Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 17(4): 992-1007, 2017 04.
Article in English | MEDLINE | ID: mdl-27700000

ABSTRACT

Currently there is no effective approach for monitoring early ß-cell loss during islet graft rejection following human islet transplantation (HIT). Due to ethical and technical constraints, it is difficult to directly study biomarkers of islet destruction in humans. Here, we established a humanized mouse model with induced human ß-cell death using adoptive lymphocyte transfer (ALT). Human islet grafts of ALT-treated mice had perigraft lymphocyte infiltration, fewer insulin+ ß cells, and increased ß-cell apoptosis. Islet-specific miR-375 was used to validate our model, and expression of miR-375 was significantly decreased in the grafts and increased in the circulation of ALT-treated mice before hyperglycemia. A NanoString expression assay was further used to profile 800 human miRNAs in the human islet grafts, and the results were validated using quantitative real-time polymerase chain reaction. We found that miR-4454 and miR-199a-5p were decreased in the human islet grafts following ALT and increased in the circulation prior to hyperglycemia. These data demonstrate that our in vivo model of induced human ß-cell destruction is a robust method for identifying and characterizing circulating biomarkers, and suggest that miR-4454 and miR-199a-5p can serve as novel biomarkers associated with early human ß-cell loss following HIT.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , MicroRNAs/genetics , Adoptive Transfer , Animals , Apoptosis , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Female , Graft Survival , Humans , Hyperglycemia/etiology , Insulin-Secreting Cells/metabolism , Lymphocytes/immunology , Lymphocytes/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID
2.
Diabetes Obes Metab ; 18 Suppl 1: 41-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27615130

ABSTRACT

Pancreatic ß-cells are highly specialized cells committed to secrete insulin in response to changes in the level of nutrients, hormones and neurotransmitters. Chronic exposure to elevated concentrations of glucose, fatty acids or inflammatory mediators can result in modifications in ß-cell gene expression that alter their functional properties. This can lead to the release of insufficient amount of insulin to cover the organism's needs, and thus to the development of diabetes mellitus. Although most of the studies carried out in the last decades to elucidate the causes of ß-cell dysfunction under disease conditions have focused on protein-coding genes, we now know that insulin-secreting cells also contain thousands of molecules of RNA that do not encode polypeptides but play key roles in the acquisition and maintenance of a highly differentiated state. In this review, we will highlight the involvement of long non-coding RNAs (lncRNAs), a particular class of non-coding transcripts, in the differentiation of ß-cells and in the regulation of their specialized tasks. We will also discuss the crosstalk between the activities of lncRNAs and microRNAs and present the emerging evidence of a potential contribution of particular lncRNAs to the development of both type 1 and type 2 diabetes.


Subject(s)
Cell Differentiation/genetics , Diabetes Mellitus/genetics , Insulin-Secreting Cells/cytology , RNA, Long Noncoding/genetics , Animals , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice
3.
Diabetes Metab ; 41(5): 369-77, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25910875

ABSTRACT

Insulin secretion from pancreatic ß cells plays a central role in the control of blood glucose levels. The amount of insulin released by ß cells is precisely adjusted to match organism requirements. A number of conditions that arise during life, including pregnancy and obesity, can result in a decreased sensitivity of insulin target tissues and a consequent rise in insulin needs. To preserve glucose homoeostasis, the augmented insulin demand requires a compensatory expansion of the pancreatic ß cell mass and an increase in its secretory activity. This compensatory process is accompanied by modifications in ß cell gene expression, although the molecular mechanisms underlying the phenomenon are still poorly understood. Emerging evidence indicates that at least part of these compensatory events may be orchestrated by changes in the level of a novel class of gene regulators, the microRNAs. Indeed, several of these small, non-coding RNAs have either positive or negative impacts on ß cell proliferation and survival. The studies reviewed here suggest that the balance between the actions of these two groups of microRNAs, which have opposing functional effects, can determine whether ß cells expand sufficiently to maintain blood glucose levels in the normal range or fail to meet insulin demand and thus lead, as a consequence, towards diabetes manifestation. A better understanding of the mechanisms governing changes in the microRNA profile will open the way for the development of new strategies to prevent and/or treat both type 2 and gestational diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Models, Biological , Prediabetic State/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Diabetes Mellitus, Type 2/pathology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Prediabetic State/pathology
4.
Diabetes Obes Metab ; 14 Suppl 3: 12-21, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22928560

