Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Animals (Basel) ; 12(5)2022 Mar 06.
Article in English | MEDLINE | ID: mdl-35268228

ABSTRACT

Visual leopard identifications performed with camera traps using the capture-recapture method only consider areas of the skin that are visible to the equipment. The method presented here considered the spot or rosette formations of either the two flanks or the face, and the captured images were then compared and matched with available photographs. Leopards were classified as new individuals if no matches were found in the existing set of photos. It was previously assumed that an individual leopard's spot or rosette pattern would not change. We established that the spot and rosette patterns change over time and that these changes are the result of injuries in certain cases. When compared to the original patterns, the number of spots may be lost or reduced, and some spots or patterns may change in terms of their prominence, shape, and size. We called these changes "obliterate changes" and "rejig changes", respectively. The implementation of an earlier method resulted in a duplication of leopard counts, achieving an error rate of more than 15% in the population at Yala National Park. The same leopard could be misidentified and counted multiple times, causing overestimated populations. To address this issue, we created a new two-step methodology for identifying Sri Lankan leopards. The multi-point identification method requires the evaluation of at least 9-10 spot areas before a leopard can be identified. Moreover, the minimum leopard population at the YNP 1 comprises at least 77 leopards and has a density of 0.5461 leopards per km2.

2.
Genes Dev ; 35(3-4): 234-249, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33446570

ABSTRACT

The physiological functions of many vital tissues and organs continue to mature after birth, but the genetic mechanisms governing this postnatal maturation remain an unsolved mystery. Human pancreatic ß cells produce and secrete insulin in response to physiological cues like glucose, and these hallmark functions improve in the years after birth. This coincides with expression of the transcription factors SIX2 and SIX3, whose functions in native human ß cells remain unknown. Here, we show that shRNA-mediated SIX2 or SIX3 suppression in human pancreatic adult islets impairs insulin secretion. However, transcriptome studies revealed that SIX2 and SIX3 regulate distinct targets. Loss of SIX2 markedly impaired expression of genes governing ß-cell insulin processing and output, glucose sensing, and electrophysiology, while SIX3 loss led to inappropriate expression of genes normally expressed in fetal ß cells, adult α cells, and other non-ß cells. Chromatin accessibility studies identified genes directly regulated by SIX2. Moreover, ß cells from diabetic humans with impaired insulin secretion also had reduced SIX2 transcript levels. Revealing how SIX2 and SIX3 govern functional maturation and maintain developmental fate in native human ß cells should advance ß-cell replacement and other therapeutic strategies for diabetes.


Subject(s)
Cell Differentiation/genetics , Eye Proteins/metabolism , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Nerve Tissue Proteins/metabolism , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin Secretion/genetics , RNA, Small Interfering/metabolism , Transcriptome , Homeobox Protein SIX3
3.
Development ; 147(6)2020 03 30.
Article in English | MEDLINE | ID: mdl-32108026

ABSTRACT

Reliance on rodents for understanding pancreatic genetics, development and islet function could limit progress in developing interventions for human diseases such as diabetes mellitus. Similarities of pancreas morphology and function suggest that porcine and human pancreas developmental biology may have useful homologies. However, little is known about pig pancreas development. To fill this knowledge gap, we investigated fetal and neonatal pig pancreas at multiple, crucial developmental stages using modern experimental approaches. Purification of islet ß-, α- and δ-cells followed by transcriptome analysis (RNA-seq) and immunohistology identified cell- and stage-specific regulation, and revealed that pig and human islet cells share characteristic features that are not observed in mice. Morphometric analysis also revealed endocrine cell allocation and architectural similarities between pig and human islets. Our analysis unveiled scores of signaling pathways linked to native islet ß-cell functional maturation, including evidence of fetal α-cell GLP-1 production and signaling to ß-cells. Thus, the findings and resources detailed here show how pig pancreatic islet studies complement other systems for understanding the developmental programs that generate functional islet cells, and that are relevant to human pancreatic diseases.


