Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Dev Orig Health Dis ; 13(6): 806-811, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35450554

ABSTRACT

With type 2 diabetes presenting at younger ages, there is a growing need to identify biomarkers of future glucose intolerance. A high (20%) prevalence of glucose intolerance at 18 years was seen in women from the Pune Maternal Nutrition Study (PMNS) birth cohort. We investigated the potential of circulating microRNAs in risk stratification for future pre-diabetes in these women. Here, we provide preliminary longitudinal analyses of circulating microRNAs in normal glucose tolerant (NGT@18y, N = 10) and glucose intolerant (N = 8) women (ADA criteria) at 6, 12 and 17 years of their age using discovery analysis (OpenArray™ platform). Machine-learning workflows involving Lasso with bootstrapping/leave-one-out cross-validation identified microRNAs associated with glucose intolerance at 18 years of age. Several microRNAs, including miR-212-3p, miR-30e-3p and miR-638, stratified glucose-intolerant women from NGT at childhood. Our results suggest that circulating microRNAs, longitudinally assessed over 17 years of life, are dynamic biomarkers associated with and predictive of pre-diabetes at 18 years of age. Validation of these findings in males and remaining participants from the PMNS birth cohort will provide a unique opportunity to study novel epigenetic mechanisms in the life-course progression of glucose intolerance and enhance current clinical risk prediction of pre-diabetes and progression to type 2 diabetes.


Subject(s)
Circulating MicroRNA , Diabetes Mellitus, Type 2 , Glucose Intolerance , MicroRNAs , Prediabetic State , Child, Preschool , Male , Humans , Adolescent , Female , Prediabetic State/diagnosis , Prediabetic State/epidemiology , Prediabetic State/genetics , Glucose Intolerance/diagnosis , Glucose Intolerance/epidemiology , Glucose Intolerance/genetics , Circulating MicroRNA/genetics , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , India , MicroRNAs/genetics , Biomarkers , Glucose
2.
Methods Mol Biol ; 2224: 87-98, 2021.
Article in English | MEDLINE | ID: mdl-33606208

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease, where insulin-producing ß-cells in the pancreas are inappropriately recognized and destroyed by immune cells. Islet transplantation is the most successful cell-based therapy for T1D individuals who experience frequent and severe life-threatening hypoglycemia. However, this therapy is extremely restricted owing to the limited availability of donor pancreas. In recent years, significant progress has been made in generating ß-cells from stem/progenitor cells using different approaches of in vitro differentiation. The insulin production from such in vitro generated ß-cells is still far less than that observed in islet ß-cells. We employed a novel strategy to improve the efficiency of progenitor cell differentiation by performing partial mouse pancreas resection after transplanting in vitro generated insulin-producing cells under the kidney capsule of these mice. Pancreas resection (pancreatectomy) has been shown to induce regenerative pathways, leading to regeneration of almost the entire resected pancreas over 3-5 weeks in mice. We found that in our method, regenerating mouse pancreas promotes better graft differentiation/maturation and insulin production from transplanted cells. In this chapter, we detail the protocols used for transplantation of in vitro differentiated cells in immunocompromised mice, partial pancreatectomy in host (NOD scid) mice, and assessment of graft function. We believe that our protocols provide a solid platform for further studies aimed at understanding growth/differentiation molecules secreted from regenerating pancreas that promote graft maturation.


Subject(s)
Cell Differentiation/physiology , Pancreas/physiology , Animals , Diabetes Mellitus, Type 1/physiopathology , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation/physiology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatectomy/methods , Stem Cells/physiology
3.
Cell Metab ; 22(2): 312-9, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26166746

ABSTRACT

People in developing countries have faced multigenerational undernutrition and are currently undergoing major lifestyle changes, contributing to an epidemic of metabolic diseases, though the underlying mechanisms remain unclear. Using a Wistar rat model of undernutrition over 50 generations, we show that Undernourished rats exhibit low birth-weight, high visceral adiposity (DXA/MRI), and insulin resistance (hyperinsulinemic-euglycemic clamps), compared to age-/gender-matched control rats. Undernourished rats also have higher circulating insulin, homocysteine, endotoxin and leptin levels, lower adiponectin, vitamin B12 and folate levels, and an 8-fold increased susceptibility to Streptozotocin-induced diabetes compared to control rats. Importantly, these metabolic abnormalities are not reversed after two generations of unrestricted access to commercial chow (nutrient recuperation). Altered epigenetic signatures in insulin-2 gene promoter region of Undernourished rats are not reversed by nutrient recuperation, and may contribute to the persistent detrimental metabolic profiles in similar multigenerational undernourished human populations.


Subject(s)
Adiposity , Diabetes Mellitus, Experimental , Malnutrition , Obesity , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Disease Susceptibility , Humans , Malnutrition/complications , Malnutrition/diet therapy , Malnutrition/metabolism , Malnutrition/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Rats , Rats, Wistar
4.
Sci Rep ; 1: 81, 2011.
Article in English | MEDLINE | ID: mdl-22355600

ABSTRACT

Visceral adiposity is a risk factor for cardiovascular disorders, type 2 diabetes mellitus (T2D) and associated metabolic diseases. Sub-cutaneous fat is believed to be intrinsically different from visceral fat. To understand molecular mechanisms involved in metabolic advantages of fat transplantation, we studied a rat model of diet-induced adiposity. Adipokine genes (Adiponectin, Leptin, Resistin and Visfatin) were expressed at 10,000 to a million-fold lower in visceral fat depot as compared to peripheral (thigh/chest) fat depots. Interestingly, autologous transplantation of visceral fat to subcutaneous sites resulted in increased gene transcript abundance in the grafts by 3 weeks post-transplantation, indicating the impact of local (residence) factors influencing epigenetic memory. We show here that active transcriptional state of adipokine genes is linked with glucose mediated recruitment of enzymes that regulate histone methylation. Adipose depots have "residence memory" and autologous transplantation of visceral fat to sub-cutaneous sites offers metabolic advantage.


Subject(s)
Adipose Tissue/transplantation , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue/metabolism , Animals , Body Composition , Female , Intra-Abdominal Fat , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...