ABSTRACT

Pancreatic ß-cells play a central role in glucose homeostasis by tightly regulating insulin release according to the organism's demand. Impairment of ß-cell function due to hostile environment, such as hyperglycaemia and hyperlipidaemia, or due to autoimmune destruction of ß-cells, results in diabetes onset. Both environmental factors and genetic predisposition are known to be involved in the development of the disease, but the exact mechanisms leading to ß-cell dysfunction and death remain to be characterized. Non-coding RNA molecules, such as microRNAs (miRNAs), have been suggested to be necessary for proper ß-cell development and function. The present review aims at summarizing the most recent findings about the role of non-coding RNAs in the control of ß-cell functions and their involvement in diabetes. We will also provide a perspective view of the future research directions in the field of non-coding RNAs. In particular, we will discuss the implications for diabetes research of the discovery of a new communication mechanism based on cell-to-cell miRNA transfer. Moreover, we will highlight the emerging interconnections between miRNAs and epigenetics and the possible role of long non-coding RNAs in the control of ß-cell activities.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Insulin-Secreting Cells/metabolism , MicroRNAs/genetics , RNA, Untranslated/metabolism , Cell Differentiation , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Gene Silencing , Homeostasis/genetics , Humans , Male , RNA, Untranslated/genetics
5.
Diabetologia ; 55(6): 1699-708, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22159912

ABSTRACT

AIMS/HYPOTHESIS: Chronic exposure of pancreatic beta cells to proinflammatory cytokines leads to impaired insulin secretion and apoptosis. ARE/poly(U)-binding factor 1 (AUF1) belongs to a protein family that controls mRNA stability and translation by associating with adenosine- and uridine-rich regions of target messengers. We investigated the involvement of AUF1 in cytokine-induced beta cell dysfunction. METHODS: Production and subcellular distribution of AUF1 isoforms were analysed by western blotting. To test for their role in the control of beta cell functions, each isoform was overproduced individually in insulin-secreting cells. The contribution to cytokine-mediated beta cell dysfunction was evaluated by preventing the production of AUF1 isoforms by RNA interference. The effect of AUF1 on the production of potential targets was assessed by western blotting. RESULTS: MIN6 cells and human pancreatic islets were found to produce four AUF1 isoforms (p42>p45>p37>p40). AUF1 isoforms were mainly localised in the nucleus but were partially translocated to the cytoplasm upon exposure of beta cells to cytokines and activation of the ERK pathway. Overproduction of AUF1 did not affect glucose-induced insulin secretion but promoted apoptosis. This effect was associated with a decrease in the production of the anti-apoptotic proteins, B cell leukaemia/lymphoma 2 (BCL2) and myeloid cell leukaemia sequence 1 (MCL1). Silencing of AUF1 isoforms restored the levels of the anti-apoptotic proteins, attenuated the activation of the nuclear factor-κB (NFκB) pathway, and protected the beta cells from cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION: Our findings point to a contribution of AUF1 to the deleterious effects of cytokines on beta cell functions and suggest a role for this RNA-binding protein in the early phases of type 1 diabetes.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Islets of Langerhans/metabolism , Protein Isoforms/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Humans , Immunohistochemistry , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Islets of Langerhans/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Protein Isoforms/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference
6.
Diabetologia ; 54(9): 2337-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21547497

