucose concentrations, palmitate, or a cytokine cocktail of IL-1ß, TNFα, and IFNγ. We indeed observed that LDB1 mRNA and/or protein levels were reduced upon palmitate and cytokine (cocktail or singly) incubation. Conversely, acute high glucose treatment of ß-cells did not impair LDB1 or ISL1 levels, but increased LDB1:ISL1 interactions. These observations suggest that LDB1:ISL1 complex formation is sensitive to ß-cell stresses and that targeting and/or stabilizing this complex may rescue lost ß-cell gene expression to preserve cellular function.


Subject(s)
LIM Domain Proteins , Transcription Factors , Animals , Cytokines , DNA-Binding Proteins , Glucose/pharmacology , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Palmitates , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Diabetes ; 2021 11 13.
Article in English | MEDLINE | ID: mdl-34957490

ABSTRACT

One hundred years have passed since the discovery of insulin-an achievement that transformed diabetes from a fatal illness into a manageable chronic condition. The decades since that momentous achievement have brought ever more rapid innovation and advancement in diabetes research and clinical care. To celebrate the important work of the past century and help to chart a course for its continuation into the next, the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases recently held a joint international symposium, bringing together a cohort of researchers with diverse interests and backgrounds from both countries and beyond to discuss their collective quest to better understand the heterogeneity of diabetes and thus gain insights to inform new directions in diabetes treatment and prevention. This article summarizes the proceedings of that symposium, which spanned cutting-edge research into various aspects of islet biology, the heterogeneity of diabetic phenotypes, and the current state of and future prospects for precision medicine in diabetes.

12.
Endocrinology ; 162(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33367814

ABSTRACT

Increased glucagon is a hallmark of diabetes and leads to worsening of the hyperglycemia, but the molecular mechanisms causing it are still unknown. We therefore investigated the possibility that microRNAs might be involved in the regulation of glucagon. Indeed, analysis of the glucagon 3' untranslated region (UTR) revealed potential binding sites for miR-320a, and using luciferase reporter assays we found that miR-320a directly targets the 3' UTRs of human and rodent glucagon. In addition, endogenous glucagon mRNA and protein expression as well as glucagon secretion were reduced in response to miR-320a overexpression, whereas inhibition of miR-320a upregulated glucagon expression. Interestingly, miR-320a expression was decreased by high glucose, and this was associated with an increase in glucagon expression in human islets and mouse αTC1-6 cells. Moreover, miR-320a overexpression completely blunted these effects. Importantly, miR-320a was also significantly downregulated in human islets of subjects with type 2 diabetes and this was accompanied by increased glucagon expression. Thus, our data suggest that glucose-induced downregulation of miR-320a may contribute to the paradoxical increase in glucagon observed in type 2 diabetes and reveal for the first time that glucagon expression is under the control by a microRNA providing novel insight into the abnormal regulation of glucagon in diabetes.


Subject(s)
Glucagon/genetics , MicroRNAs/physiology , 3' Untranslated Regions/drug effects , 3' Untranslated Regions/genetics , Adolescent , Adult , Aged , Animals , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucose/pharmacology , HEK293 Cells , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Middle Aged
13.
medRxiv ; 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32766607

ABSTRACT

BACKGROUND: Coronavirus disease-2019 (COVID-19) is a growing pandemic with an increasing death toll that has been linked to various comorbidities as well as racial disparity. However, the specific characteristics of these at-risk populations are still not known and approaches to lower mortality are lacking. METHODS: We conducted a retrospective electronic health record data analysis of 25,326 subjects tested for COVID-19 between 2/25/20 and 6/22/20 at the University of Alabama at Birmingham Hospital, a tertiary health care center in the racially diverse Southern U.S. The primary outcome was mortality in COVID-19-positive subjects and the association with subject characteristics and comorbidities was analyzed using simple and multiple linear logistic regression. RESULTS: The odds ratio of contracting COVID-19 was disproportionately high in Blacks/African-Americans (OR 2.6; 95%CI 2.19-3.10; p<0.0001) and in subjects with obesity (OR 1.93; 95%CI 1.64-2.28; p<0.0001), hypertension (OR 2.46; 95%CI 2.07-2.93; p<0.0001), and diabetes (OR 2.11; 95%CI 1.78-2.48; p<0.0001). Diabetes was also associated with a dramatic increase in mortality (OR 3.62; 95%CI 2.11-6.2; p<0.0001) and emerged as an independent risk factor in this diverse population even after correcting for age, race, sex, obesity and hypertension. Interestingly, we found that metformin treatment was independently associated with a significant reduction in mortality in subjects with diabetes and COVID-19 (OR 0.33; 95%CI 0.13-0.84; p=0.0210). CONCLUSION: Thus, these results suggest that while diabetes is an independent risk factor for COVID-19-related mortality, this risk is dramatically reduced in subjects taking metformin, raising the possibility that metformin may provide a protective approach in this high risk population.