Subject(s)
Cell Differentiation/genetics , Insulin-Secreting Cells/physiology , Islets of Langerhans/embryology , Islets of Langerhans/growth & development , Swine , Animals , Animals, Newborn , Cells, Cultured , Embryo, Mammalian , Female , Fetus/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/physiology , Humans , Islets of Langerhans/cytology , Mice , Organogenesis/genetics , Pregnancy , Swine/embryology , Swine/genetics , Swine/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
4.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30389725

ABSTRACT

Increasing non-shivering thermogenesis (NST), which expends calories as heat rather than storing them as fat, is championed as an effective way to combat obesity and metabolic disease. Innate mechanisms constraining the capacity for NST present a fundamental limitation to this approach, yet are not well understood. Here, we provide evidence that Regulator of Calcineurin 1 (RCAN1), a feedback inhibitor of the calcium-activated protein phosphatase calcineurin (CN), acts to suppress two distinctly different mechanisms of non-shivering thermogenesis (NST): one involving the activation of UCP1 expression in white adipose tissue, the other mediated by sarcolipin (SLN) in skeletal muscle. UCP1 generates heat at the expense of reducing ATP production, whereas SLN increases ATP consumption to generate heat. Gene expression profiles demonstrate a high correlation between Rcan1 expression and metabolic syndrome. On an evolutionary timescale, in the context of limited food resources, systemic suppression of prolonged NST by RCAN1 might have been beneficial; however, in the face of caloric abundance, RCAN1-mediated suppression of these adaptive avenues of energy expenditure may now contribute to the growing epidemic of obesity.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Metabolism , Muscle Proteins/metabolism , Thermogenesis , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adrenergic Agents/pharmacology , Animals , Calcineurin/metabolism , Calcium-Binding Proteins , Cell Differentiation/drug effects , Cold Temperature , Female , Insulin Resistance , Intracellular Signaling Peptides and Proteins/deficiency , Lipid Metabolism/drug effects , Liver/metabolism , Male , Metabolic Syndrome/metabolism , Metabolism/drug effects , Mice , Mice, Knockout , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Striated/metabolism , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Promoter Regions, Genetic/genetics , Proteolipids/genetics , Proteolipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism
5.
Nat Commun ; 9(1): 3855, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242153

ABSTRACT

Developing systems to identify the cell type-specific functions regulated by genes linked to type 2 diabetes (T2D) risk could transform our understanding of the genetic basis of this disease. However, in vivo systems for efficiently discovering T2D risk gene functions relevant to human cells are currently lacking. Here we describe powerful interdisciplinary approaches combining Drosophila genetics and physiology with human islet biology to address this fundamental gap in diabetes research. We identify Drosophila orthologs of T2D-risk genes that regulate insulin output. With human islets, we perform genetic studies and identify cognate human T2D-risk genes that regulate human beta cell function. Loss of BCL11A, a transcriptional regulator, in primary human islet cells leads to enhanced insulin secretion. Gene expression profiling reveals BCL11A-dependent regulation of multiple genes involved in insulin exocytosis. Thus, genetic and physiological systems described here advance the capacity to identify cell-specific T2D risk gene functions.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Genes, Insect , Islets of Langerhans/metabolism , Nuclear Proteins/metabolism , Animals , Drosophila , Exocytosis , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Insulin Secretion/genetics , Repressor Proteins , Risk Assessment
6.
Clin Exp Pharmacol Physiol ; 45(4): 377-383, 2018 04.
Article in English | MEDLINE | ID: mdl-29094385