ABSTRACT

AIMS/HYPOTHESIS: Pro-atherogenic and pro-oxidant, oxidised LDL trigger adverse effects on pancreatic beta cells, possibly contributing to diabetes progression. Because oxidised LDL diminish the expression of genes regulated by the inducible cAMP early repressor (ICER), we investigated the involvement of this transcription factor and of oxidative stress in beta cell failure elicited by oxidised LDL. METHODS: Isolated human and rat islets, and insulin-secreting cells were cultured with human native or oxidised LDL or with hydrogen peroxide. The expression of genes was determined by quantitative real-time PCR and western blotting. Insulin secretion was monitored by EIA kit. Cell apoptosis was determined by scoring cells displaying pycnotic nuclei. RESULTS: Exposure of beta cell lines and islets to oxidised LDL, but not to native LDL raised the abundance of ICER. Induction of this repressor by the modified LDL compromised the expression of important beta cell genes, including insulin and anti-apoptotic islet brain 1, as well as of genes coding for key components of the secretory machinery. This led to hampering of insulin production and secretion, and of cell survival. Silencing of this transcription factor by RNA interference restored the expression of its target genes and alleviated beta cell dysfunction and death triggered by oxidised LDL. Induction of ICER was stimulated by oxidative stress, whereas antioxidant treatment with N-acetylcysteine or HDL prevented the rise of ICER elicited by oxidised LDL and restored beta cell functions. CONCLUSIONS/INTERPRETATION: Induction of ICER links oxidative stress to beta cell failure caused by oxidised LDL and can be effectively abrogated by antioxidant treatment.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiopathology , Oxidative Stress/physiology , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cyclic AMP Response Element Modulator/drug effects , Cyclic AMP Response Element Modulator/genetics , Humans , Hydrogen Peroxide/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Lipoproteins, LDL/pharmacology , Male , Models, Animal , Oxidative Stress/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
7.
Diabetes Obes Metab ; 11 Suppl 4: 118-29, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19817794

ABSTRACT

The discovery in mammalian cells of hundreds of small RNA molecules, called microRNAs, with the potential to modulate the expression of the majority of the protein-coding genes has revolutionized many areas of biomedical research, including the diabetes field. MicroRNAs function as translational repressors and are emerging as key regulators of most, if not all, physiological processes. Moreover, alterations in the level or function of microRNAs are associated with an increasing number of diseases. Here, we describe the mechanisms governing the biogenesis and activities of microRNAs. We present evidence for the involvement of microRNAs in diabetes mellitus, by outlining the contribution of these small RNA molecules in the control of pancreatic beta-cell functions and by reviewing recent studies reporting changes in microRNA expression in tissues isolated from diabetes animal models. MicroRNAs hold great potential as therapeutic targets. We describe the strategies developed for the delivery of molecules mimicking or blocking the function of these tiny regulators of gene expression in living animals. In addition, because changes in serum microRNA profiles have been shown to occur in association with different human diseases, we also discuss the potential use of microRNAs as blood biomarkers for prevention and management of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , MicroRNAs/physiology , Animals , Diabetes Mellitus, Type 2/diet therapy , Gene Expression Regulation , Humans , Mice , MicroRNAs/biosynthesis
8.
Diabetologia ; 51(8): 1429-39, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18385973

ABSTRACT

AIMS/HYPOTHESIS: The expression of several neuronal genes in pancreatic beta cells is due to the absence of the transcription factor repressor element 1 (RE-1) silencing transcription factor (REST). The identification of these traits and their functional significance in beta cells has only been partly elucidated. Herein, we investigated the biological consequences of a repression of REST target genes by expressing REST in beta cells. METHODS: The effect of REST expression on glucose homeostasis, insulin content and release, and beta cell mass was analysed in transgenic mice selectively expressing REST in beta cells. Relevant target genes were identified in INS-1E and primary beta cells expressing REST. RESULTS: Transgenic mice featuring a beta cell-targeted expression of REST exhibited glucose intolerance and reduced beta cell mass. In primary beta cells, REST repressed several proteins of the exocytotic machinery, including synaptosomal-associated protein (SNAP) 25, synaptotagmin (SYT) IV, SYT VII, SYT IX and complexin II; it impaired first and second phases of insulin secretion. Using RNA interference in INS-1E cells, we showed that SYT IV and SYT VII were implicated in the control of insulin release. CONCLUSIONS/INTERPRETATION: The data document the critical role of REST target genes in pancreatic beta cells. Specifically, we provide evidence that the downregulation of these genes is detrimental for the exocytosis of large dense core vesicles, thus contributing to beta cell dysfunction and impaired glucose homeostasis.