14.
Diabetes ; 69(9): 2003-2016, 2020 09.
Article in English | MEDLINE | ID: mdl-32611548

ABSTRACT

Targeting retinoid X receptor (RXR) has been proposed as one of the therapeutic strategies to treat individuals with metabolic syndrome, as RXR heterodimerizes with multiple nuclear receptors that regulate genes involved in metabolism. Despite numerous efforts, RXR ligands (rexinoids) have not been approved for clinical trials to treat metabolic syndrome due to the serious side effects such as hypertriglyceridemia and altered thyroid hormone axis. In this study, we demonstrate a novel rexinoid-like small molecule, UAB126, which has positive effects on metabolic syndrome without the known side effects of potent rexinoids. Oral administration of UAB126 ameliorated obesity, insulin resistance, hepatic steatosis, and hyperlipidemia without changes in food intake, physical activity, and thyroid hormone levels. RNA-sequencing analysis revealed that UAB126 regulates the expression of genes in the liver that are modulated by several nuclear receptors, including peroxisome proliferator-activated receptor α and/or liver X receptor in conjunction with RXR. Furthermore, UAB126 not only prevented but also reversed obesity-associated metabolic disorders. The results suggest that optimized modulation of RXR may be a promising strategy to treat metabolic disorders without side effects. Thus, the current study reveals that UAB126 could be an attractive therapy to treat individuals with obesity and its comorbidities.


Subject(s)
Diet, High-Fat , Fatty Liver/drug therapy , Hyperlipidemias/drug therapy , Insulin Resistance/physiology , Liver/drug effects , Obesity/drug therapy , Retinoid X Receptors/agonists , Animals , Fatty Liver/blood , Hyperlipidemias/blood , Lipids/blood , Male , Mice , Obesity/blood
15.
Cell Metab ; 32(3): 353-365.e8, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32726606

ABSTRACT

Diabetes is characterized by hyperglycemia, loss of functional islet beta cell mass, deficiency of glucose-lowering insulin, and persistent alpha cell secretion of gluconeogenic glucagon. Still, no therapies that target these underlying processes are available. We therefore performed high-throughput screening of 300,000 compounds and extensive medicinal chemistry optimization and here report the discovery of SRI-37330, an orally bioavailable, non-toxic small molecule, which effectively rescued mice from streptozotocin- and obesity-induced (db/db) diabetes. Interestingly, in rat cells and in mouse and human islets, SRI-37330 inhibited expression and signaling of thioredoxin-interacting protein, which we have previously found to be elevated in diabetes and to have detrimental effects on islet function. In addition, SRI-37330 treatment inhibited glucagon secretion and function, reduced hepatic glucose production, and reversed hepatic steatosis. Thus, these studies describe a newly designed chemical compound that, compared to currently available therapies, may provide a distinct and effective approach to treating diabetes.


