Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










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.
Cell Mol Gastroenterol Hepatol ; 13(5): 1530-1553.e4, 2022.
Article in English | MEDLINE | ID: mdl-35032693

ABSTRACT

BACKGROUND & AIMS: Pancreatic islet ß-cells are factories for insulin production; however, ectopic expression of insulin also is well recognized. The gallbladder is a next-door neighbor to the developing pancreas. Here, we wanted to understand if gallbladders contain functional insulin-producing cells. METHODS: We compared developing and adult mouse as well as human gallbladder epithelial cells and islets using immunohistochemistry, flow cytometry, enzyme-linked immunosorbent assays, RNA sequencing, real-time polymerase chain reaction, chromatin immunoprecipitation, and functional studies. RESULTS: We show that the epithelial lining of developing, as well as adult, mouse and human gallbladders naturally contain interspersed cells that retain the capacity to actively transcribe, translate, package, and release insulin. We show that human gallbladders also contain functional insulin-secreting cells with the potential to naturally respond to glucose in vitro and in situ. Notably, in a non-obese diabetic (NOD) mouse model of type 1 diabetes, we observed that insulin-producing cells in the gallbladder are not targeted by autoimmune cells. Interestingly, in human gallbladders, insulin splice variants are absent, although insulin splice forms are observed in human islets. CONCLUSIONS: In summary, our biochemical, transcriptomic, and functional data in mouse and human gallbladder epithelial cells collectively show the evolutionary and developmental similarities between gallbladder and the pancreas that allow gallbladder epithelial cells to continue insulin production in adult life. Understanding the mechanisms regulating insulin transcription and translation in gallbladder epithelial cells would help guide future studies in type 1 diabetes therapy.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Epithelial Cells/metabolism , Gallbladder/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD
3.
iScience ; 24(4): 102379, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33981968

ABSTRACT

Dicer knockout mouse models demonstrated a key role for microRNAs in pancreatic ß-cell function. Studies to identify specific microRNA(s) associated with human (pro-)endocrine gene expression are needed. We profiled microRNAs and key pancreatic genes in 353 human tissue samples. Machine learning workflows identified microRNAs associated with (pro-)insulin transcripts in a discovery set of islets (n = 30) and insulin-negative tissues (n = 62). This microRNA signature was validated in remaining 261 tissues that include nine islet samples from individuals with type 2 diabetes. Top eight microRNAs (miR-183-5p, -375-3p, 216b-5p, 183-3p, -7-5p, -217-5p, -7-2-3p, and -429-3p) were confirmed to be associated with and predictive of (pro-)insulin transcript levels. Use of doxycycline-inducible microRNA-overexpressing human pancreatic duct cell lines confirmed the regulatory roles of these microRNAs in (pro-)endocrine gene expression. Knockdown of these microRNAs in human islet cells reduced (pro-)insulin transcript abundance. Our data provide specific microRNAs to further study microRNA-mRNA interactions in regulating insulin transcription.

4.
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
5.
Methods Protoc ; 3(2)2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32260112

ABSTRACT

Telomeres represent the nucleotide repeat sequences at the ends of chromosomes and are essential for chromosome stability. They can shorten at each round of DNA replication mainly because of incomplete DNA synthesis of the lagging strand. Reduced relative telomere length is associated with aging and a range of disease states. Different methods such as terminal restriction fragment analysis, real-time quantitative PCR (qPCR) and fluorescence in situ hybridization are available to measure telomere length; however, the qPCR-based method is commonly used for large population-based studies. There are multiple variations across qPCR-based methods, including the choice of the single-copy gene, primer sequences, reagents, and data analysis methods in the different reported studies so far. Here, we provide a detailed step-by-step protocol that we have optimized and successfully tested in the hands of other users. This protocol will help researchers interested in measuring relative telomere lengths in cells or across larger clinical cohort/study samples to determine associations of telomere length with health and disease.