Subject(s)
Gene Silencing , Insulin-Secreting Cells/physiology , Repressor Proteins/genetics , Animals , Chromatin/physiology , Consensus Sequence , Cytosol/physiology , Genes, Reporter , Glucose/metabolism , Glucose Tolerance Test , Humans , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Membrane Potentials , Mice , Mice, Transgenic , Mitochondrial Membranes/physiology , Repressor Proteins/physiology
9.
Diabetologia ; 50(6): 1304-14, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17437081

ABSTRACT

AIMS/HYPOTHESIS: We explored the potential adverse effects of pro-atherogenic oxidised LDL-cholesterol particles on beta cell function. MATERIALS AND METHODS: Isolated human and rat islets and different insulin-secreting cell lines were incubated with human oxidised LDL with or without HDL particles. The insulin level was monitored by ELISA, real-time PCR and a rat insulin promoter construct linked to luciferase gene reporter. Cell apoptosis was determined by scoring cells displaying pycnotic nuclei. RESULTS: Prolonged incubation with human oxidised LDL particles led to a reduction in preproinsulin expression levels, whereas the insulin level was preserved in the presence of native LDL-cholesterol. The loss of insulin production occurred at the transcriptional levels and was associated with an increase in activator protein-1 transcriptional activity. The rise in activator protein-1 activity resulted from activation of c-Jun N-terminal kinases (JNK, now known as mitogen-activated protein kinase 8 [MAPK8]) due to a subsequent decrease in islet-brain 1 (IB1; now known as MAPK8 interacting protein 1) levels. Consistent with the pro-apoptotic role of the JNK pathway, oxidised LDL also induced a twofold increase in the rate of beta cell apoptosis. Treatment of the cells with JNK inhibitor peptides or HDL countered the effects mediated by oxidised LDL. CONCLUSIONS/INTERPRETATION: These data provide strong evidence that oxidised LDL particles exert deleterious effects in the progression of beta cell failure in diabetes and that these effects can be countered by HDL particles.


Subject(s)
Insulin-Secreting Cells/enzymology , Insulin/genetics , Lipoproteins, HDL/pharmacology , Lipoproteins, LDL/pharmacology , MAP Kinase Kinase 4/metabolism , Animals , Apoptosis , Cell Line , Diabetes Mellitus/enzymology , Disease Progression , Enzyme Activation , Genes, Reporter , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , MAP Kinase Kinase 4/antagonists & inhibitors , Male , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA/genetics , RNA/isolation & purification , Rats , Rats, Sprague-Dawley
10.
Biochem Soc Trans ; 34(Pt 5): 696-700, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17052177

ABSTRACT

The last decade has witnessed spectacular progress in the identification of the protein apparatus required for exocytosis of neurotransmitters, peptide hormones and other bioactive products. In striking contrast, our knowledge of the mechanisms determining the expression of the components of the secretory machinery has remained rudimentary. Since modifications in secretory functions are associated with several physiological processes and contribute to the development of human pathologies, a better knowledge of the control of the expression of the genes involved in exocytosis is urgently needed. Recent studies have led to the identification of transcription factors and other regulatory molecules such as microRNAs that modulate the cellular level of key controllers of the exocytotic process. These findings furnish a new perspective for understanding how secretory functions can adapt to normal physiological conditions and shed light on the mechanisms involved in the development of important human diseases such as diabetes mellitus characterized by defective release of bioactive compounds.


Subject(s)
Exocytosis , Gene Expression Regulation/physiology , Animals , MicroRNAs/genetics , RNA, Messenger/genetics , SNARE Proteins/genetics , Transcription Factors/metabolism , rab GTP-Binding Proteins/genetics
11.
Cell Motil Cytoskeleton ; 62(2): 124-32, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16145686