ABSTRACT

The regulator of calcineurin 1 (RCAN1) was first discovered as a gene located on human chromosome 21, expressed in neurons and overexpressed in the brains of Down syndrome individuals. Increased expression of RCAN1 has been linked with not only Down syndrome-associated pathology but also an associated neurological disorder, Alzheimer's Disease, in which neuronal RCAN1 expression is also increased. RCAN1 has additionally been demonstrated to affect other cell types including endocrine cells, with links to the pathogenesis of ß-cell dysfunction in type 2 diabetes. The primary functions of RCAN1 relate to the inhibition of the phosphatase calcineurin, and to the regulation of mitochondrial function. Various forms of cellular stress such as reactive oxygen species and hyperglycaemia cause transient increases in RCAN1 expression. The short term (hours to days) induction of RCAN1 expression is generally thought to have a protective effect by regulating the expression of pro-survival genes in multiple cell types, many of which are mediated via the calcineurin/NFAT transcriptional pathway. However, strong evidence also supports the notion that chronic (weeks-years) overexpression of RCAN1 has a detrimental effect on cells and that this may drive pathophysiological changes in neurons and endocrine cells linked to Down syndrome, Alzheimer's Disease and type 2 diabetes. Here we review the evidence related to these roles of RCAN1 in neurons and endocrine cells and their relationship to these human health disorders.


Subject(s)
Disease , Endocrine System/metabolism , Health , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Animals , Humans
7.
JCI Insight ; 2(20)2017 10 19.
Article in English | MEDLINE | ID: mdl-29046484

ABSTRACT

Cellular therapies based on permanent genetic modification of conventional T cells have emerged as a promising strategy for cancer. However, it remains unknown if modification of T cell subsets, such as Tregs, could be useful in other settings, such as allograft transplantation. Here, we use a modular system based on a chimeric antigen receptor (CAR) that binds covalently modified mAbs to control Treg activation in vivo. Transient expression of this mAb-directed CAR (mAbCAR) in Tregs permitted Treg targeting to specific tissue sites and mitigated allograft responses, such as graft-versus-host disease. mAbCAR Tregs targeted to MHC class I proteins on allografts prolonged islet allograft survival and also prolonged the survival of secondary skin grafts specifically matched to the original islet allograft. Thus, transient genetic modification to produce mAbCAR T cells led to durable immune modulation, suggesting therapeutic targeting strategies for controlling alloreactivity in settings such as organ or tissue transplantation.


Subject(s)
Immune Tolerance/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Animals , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Culture Techniques , Disease Models, Animal , Graft Rejection/prevention & control , Graft Survival/immunology , Histocompatibility Antigens Class I , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Transplantation , Receptors, Chimeric Antigen/genetics , STAT5 Transcription Factor , T-Lymphocytes, Regulatory/immunology , Tissue Transplantation , Transplantation Tolerance/immunology , Transplantation, Homologous
8.
Diabetes ; 66(5): 1301-1311, 2017 05.
Article in English | MEDLINE | ID: mdl-28174291

ABSTRACT

Pancreatic islet transplantation is a promising clinical treatment for type 1 diabetes, but success is limited by extensive ß-cell death in the immediate posttransplant period and impaired islet function in the longer term. Following transplantation, appropriate vascular remodeling is crucial to ensure the survival and function of engrafted islets. The sphingosine kinase (SK) pathway is an important regulator of vascular beds, but its role in the survival and function of transplanted islets is unknown. We observed that donor islets from mice deficient in SK1 (Sphk1 knockout) contain a reduced number of resident intraislet vascular endothelial cells. Furthermore, we demonstrate that the main product of SK1, sphingosine-1-phosphate, controls the migration of intraislet endothelial cells in vitro. We reveal in vivo that Sphk1 knockout islets have an impaired ability to cure diabetes compared with wild-type controls. Thus, SK1-deficient islets not only contain fewer resident vascular cells that participate in revascularization, but likely also a reduced ability to recruit new vessels into the transplanted islet. Together, our data suggest that SK1 is important for islet revascularization following transplantation and represents a novel clinical target for improving transplant outcomes.