6.
Gene ; 694: 97-101, 2019 Apr 30.
Article in English | MEDLINE | ID: mdl-30738962

ABSTRACT

BACKGROUND: Previous studies indicate that low birth weight and exposure to maternal stress during pregnancy may result in shortened telomeres in infants. Shorter telomere length has in turn been linked with accelerated ageing and with age-related diseases. This study aimed to investigate the association between pregnancy and birth factors and relative telomere length in offspring at 11 years of age. METHODS: Participants were aged 11 years enrolled in the Auckland Birthweight Collaborative Study at birth (n = 380). Half of the children were born small for gestational age (SGA = birthweight ≤ 10th percentile) and half were appropriate for gestational age (AGA = birthweight > 10th percentile). Maternal stress during pregnancy was assessed using the Perceived Stress Scale. Relative leukocyte telomere length (RTL) in leukocytes was measured at 11 years of age using quantitative real-time PCR. RESULTS: RTL was normally distributed (mean = 3.78, SD = 1.05). There were no significant associations between RTL at age 11 years and birthweight, sex, maternal smoking, maternal stress during pregnancy or maternal pre-pregnancy body mass index. CONCLUSION: At age 11 years, RTL did not differ between children by birthweight or pregnancy-related stressors. Further telomere-related studies in newborns, children and adolescents are merited to increase knowledge of potential telomere modulating factors.


Subject(s)
Birth Weight/genetics , Stress, Psychological/genetics , Telomere Homeostasis/genetics , Adult , Birth Weight/physiology , Body Mass Index , Child , Female , Gestational Age , Humans , Infant, Newborn , Infant, Small for Gestational Age/metabolism , Leukocytes , Male , Maternal Inheritance/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Stress, Psychological/metabolism , Telomere/genetics , Telomere Shortening/genetics
7.
Cell Transplant ; 28(1): 89-104, 2019 01.
Article in English | MEDLINE | ID: mdl-30376726

ABSTRACT

Type 1 diabetes (T1D) is characterized by the loss of insulin-producing ß-cells in the pancreas. T1D can be treated using cadaveric islet transplantation, but this therapy is severely limited by a lack of pancreas donors. To develop an alternative cell source for transplantation therapy, we carried out the epigenetic characterization in nine different adult mouse tissues and identified visceral adipose-derived progenitors as a candidate cell population. Chromatin conformation, assessed using chromatin immunoprecipitation (ChIP) sequencing and validated by ChIP-polymerase chain reaction (PCR) at key endocrine pancreatic gene promoters, revealed similarities between visceral fat and endocrine pancreas. Multiple techniques involving quantitative PCR, in-situ PCR, confocal microscopy, and flow cytometry confirmed the presence of measurable (2-1000-fold over detectable limits) pancreatic gene transcripts and mesenchymal progenitor cell markers (CD73, CD90 and CD105; >98%) in visceral adipose tissue-derived mesenchymal cells (AMCs). The differentiation potential of AMCs was explored in transgenic reporter mice expressing green fluorescent protein (GFP) under the regulation of the Pdx1 (pancreatic and duodenal homeobox-1) gene promoter. GFP expression was measured as an index of Pdx1 promoter activity to optimize culture conditions for endocrine pancreatic differentiation. Differentiated AMCs demonstrated their capacity to induce pancreatic endocrine genes as evidenced by increased GFP expression and validated using TaqMan real-time PCR (at least 2-200-fold relative to undifferentiated AMCs). Human AMCs differentiated using optimized protocols continued to produce insulin following transplantation in NOD/SCID mice. Our studies provide a systematic analysis of potential islet progenitor populations using genome-wide profiling studies and characterize visceral adipose-derived cells for replacement therapy in diabetes.