ABSTRACT

The cytoskeleton, composed of actin filaments, intermediate filaments, and microtubules, is a highly dynamic supramolecular network actively involved in many essential biological mechanisms such as cellular structure, transport, movements, differentiation, and signaling. As a first step to characterize the biophysical changes associated with cytoskeleton functions, we have developed finite elements models of the organization of the cell that has allowed us to interpret atomic force microscopy (AFM) data at a higher resolution than that in previous work. Thus, by assuming that living cells behave mechanically as multilayered structures, we have been able to identify superficial and deep effects that could be related to actin and microtubule disassembly, respectively. In Cos-7 cells, actin destabilization with Cytochalasin D induced a decrease of the visco-elasticity close to the membrane surface, while destabilizing microtubules with Nocodazole produced a stiffness decrease only in deeper parts of the cell. In both cases, these effects were reversible. Cell softening was measurable with AFM at concentrations of the destabilizing agents that did not induce detectable effects on the cytoskeleton network when viewing the cells with fluorescent confocal microscopy. All experimental results could be simulated by our models. This technology opens the door to the study of the biophysical properties of signaling domains extending from the cell surface to deeper parts of the cell.


Subject(s)
Cytoskeleton/physiology , Actins/antagonists & inhibitors , Animals , Biomechanical Phenomena , COS Cells , Chlorocebus aethiops , Computer Simulation , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Genes, Reporter , Microscopy, Confocal , Microtubules/drug effects , Microtubules/physiology , Models, Biological , Transfection
12.
Proc Natl Acad Sci U S A ; 100(15): 8736-41, 2003 Jul 22.
Article in English | MEDLINE | ID: mdl-12853568

ABSTRACT

Measuring the biophysical properties of macromolecular complexes at work is a major challenge of modern biology. The protein complex composed of vesicle-associated membrane protein 2, synaptosomal-associated protein of 25 kDa, and syntaxin 1 [soluble N-ethyl-maleimide-sensitive factor attachment protein receptor (SNARE) complex] is essential for docking and fusion of neurotransmitter-filled synaptic vesicles with the presynaptic membrane. To better understand the fusion mechanisms, we reconstituted the synaptic SNARE complex in the imaging chamber of an atomic force microscope and measured the interaction forces between its components. Each protein was tested against the two others, taken either individually or as binary complexes. This approach allowed us to determine specific interaction forces and dissociation kinetics of the SNAREs and led us to propose a sequence of interactions. A theoretical model based on our measurements suggests that a minimum of four complexes is probably necessary for fusion to occur. We also showed that the regulatory protein neuronal Sec1 injected into the atomic force microscope chamber prevented the complex formation. Finally, we measured the effect of tetanus toxin protease on the SNARE complex and its activity by on-line registration during tetanus toxin injection. These experiments provide a basis for the functional study of protein microdomains and also suggest opportunities for sensitive screening of drugs that can modulate protein-protein interactions.


Subject(s)
Membrane Fusion/physiology , Membrane Proteins/physiology , Synaptic Vesicles/physiology , Antigens, Surface/chemistry , Antigens, Surface/physiology , Biophysical Phenomena , Biophysics , In Vitro Techniques , Kinetics , Macromolecular Substances , Membrane Proteins/chemistry , Microscopy, Atomic Force , Munc18 Proteins , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/pharmacology , Nerve Tissue Proteins/physiology , Protein Binding , R-SNARE Proteins , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SNARE Proteins , Synaptosomal-Associated Protein 25 , Syntaxin 1 , Tetanus Toxin/pharmacology , Vesicular Transport Proteins/pharmacology , Vesicular Transport Proteins/physiology
13.
FEBS Lett ; 525(1-3): 126-30, 2002 Aug 14.
Article in English | MEDLINE | ID: mdl-12163174

ABSTRACT

The interaction between Rab3A and calmodulin is necessary for the inhibitory effect of Rab3A in neuroendocrine cells. Contrastingly, Rab3A triggers the exocytosis known as acrosome reaction in permeabilized spermatozoa. Here we show that a Rab3A mutant that cannot bind calmodulin was fully capable of triggering acrosomal exocytosis. Additionally, calmodulin by itself abrogated the exocytosis triggered by Rab3A. The effect was observed with both the wild type protein and the calmodulin binding deficient mutant. Our results indicate that the inhibitory and stimulatory effects of Rab3A in different exocytic processes are mediated by different effectors.