Subject(s)
Cell Movement/genetics , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Endothelial Cells/cytology , Islets of Langerhans Transplantation , Islets of Langerhans/blood supply , Lysophospholipids/metabolism , Neovascularization, Physiologic/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine/analogs & derivatives , Animals , Flow Cytometry , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Sphingosine/metabolism , Transplants/blood supply
9.
Cell Metab ; 23(5): 909-20, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27133132

ABSTRACT

Intensive efforts are focused on identifying regulators of human pancreatic islet cell growth and maturation to accelerate development of therapies for diabetes. After birth, islet cell growth and function are dynamically regulated; however, establishing these age-dependent changes in humans has been challenging. Here, we describe a multimodal strategy for isolating pancreatic endocrine and exocrine cells from children and adults to identify age-dependent gene expression and chromatin changes on a genomic scale. These profiles revealed distinct proliferative and functional states of islet α cells or ß cells and histone modifications underlying age-dependent gene expression changes. Expression of SIX2 and SIX3, transcription factors without prior known functions in the pancreas and linked to fasting hyperglycemia risk, increased with age specifically in human islet ß cells. SIX2 and SIX3 were sufficient to enhance insulin content or secretion in immature ß cells. Our work provides a unique resource to study human-specific regulators of islet cell maturation and function.


Subject(s)
Aging/genetics , Gene Expression Regulation, Developmental , Insulin-Secreting Cells/metabolism , Adult , Cell Differentiation , Cell Separation , Child , Child, Preschool , Chromatin/metabolism , Chromatin Immunoprecipitation , Diabetes Mellitus/genetics , Histone Code , Homeodomain Proteins/metabolism , Humans , Infant , Middle Aged , Transcription Factors/metabolism , Transcriptome/genetics , Young Adult
10.
Diabetes ; 65(8): 2331-41, 2016 08.
Article in English | MEDLINE | ID: mdl-27217483

ABSTRACT

ß-Cell proliferation and expansion during pregnancy are crucial for maintaining euglycemia in response to increased metabolic demands placed on the mother. Prolactin and placental lactogen signal through the prolactin receptor (PRLR) and contribute to adaptive ß-cell responses in pregnancy; however, the in vivo requirement for PRLR signaling specifically in maternal ß-cell adaptations remains unknown. We generated a floxed allele of Prlr, allowing conditional loss of PRLR in ß-cells. In this study, we show that loss of PRLR signaling in ß-cells results in gestational diabetes mellitus (GDM), reduced ß-cell proliferation, and failure to expand ß-cell mass during pregnancy. Targeted PRLR loss in maternal ß-cells in vivo impaired expression of the transcription factor Foxm1, both G1/S and G2/M cyclins, tryptophan hydroxylase 1 (Tph1), and islet serotonin production, for which synthesis requires Tph1. This conditional system also revealed that PRLR signaling is required for the transient gestational expression of the transcription factor MafB within a subset of ß-cells during pregnancy. MafB deletion in maternal ß-cells also produced GDM, with inadequate ß-cell expansion accompanied by failure to induce PRLR-dependent target genes regulating ß-cell proliferation. These results unveil molecular roles for PRLR signaling in orchestrating the physiologic expansion of maternal ß-cells during pregnancy.


Subject(s)
Diabetes, Gestational/metabolism , Insulin-Secreting Cells/metabolism , MafB Transcription Factor/metabolism , Receptors, Prolactin/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Cyclin A2/genetics , Cyclin B1/genetics , Cyclin B2/genetics , Cyclin D1/genetics , Cyclin D2/genetics , Diabetes, Gestational/physiopathology , Female , Forkhead Box Protein M1/genetics , Insulin/metabolism , MafB Transcription Factor/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptors, Prolactin/genetics , Serotonin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
11.
PLoS Genet ; 12(5): e1006033, 2016 05.
Article in English | MEDLINE | ID: mdl-27195491