Subject(s)
Epigenesis, Genetic/genetics , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Flow Cytometry , Gene Expression Profiling/methods , Humans , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Stem Cells/cytology , Stem Cells/metabolism
8.
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
9.
Methods Mol Biol ; 1194: 271-88, 2014.
Article in English | MEDLINE | ID: mdl-25064109

ABSTRACT

It is well known that human cells are diverse with respect to their epigenome, transcriptome, and proteome. In the context of regenerative medicine, it is important for the transplanted cells or tissues to faithfully recapitulate their intended tissue type in each of these respects. Whether the cells chosen for such an application are embryonic, postnatal, or induced pluripotent stem cells, the transplanted product must behave in a predictable and reliable manner to be a safe and effective treatment option. Irrespective of the choice of cells used in such an application, the characterization and understanding of the developmental cues responsible for establishing and maintaining the desired cell phenotype are essential.Animal models are extremely important in understanding the development of a specific tissue, which can then be subsequently extrapolated to human studies. Generation of transgenic animal models with whole-body gene knockout, conditional knockout, constitutive fluorescent gene reporters, and Cre-Lox-based conditional and lineage reporters has revolutionized the field of developmental biology. An intrinsically complex network of the actions and interactions of the multitude of different signalling cascades is required for development. A thorough understanding of such networks, gained through studies on transgenic animal models, is essential for the development of the techniques necessary to reliably differentiate a given stem or progenitor cell population into a specific cell type, such as an islet-like, insulin-producing cell aggregate.In this chapter, we describe the use of GFP (green fluorescent protein)-based reporter mice for isolation of cells of choice, analyzing gene expression in those cells as well as their use for screening signalling molecules to understand their effect on differentiation.


Subject(s)
Cell Differentiation , Cell Separation/methods , Homeodomain Proteins/genetics , Pancreas/cytology , Stem Cells/cytology , Trans-Activators/genetics , Animals , Cloning, Molecular , DNA, Complementary/genetics , Embryo, Mammalian/cytology , Female , Genes, Reporter/genetics , Male , Mice , Mice, Transgenic , Phenotype , Polymerase Chain Reaction , Pregnancy , Taq Polymerase/metabolism
10.
Methods Mol Biol ; 1194: 449-69, 2014.
Article in English | MEDLINE | ID: mdl-25064120

ABSTRACT

The mammalian gut is inhabited by a complex and highly diverse population of bacteria. About 100 trillion microbes are present in the human gut, a number ten times more than the total number of cells in an adult human body. These microorganisms play an important role in several fundamental and crucial processes such as immunity, digestion, synthesis of vitamins, and metabolizing bile acids, sterols, and xenobiotics in the host, thereby influencing human health. Identification and manipulation of these metabolic interfaces is therefore critical. Here, we present a set of methods for manipulation and targeting the 16S rRNA based identification of rodent gut microbiota using Sanger's and next-generation sequencing platforms. Novel methods for manipulation of gut microbiota are also presented. In principle, these methods can be easily adapted to most rodent models for successful screening and manipulation of gut microbiome, to generate a better understanding of their role in metabolic disease.


Subject(s)
Bacteria/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Microbiota , Animals , Bacteria/genetics , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Feces/microbiology , Mice , Microbiota/genetics , Microbiota/physiology , Polymerase Chain Reaction , RNA, Ribosomal, 16S/genetics , Rats , Sequence Analysis, DNA
12.
Exp Diabetes Res ; 2012: 168368, 2012.
Article in English | MEDLINE | ID: mdl-22811698

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that act as regulators of gene expression by targeting mature messenger RNAs. Following the initial report of the presence of miRNAs in serum and plasma a number of studies have successfully demonstrated the use of these miRNAs as biomarkers of disease. Currently, there are many methods of isolating total RNA from liquid samples. Here, we describe a simple, cost effective method for extraction of RNA from human serum as well as subsequent real time PCR analysis of miRNA levels.


Subject(s)
Gene Expression Regulation , Genetic Techniques , RNA, Untranslated/blood , RNA, Untranslated/metabolism , Real-Time Polymerase Chain Reaction/methods , Serum/metabolism , Biomarkers/metabolism , Humans , MicroRNAs/analysis , MicroRNAs/blood , MicroRNAs/metabolism , RNA, Messenger/metabolism , RNA, Untranslated/analysis
13.
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
...