Subject(s)
Acrosome Reaction/physiology , Calmodulin/metabolism , Exocytosis/physiology , rab3A GTP-Binding Protein/metabolism , Acrosome Reaction/drug effects , Calcimycin/pharmacology , Calmodulin/antagonists & inhibitors , Calmodulin/pharmacology , Cell Membrane Permeability , Chlorpromazine/pharmacology , Dopamine Antagonists/pharmacology , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Fluorescent Dyes , Humans , Ionophores/pharmacology , Male , Mutation , Progesterone/pharmacology , Protein Binding/physiology , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/pharmacology
14.
FEBS Lett ; 503(1): 19-24, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11513847

ABSTRACT

The expression of rab3A and rab3D isoforms in the enteroendocrine, cholecystokinin-secreting, cell lines STC-1 and GLUTag is here demonstrated. In contrast, rab3B is undetectable in these two cell lines, and rab3C is only slightly expressed in GLUTag cells. Using a transient co-transfection system with human growth hormone as reporter protein, we show that overexpression of the GTPase-deficient mutant rab3AQ81L, but not rab3DQ81L, significantly decreases human growth hormone secretory responses to various agonists in STC-1 cells. These results indicate that endocrine cell lines of intestinal origin express rab3A and rab3D proteins, but the GTP-bound form of rab3A only acts as a negative modulator in the control of cholecystokinin secretion from STC-1 cells.


Subject(s)
Cholecystokinin/metabolism , Exocytosis/physiology , rab3A GTP-Binding Protein/physiology , Animals , Cell Line , Genes, Reporter , Growth Hormone/genetics , Immunohistochemistry , Mice , Mutation , Rats , Transfection , rab3A GTP-Binding Protein/genetics
15.
J Biol Chem ; 276(35): 32756-62, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11438518

ABSTRACT

To define the role of the Rab3-interacting molecule RIM in exocytosis we searched for additional binding partners of the protein. We found that the two C(2) domains of RIM display properties analogous to those of the C(2)B domain of synaptotagmin-I. Thus, RIM-C(2)A and RIM-C(2)B bind in a Ca(2+)-independent manner to alpha1B, the pore-forming subunit of N-type Ca(2+) channels (EC(50) = approximately 20 nm). They also weakly interact with the alpha1C but not the alpha1D subunit of L-type Ca(2+) channels. In addition, the C(2) domains of RIM associate with SNAP-25 and synaptotagmin-I. The binding affinities for these two proteins are 203 and 24 nm, respectively, for RIM-C(2)A and 224 and 16 nm for RIM-C(2)B. The interactions of the C(2) domains of RIM with SNAP-25 and synaptotagmin-I are modulated by Ca(2+). Thus, in the presence of Ca(2+) (EC(50) = approximately 75 microm) the interaction with synaptotagmin-I is increased, whereas SNAP-25 binding is reduced. Synaptotagmin-I binding is abolished by mutations in two positively charged amino acids in the C(2) domains of RIM and by the addition of inositol polyphosphates. We propose that the Rab3 effector RIM is a scaffold protein that participates through its multiple binding partners in the docking and fusion of secretory vesicles at the release sites.


Subject(s)
Calcium Channels/metabolism , Calcium-Binding Proteins , GTP-Binding Proteins , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Surface/chemistry , Antigens, Surface/metabolism , Binding Sites , Brain/metabolism , Calcium/metabolism , Calcium/pharmacology , Calcium Channels/chemistry , Cloning, Molecular , Humans , Kinetics , Membrane Glycoproteins/chemistry , Membrane Proteins/chemistry , Molecular Sequence Data , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits , RNA, Messenger/genetics , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Synaptosomal-Associated Protein 25 , Synaptotagmin I , Synaptotagmins , Syntaxin 1 , Zinc Fingers , rab3 GTP-Binding Proteins/metabolism
16.
J Cell Sci ; 114(Pt 9): 1757-64, 2001 May.
Article in English | MEDLINE | ID: mdl-11309205

ABSTRACT

Rabphilin is a secretory vesicle protein that interacts with the GTP-bound form of the small GTPase Rab3. We investigated the involvement of Rabphilin in endocytosis using different point mutants of the protein. Overexpression of wild-type Rabphilin in the insulin-secreting cell line HIT-T15 did not affect receptor-mediated transferrin endocytosis. By contrast, Rabphilin V61A, a mutant that is unable to interact with Rab3, enhanced the rate of transferrin internalization. The effect of Rabphilin V61A was not mimicked by Rabphilin L83A, another mutant with impaired Rab3 binding. Careful analysis of the properties of the two mutants revealed that Rabphilin V61A and Rabphilin L83A are both targeted to secretory vesicles, have stimulatory activity on exocytosis, and bind equally well to alpha-actinin. However, Rabphilin L83A fails to interact with Rabaptin-5, an important component of the endocytotic machinery. These results indicate that Rabphilin promotes receptor-mediated endocytosis and that its action is negatively modulated by Rab3. We propose that the hydrolysis of GTP that is coupled to the exocytotic event disrupts the Rabphilin-Rab3 complex and permits the recruitment of Rabaptin-5 at the fusion site. Our data show that immediately after internalization the transferrin receptor and VAMP-2 colocalize on the same vesicular structures, suggesting that Rabphilin favors the rapid recycling of the components of the secretory vesicle.


Subject(s)
Endocytosis , GTP-Binding Proteins/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins , rab3 GTP-Binding Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Protein Binding
17.
Eur J Neurosci ; 12(6): 1913-23, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10886332

ABSTRACT

In addition to its role in exocytosis, SNAP-25 is essential for axonal outgrowth. In order to identify SNARE proteins involved in neurite growth we have used SNAP-25 antibodies to affinity-purify protein complexes enriched in developing rat brain membrane extracts. We have identified a complex between SNAP-25 and syntaxin 13 predominantly present in brain at embryonic or early postnatal stages. We show that syntaxin 13 is developmentally regulated with a decrease in adult brain. In differentiated neuroendocrine PC12 cells as well as primary cortical neurons the protein is localized to a punctated and tubular staining in the perinuclear region and along processes with high levels in the central region of growth cones. Carboxy-terminally tagged syntaxin 13 was also detected on the plasma membrane by in vivo surface-labelling where it colocalized with SNAP-25. Syntaxin 13 has recently been shown to be implicated in early endosomal trafficking. In our study, colocalization with internalized transferrin in the cell body and along neurites confirmed endosomal location in both compartments. Finally, overexpression of full-length syntaxin 13 enhanced neurite outgrowth in NGF-stimulated PC12 cells, whilst it had no effect on regulated secretion. The data suggest that a syntaxin 13-dependent endocytic trafficking step plays a limiting role in membrane expansion during neuronal development.


Subject(s)
Endosomes/metabolism , Membrane Proteins/metabolism , Neurites/metabolism , Vesicular Transport Proteins , Age Factors , Amino Acid Sequence , Animals , Biological Transport/physiology , Cell Membrane/chemistry , Cell Membrane/metabolism , Endosomes/chemistry , Exocytosis/physiology , Growth Cones/chemistry , Growth Cones/metabolism , Membrane Proteins/analysis , Membrane Proteins/genetics , Molecular Sequence Data , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurites/chemistry , PC12 Cells , Qa-SNARE Proteins , Rats , SNARE Proteins , Synaptosomal-Associated Protein 25 , Transfection
18.
Mol Biol Cell ; 11(7): 2403-17, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888677

ABSTRACT

Like neuronal synaptic vesicles, intracellular GLUT4-containing vesicles must dock and fuse with the plasma membrane, thereby facilitating insulin-regulated glucose uptake into muscle and fat cells. GLUT4 colocalizes in part with the vesicle SNAREs VAMP2 and VAMP3. In this study, we used a single-cell fluorescence-based assay to compare the functional involvement of VAMP2 and VAMP3 in GLUT4 translocation. Transient transfection of proteolytically active tetanus toxin light chain cleaved both VAMP2 and VAMP3 proteins in L6 myoblasts stably expressing exofacially myc-tagged GLUT4 protein and inhibited insulin-stimulated GLUT4 translocation. Tetanus toxin also caused accumulation of the remaining C-terminal VAMP2 and VAMP3 portions in Golgi elements. This behavior was exclusive to these proteins, because the localization of intracellular myc-tagged GLUT4 protein was not affected by the toxin. Upon cotransfection of tetanus toxin with individual vesicle SNARE constructs, only toxin-resistant VAMP2 rescued the inhibition of insulin-dependent GLUT4 translocation by tetanus toxin. Moreover, insulin caused a cortical actin filament reorganization in which GLUT4 and VAMP2, but not VAMP3, were clustered. We propose that VAMP2 is a resident protein of the insulin-sensitive GLUT4 compartment and that the integrity of this protein is required for GLUT4 vesicle incorporation into the cell surface in response to insulin.


Subject(s)
Insulin/metabolism , Membrane Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Actins/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane/metabolism , Glucose Transporter Type 4 , Insulin/pharmacology , Monosaccharide Transport Proteins/genetics , Muscle, Skeletal/cytology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , R-SNARE Proteins , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetanus Toxin/metabolism , Vesicle-Associated Membrane Protein 3
19.
FEBS Lett ; 474(1): 66-70, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10828453

ABSTRACT

The putative Rab3 effector RIM (Rab3-interacting molecule) was detected by Northern blotting, RT-PCR and Western blotting in native pancreatic beta-cells as well as in the derived cell lines INS-1E and HIT-T15. RIM was localized on the plasma membrane of INS-1E cells and beta-cells. An involvement of RIM in insulin exocytosis was indicated by transfection experiments of INS-1E cells with the Rab3 binding domain of RIM. This domain enhanced glucose-stimulated secretion in intact cells and Ca(2+)-stimulated exocytosis in permeabilized cells. Co-expression of Rab3A reversed the effect of RIM on exocytosis. These results suggest an implication of RIM in the control of insulin secretion.


Subject(s)
Exocytosis , GTP-Binding Proteins , Gene Expression , Insulin/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Animals , Cell Line , Cell Membrane/chemistry , Cricetinae , Humans , Insulin Secretion , Insulinoma , Islets of Langerhans/ultrastructure , Mice , Nerve Tissue Proteins/analysis , Pancreatic Neoplasms , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , rab3 GTP-Binding Proteins/metabolism
20.
EMBO J ; 18(21): 5885-91, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10545100

ABSTRACT

Rab GTPases regulate membrane traffic between the cellular compartments of eukaryotic cells. Rab3 is associated with secretory vesicles of neuronal and endocrine cells and controls the Ca(2+)-triggered release of neurotransmitters and hormones. To clarify the mode of action of Rab3 we generated mutants of the GTPase that do not interact efficiently with its putative effectors Rabphilin and RIM. Surprisingly, these mutants transfected in PC12 cells were still capable of inhibiting Ca(2+)-evoked secretion. Rab3 was shown previously to bind to calmodulin in a Ca(2+)-dependent manner. By replacing two arginines conserved between Rab3 isoforms, we generated a mutant with a reduced affinity for calmodulin. This mutant retained the capacity to interact with the Rab3 regulatory proteins, Rabphilin, RIM, Mss4 and RabGDI, and was correctly targeted to dense-core secretory granules. However, replacement of the two arginines abolished the ability of the GTP-bound form of Rab3 to inhibit exocytosis of catecholamine- and insulin-secreting cells. We propose that a Rab3-calmodulin complex generated by elevated Ca(2+) concentrations mediated at least some of the effects of the GTPase and limited the number of exocytotic events that occurred in response to secretory stimuli.


Subject(s)
Calmodulin/metabolism , Exocytosis/genetics , Guanine Nucleotide Exchange Factors , rab3 GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium/pharmacology , Cytoplasmic Granules/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Microscopy, Fluorescence , Mutation , Nerve Tissue Proteins/metabolism , PC12 Cells , Protein Binding/genetics , Proteins/metabolism , Rats , Transfection , Vesicular Transport Proteins , rab GTP-Binding Proteins/metabolism , rab3 GTP-Binding Proteins/genetics , Rabphilin-3A
SELECTION OF CITATIONS
SEARCH DETAIL
...