Subject(s)
Carrier Proteins/genetics , Diabetes Mellitus, Experimental/drug therapy , Glucagon/metabolism , Hypoglycemic Agents/pharmacology , Small Molecule Libraries/pharmacology , Administration, Oral , Animals , Carrier Proteins/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Male , Mice , Mice, Inbred C57BL , Rats , Small Molecule Libraries/administration & dosage , Streptozocin
16.
Front Endocrinol (Lausanne) ; 11: 600439, 2020.
Article in English | MEDLINE | ID: mdl-33519709

ABSTRACT

Background: Coronavirus disease-2019 (COVID-19) is a growing pandemic with an increasing death toll that has been linked to various comorbidities as well as racial disparity. However, the specific characteristics of these at-risk populations are still not known and approaches to lower mortality are lacking. Methods: We conducted a retrospective electronic health record data analysis of 25,326 subjects tested for COVID-19 between 2/25/20 and 6/22/20 at the University of Alabama at Birmingham Hospital, a tertiary health care center in the racially diverse Southern U.S. The primary outcome was mortality in COVID-19-positive subjects and the association with subject characteristics and comorbidities was analyzed using simple and multiple linear logistic regression. Results: The odds ratio of contracting COVID-19 was disproportionately high in Blacks/African-Americans (OR 2.6; 95% CI 2.19-3.10; p<0.0001) and in subjects with obesity (OR 1.93; 95% CI 1.64-2.28; p<0.0001), hypertension (OR 2.46; 95% CI 2.07-2.93; p<0.0001), and diabetes (OR 2.11; 95% CI 1.78-2.48; p<0.0001). Diabetes was also associated with a dramatic increase in mortality (OR 3.62; 95% CI 2.11-6.2; p<0.0001) and emerged as an independent risk factor in this diverse population even after correcting for age, race, sex, obesity, and hypertension. Interestingly, we found that metformin treatment prior to diagnosis of COVID-19 was independently associated with a significant reduction in mortality in subjects with diabetes and COVID-19 (OR 0.33; 95% CI 0.13-0.84; p=0.0210). Conclusion: Thus, these results suggest that while diabetes is an independent risk factor for COVID-19-related mortality, this risk is dramatically reduced in subjects taking metformin prior to diagnosis of COVID-19, raising the possibility that metformin may provide a protective approach in this high risk population.


Subject(s)
COVID-19/mortality , Diabetes Mellitus/mortality , Ethnicity/statistics & numerical data , Hospital Mortality/trends , Hospitalization/statistics & numerical data , Metformin/therapeutic use , SARS-CoV-2/drug effects , Aged , COVID-19/transmission , COVID-19/virology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/epidemiology , Diabetes Mellitus/virology , Female , Follow-Up Studies , Humans , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Prognosis , Retrospective Studies , SARS-CoV-2/isolation & purification , Survival Rate , United States/epidemiology , COVID-19 Drug Treatment
17.
Am J Physiol Endocrinol Metab ; 317(4): E723-E730, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31408375

ABSTRACT

Pancreatic beta-cell death is a major factor in the pathogenesis of type 1 diabetes (T1D), but straightforward methods to measure beta-cell loss in humans are lacking, underlining the need for novel biomarkers. Using studies in INS-1 cells, human islets, diabetic mice, and serum samples of subjects with T1D at different stages, we have identified serum miR-204 as an early biomarker of T1D-associated beta-cell loss in humans. MiR-204 is a highly enriched microRNA in human beta-cells, and we found that it is released from dying beta-cells and detectable in human serum. We further discovered that serum miR-204 was elevated in children and adults with T1D and in autoantibody-positive at-risk subjects but not in type 2 diabetes or other autoimmune diseases and was inversely correlated with remaining beta-cell function in recent-onset T1D. Thus, serum miR-204 may provide a much needed novel approach to assess early T1D-associated human beta-cell loss even before onset of overt disease.


Subject(s)
Biomarkers/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , MicroRNAs/blood , Adolescent , Adult , Animals , Autoimmune Diseases/blood , Case-Control Studies , Cell Line , Child , Female , Humans , Islets of Langerhans Transplantation , Male , Mice , Mice, Inbred C57BL , Middle Aged , Primary Cell Culture
18.
Nat Med ; 24(8): 1108-1112, 2018 08.
Article in English | MEDLINE | ID: mdl-29988125