ABSTRACT

Type 2 diabetes (T2D) is a complex metabolic disease associated with obesity, insulin resistance and hypoinsulinemia due to pancreatic ß-cell dysfunction. Reduced mitochondrial function is thought to be central to ß-cell dysfunction. Mitochondrial dysfunction and reduced insulin secretion are also observed in ß-cells of humans with the most common human genetic disorder, Down syndrome (DS, Trisomy 21). To identify regions of chromosome 21 that may be associated with perturbed glucose homeostasis we profiled the glycaemic status of different DS mouse models. The Ts65Dn and Dp16 DS mouse lines were hyperglycemic, while Tc1 and Ts1Rhr mice were not, providing us with a region of chromosome 21 containing genes that cause hyperglycemia. We then examined whether any of these genes were upregulated in a set of ~5,000 gene expression changes we had identified in a large gene expression analysis of human T2D ß-cells. This approach produced a single gene, RCAN1, as a candidate gene linking hyperglycemia and functional changes in T2D ß-cells. Further investigations demonstrated that RCAN1 methylation is reduced in human T2D islets at multiple sites, correlating with increased expression. RCAN1 protein expression was also increased in db/db mouse islets and in human and mouse islets exposed to high glucose. Mice overexpressing RCAN1 had reduced in vivo glucose-stimulated insulin secretion and their ß-cells displayed mitochondrial dysfunction including hyperpolarised membrane potential, reduced oxidative phosphorylation and low ATP production. This lack of ß-cell ATP had functional consequences by negatively affecting both glucose-stimulated membrane depolarisation and ATP-dependent insulin granule exocytosis. Thus, from amongst the myriad of gene expression changes occurring in T2D ß-cells where we had little knowledge of which changes cause ß-cell dysfunction, we applied a trisomy 21 screening approach which linked RCAN1 to ß-cell mitochondrial dysfunction in T2D.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Down Syndrome/genetics , Insulin/genetics , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Adenosine Triphosphate/metabolism , Aneuploidy , Animals , Calcium-Binding Proteins , Chromosomes, Human, Pair 21/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Down Syndrome/metabolism , Down Syndrome/pathology , Gene Expression Regulation , Glucose/metabolism , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/pathology , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mitochondria/genetics , Mitochondria/pathology , Muscle Proteins/metabolism , Protein Biosynthesis/genetics
12.
Mol Endocrinol ; 30(2): 248-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26745668

ABSTRACT

The 2 most abundant human pancreatic islet cell types are insulin-producing ß-cells and glucagon-producing α-cells. Defined cis-regulatory elements from rodent Insulin genes have permitted genetic labeling of human islet ß-cells, enabling lineage tracing and generation of human ß-cell lines, but analogous elements for genetically labeling human α-cells with high specificity do not yet exist. To identify genetic elements that specifically direct reporter expression to human α-cells, we investigated noncoding sequences adjacent to the human GLUCAGON and ARX genes, which are expressed in islet α-cells. Elements with high evolutionary conservation were cloned into lentiviral vectors to direct fluorescent reporter expression in primary human islets. Based on the specificity of reporter expression for α- and ß-cells, we found that rat glucagon promoter was not specific for human α-cells but that addition of human GLUCAGON untranslated region sequences substantially enhanced specificity of labeling in both cultured and transplanted islets to a degree not previously reported, to our knowledge. Specific transgene expression from these cis-regulatory sequences in human α-cells should enable targeted genetic modification and lineage tracing.


Subject(s)
Genetic Techniques , Insulin-Secreting Cells/metabolism , Staining and Labeling , Animals , Base Sequence , Genetic Loci , HEK293 Cells , Humans , Islets of Langerhans Transplantation , Mice, SCID , Regulatory Sequences, Nucleic Acid/genetics
13.
Cell Transplant ; 24(1): 37-48, 2015.
Article in English | MEDLINE | ID: mdl-24069942

ABSTRACT

The success of pancreatic islet transplantation is limited by delayed engraftment and suboptimal function in the longer term. Endothelial progenitor cells (EPCs) represent a potential cellular therapy that may improve the engraftment of transplanted pancreatic islets. In addition, EPCs may directly affect the function of pancreatic ß-cells. The objective of this study was to examine the ability of EPCs to enhance pancreatic islet transplantation in a murine syngeneic marginal mass transplant model and to examine the mechanisms through which this occurs. We found that cotransplanted EPCs improved the cure rate and initial glycemic control of transplanted islets. Gene expression data indicate that EPCs, or their soluble products, modulate the expression of the ß-cell surface molecule connexin 36 and affect glucose-stimulated insulin release in vitro. In conclusion, EPCs are a promising candidate for improving outcomes in islet transplantation, and their mechanisms of action warrant further study.


Subject(s)
Connexins/biosynthesis , Endothelial Cells/metabolism , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Islets of Langerhans Transplantation , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Endothelial Cells/pathology , Endothelial Cells/transplantation , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Male , Mice , Stem Cells/pathology , Sweetening Agents/pharmacology , Transplantation, Isogeneic , Gap Junction delta-2 Protein
14.
Oxid Med Cell Longev ; 2014: 520316, 2014.
Article in English | MEDLINE | ID: mdl-25009690

ABSTRACT

Mitochondria are the primary site of cellular energy generation and reactive oxygen species (ROS) accumulation. Elevated ROS levels are detrimental to normal cell function and have been linked to the pathogenesis of neurodegenerative disorders such as Down's syndrome (DS) and Alzheimer's disease (AD). RCAN1 is abundantly expressed in the brain and overexpressed in brain of DS and AD patients. Data from nonmammalian species indicates that increased RCAN1 expression results in altered mitochondrial function and that RCAN1 may itself regulate neuronal ROS production. In this study, we have utilized mice overexpressing RCAN1 (RCAN1(ox)) and demonstrate an increased susceptibility of neurons from these mice to oxidative stress. Mitochondria from these mice are more numerous and smaller, indicative of mitochondrial dysfunction, and mitochondrial membrane potential is altered under conditions of oxidative stress. We also generated a PC12 cell line overexpressing RCAN1 (PC12(RCAN1)). Similar to RCAN1(ox) neurons, PC12(RCAN1) cells have an increased susceptibility to oxidative stress and produce more mitochondrial ROS. This study demonstrates that increasing RCAN1 expression alters mitochondrial function and increases the susceptibility of neurons to oxidative stress in mammalian cells. These findings further contribute to our understanding of RCAN1 and its potential role in the pathogenesis of neurodegenerative disorders such as AD and DS.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Muscle Proteins/metabolism , Oxidative Stress , Animals , Cell Survival/drug effects , DNA-Binding Proteins , Female , Hydrogen Peroxide/toxicity , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Models, Biological , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
15.
Diabetes ; 63(1): 3-11, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24357688

ABSTRACT

Within the pancreatic islet, the ß-cell represents the ultimate biosensor. Its central function is to accurately sense glucose levels in the blood and consequently release appropriate amounts of insulin. As the only cell type capable of insulin production, the ß-cell must balance this crucial workload with self-preservation and, when required, regeneration. Evidence suggests that the ß-cell has an important ally in intraislet endothelial cells (ECs). As well as providing a conduit for delivery of the primary input stimulus (glucose) and dissemination of its most important effector (insulin), intraislet blood vessels deliver oxygen to these dense clusters of metabolically active cells. Furthermore, it appears that ECs directly impact insulin gene expression and secretion and ß-cell survival. This review discusses the molecules and pathways involved in the crosstalk between ß-cells and intraislet ECs. The evidence supporting the intraislet EC as an important partner for ß-cell function is examined to highlight the relevance of this axis in the context of type 1 and type 2 diabetes. Recent work that has established the potential of ECs or their progenitors to enhance the re-establishment of glycemic control following pancreatic islet transplantation in animal models is discussed.


Subject(s)
Cell Communication/physiology , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Insulin-Secreting Cells/metabolism , Animals , Blood Glucose/metabolism , Endothelial Cells/cytology , Insulin/metabolism , Insulin-Secreting Cells/cytology
16.
J Physiol ; 592(7): 1505-18, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24366265

ABSTRACT

Huntingtin-associated protein 1 (HAP1) was initially established as a neuronal binding partner of huntingtin, mutations in which underlie Huntington's disease. Subcellular localization and protein interaction data indicate that HAP1 may be important in vesicle trafficking and cell signalling. In this study, we establish that HAP1 is important in several steps of exocytosis in adrenal chromaffin cells. Using carbon-fibre amperometry, we measured single vesicle exocytosis in chromaffin cells obtained from HAP1(-/-) and HAP1(+/+) littermate mice. Numbers of Ca(2+)-dependent and Ca(2+)-independent full fusion events in HAP1(-/-) cells are significantly decreased compared with those in HAP1(+/+) cells. We observed no change in the frequency of 'kiss-and-run' fusion events or in Ca(2+) entry. Whereas release per full fusion event is unchanged in HAP1(-/-) cells, early fusion pore duration is prolonged, as indicated by the increased duration of pre-spike foot signals. Kiss-and-run events have a shorter duration, indicating opposing roles for HAP1 in the stabilization of the fusion pore during full fusion and transient fusion, respectively. We use electron microscopy to demonstrate a reduction in the number of vesicles docked at the plasma membrane of HAP1(-/-) cells, where membrane capacitance measurements reveal the readily releasable pool of vesicles to be reduced in size. Our study therefore illustrates that HAP1 regulates exocytosis by influencing the morphological docking of vesicles at the plasma membrane, the ability of vesicles to be released rapidly upon stimulation, and the early stages of fusion pore formation.


Subject(s)
Adrenal Medulla/metabolism , Cell Membrane/metabolism , Chromaffin Cells/metabolism , Exocytosis , Membrane Fusion , Nerve Tissue Proteins/metabolism , Secretory Vesicles/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Catecholamines/metabolism , Cells, Cultured , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Secretory Pathway , Time Factors
17.
J Neurochem ; 124(3): 290-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23134420

ABSTRACT

We have previously shown that Regulator of Calcineurin 1 (RCAN1) regulates multiple stages of vesicle exocytosis. However, the mechanisms by which RCAN1 affects secretory vesicle exocytosis and quantal release kinetics remain unknown. Here, we use carbon fibre amperometry to detect exocytosis from chromaffin cells and identify these underlying mechanisms. We observe reduced exocytosis with repeated stimulations in chromaffin cells over-expressing RCAN1 (RCAN1(ox)), but not in wild-type (WT) cells, indicating a negative effect of RCAN1 on vesicle recycling and endocytosis. Acute exposure to calcineurin inhibitors, cyclosporine A and FK-506, replicates this effect in WT cells but has no additional effect in RCAN1(ox) cells. When we chronically expose WT cells to cyclosporine A and FK-506 we find that catecholamine release per vesicle and pre-spike foot (PSF) signal parameters are decreased, similar to that in RCAN1(ox) cells. Inhibiting calcineurin activity in RCAN1(ox) cells has no additional effect on the amount of catecholamine release per vesicle but further reduces PSF signal parameters. Although electron microscopy studies indicate these changes are not because of altered vesicle number or distribution in RCAN1(ox) cells, the smaller vesicle and dense core size we observe in RCAN1(ox) cells may underlie the reduced quantal release in these cells. Thus, our results indicate that RCAN1 most likely affects vesicle recycling and quantal release kinetics via the inhibition of calcineurin activity.


Subject(s)
Calcineurin/metabolism , Calcineurin/pharmacokinetics , Intracellular Signaling Peptides and Proteins/physiology , Muscle Proteins/physiology , Secretory Vesicles/metabolism , Animals , Calcineurin Inhibitors , Calcium-Binding Proteins , Cells, Cultured , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Chromaffin Cells/physiology , Endocytosis/physiology , Female , Kinetics , Male , Mice , Mice, Mutant Strains , Quantum Theory , Secretory Vesicles/physiology
18.
Endocrinology ; 153(11): 5212-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23011918

ABSTRACT

RCAN1 is a chromosome 21 gene that controls secretion in endocrine cells, regulates mitochondrial function, and is sensitive to oxidative stress. Regulator of calcineurin 1 (RCAN1) is also an endogenous inhibitor of the protein phosphatase calcineurin, the inhibition of which leads to hypoinsulinemia and diabetes in humans and mice. However, the presence or the role of RCAN1 in insulin-secreting ß-cells and its potential role in the pathogenesis of diabetes is unknown. Hence, the aim of this study is to investigate the presence of RCAN1 in ß-cells and identify its role in ß-cell function. RCAN1 is expressed in mouse islets and in the cytosol of pancreatic ß-cells. We find RCAN1 is a glucose-responsive gene with a 1.5-fold increase in expression observed in pancreatic islets in response to chronic hyperglycemia. The overexpression of the human RCAN1.1 isoform in mice under the regulation of its endogenous promoter causes diabetes, age-associated hyperglycemia, reduced glucose tolerance, hypoinsulinemia, loss of ß-cells, reduced ß-cell insulin secretion, aberrant mitochondrial reactive oxygen species production, and the down-regulation of key ß-cell genes. Our data therefore identifies a novel molecular link between the overexpression of RCAN1 and ß-cell dysfunction. The glucose-responsive nature of RCAN1 provides a potential mechanism of action associated with the ß-cell dysfunction observed in diabetes.


Subject(s)
Diabetes Mellitus/metabolism , Glucose Intolerance/metabolism , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Animals , Calcium-Binding Proteins , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Hyperglycemia/genetics , Hyperglycemia/pathology , Insulin Secretion , Insulin-Secreting Cells/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mitochondria/genetics , Mitochondria/metabolism , Muscle Proteins/genetics , Reactive Oxygen Species/metabolism
19.
J Physiol ; 589(Pt 7): 1691-706, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21320883

ABSTRACT

Endothelin peptides and their endogenous receptors play a major role in nociception in a variety of different organs. They also play an essential role in the development of the enteric nervous system. Mice with deletions of the endothelin-3 gene (lethal spotted mice, ls/ls) develop congenital aganglionosis. However, little is known about how nociception might be affected in the aganglionic rectum of mice deficient in endothelin-3. In this study we investigated changes in spinal afferent innervation and visceral pain transmission from the aganglionic rectum in ls/ls mice. Electromyogram recordings from anaesthetized ls/ls mice revealed a deficit in visceromotor responses arising from the aganglionic colorectum in response to noxious colorectal distension. Loss of visceromotor responses (VMRs) in ls/ls mice was selective, as no reduction in VMRs was detected after stimulation of the bladder or somatic organs. Calcitonin gene related peptide (CGRP) immunoreactivity, retrograde neuronal tracing and extracellular afferent recordings from the aganglionic rectum revealed decreased colorectal spinal innervation, combined with a reduction in mechanosensitivity of rectal afferents. The sensory defect in ls/ls mice is primarily associated with changes in low threshold wide dynamic range rectal afferents. In conclusion, disruption of endothelin 3 gene expression not only affects development and function of the enteric nervous system, but also specific classes of spinal rectal mechanoreceptors, which are required for visceral nociception from the colorectum.


Subject(s)
Endothelin-3/deficiency , Enteric Nervous System/physiopathology , Hirschsprung Disease/physiopathology , Pain/physiopathology , Afferent Pathways/physiopathology , Animals , Colon/innervation , Colon/physiopathology , Compliance/physiology , Dilatation, Pathologic , Disease Models, Animal , Electric Stimulation , Endothelin-3/genetics , Enteric Nervous System/abnormalities , Enteric Nervous System/pathology , Hirschsprung Disease/etiology , Humans , Mechanoreceptors/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nociceptors/physiology , Rectum/innervation , Rectum/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...