ABSTRACT

Pancreatic beta cell loss is a key factor in the pathogenesis of type 1 diabetes (T1D), but therapies to halt this process are lacking. We previously reported that the approved antihypertensive calcium-channel blocker verapamil, by decreasing the expression of thioredoxin-interacting protein, promotes the survival of insulin-producing beta cells and reverses diabetes in mouse models1. To translate these findings into humans, we conducted a randomized double-blind placebo-controlled phase 2 clinical trial ( NCT02372253 ) to assess the efficacy and safety of oral verapamil added for 12 months to a standard insulin regimen in adult subjects with recent-onset T1D. Verapamil treatment, compared with placebo was well tolerated and associated with an improved mixed-meal-stimulated C-peptide area under the curve, a measure of endogenous beta cell function, at 3 and 12 months (prespecified primary endpoint), as well as with a lower increase in insulin requirements, fewer hypoglycemic events and on-target glycemic control (secondary endpoints). Thus, addition of once-daily oral verapamil may be a safe and effective novel approach to promote endogenous beta cell function and reduce insulin requirements and hypoglycemic episodes in adult individuals with recent-onset T1D.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Verapamil/therapeutic use , Adult , Blood Pressure/drug effects , Diabetes Mellitus, Type 1/physiopathology , Heart Rate/drug effects , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Verapamil/pharmacology
19.
Curr Opin Endocrinol Diabetes Obes ; 25(2): 75-80, 2018 04.
Article in English | MEDLINE | ID: mdl-29356688

ABSTRACT

PURPOSE OF REVIEW: Thioredoxin-interacting protein has emerged as a major factor regulating pancreatic ß-cell dysfunction and death, key processes in the pathogenesis of type 1 and type 2 diabetes. Accumulating evidence based on basic, preclinical, and retrospective epidemiological research suggests that TXNIP represents a promising therapeutic target for diabetes. The present review is aimed at providing an update regarding these developments. RECENT FINDINGS: TXNIP has been shown to be induced by glucose and increased in diabetes and to promote ß-cell apoptosis, whereas TXNIP deletion protected against diabetes. More recently, TXNIP inhibition has also been found to promote insulin production and glucagon-like peptide 1 signaling via regulation of a microRNA. ß-Cell TXNIP expression itself was found to be regulated by hypoglycemic agents, carbohydrate-response-element-binding protein, and cytosolic calcium or the calcium channel blocker, verapamil. Retrospective studies now further suggest that verapamil use might be associated with a lower incidence of type 2 diabetes in humans. SUMMARY: TXNIP has emerged as a key factor in the regulation of functional ß-cell mass and TXNIP inhibition has shown beneficial effects in a variety of studies. Thus, the inhibition of TXNIP may provide a novel approach to the treatment of diabetes.


Subject(s)
Carrier Proteins/physiology , Diabetes Mellitus/therapy , Molecular Targeted Therapy , Therapies, Investigational/methods , Animals , Carrier Proteins/antagonists & inhibitors , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Diabetes ; 67(2): 256-264, 2018 02.
Article in English | MEDLINE | ID: mdl-29101219

ABSTRACT

Glucagon-like peptide 1 receptor (GLP1R) agonists are widely used to treat diabetes. However, their function is dependent on adequate GLP1R expression, which is downregulated in diabetes. GLP1R is highly expressed on pancreatic ß-cells, and activation by endogenous incretin or GLP1R agonists increases cAMP generation, which stimulates glucose-induced ß-cell insulin secretion and helps maintain glucose homeostasis. We now have discovered that the highly ß-cell-enriched microRNA, miR-204, directly targets the 3' UTR of GLP1R and thereby downregulates its expression in the ß-cell-derived rat INS-1 cell line and primary mouse and human islets. Furthermore, in vivo deletion of miR-204 promoted islet GLP1R expression and enhanced responsiveness to GLP1R agonists, resulting in improved glucose tolerance, cAMP production, and insulin secretion as well as protection against diabetes. Since we recently identified thioredoxin-interacting protein (TXNIP) as an upstream regulator of miR-204, we also assessed whether in vivo deletion of TXNIP could mimic that of miR-204. Indeed, it also enhanced islet GLP1R expression and GLP1R agonist-induced insulin secretion and glucose tolerance. Thus, the present studies show for the first time that GLP1R is under the control of a microRNA, miR-204, and uncover a previously unappreciated link between TXNIP and incretin action.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Gene Expression Regulation , Glucagon-Like Peptide-1 Receptor/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , HEK293 Cells , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mutation , RNA , RNA Interference , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL