Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mol Metab ; 84: 101955, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704026

ABSTRACT

OBJECTIVE: The contribution of the mitochondrial electron transfer system to insulin secretion involves more than just energy provision. We identified a small RNA fragment (mt-tRF-LeuTAA) derived from the cleavage of a mitochondrially-encoded tRNA that is conserved between mice and humans. The role of mitochondrially-encoded tRNA-derived fragments remains unknown. This study aimed to characterize the impact of mt-tRF-LeuTAA, on mitochondrial metabolism and pancreatic islet functions. METHODS: We used antisense oligonucleotides to reduce mt-tRF-LeuTAA levels in primary rat and human islet cells, as well as in insulin-secreting cell lines. We performed a joint transcriptome and proteome analysis upon mt-tRF-LeuTAA inhibition. Additionally, we employed pull-down assays followed by mass spectrometry to identify direct interactors of the fragment. Finally, we characterized the impact of mt-tRF-LeuTAA silencing on the coupling between mitochondrial metabolism and insulin secretion using high-resolution respirometry and insulin secretion assays. RESULTS: Our study unveils a modulation of mt-tRF-LeuTAA levels in pancreatic islets in different Type 2 diabetes models and in response to changes in nutritional status. The level of the fragment is finely tuned by the mechanistic target of rapamycin complex 1. Located within mitochondria, mt-tRF-LeuTAA interacts with core subunits and assembly factors of respiratory complexes of the electron transfer system. Silencing of mt-tRF-LeuTAA in islet cells limits the inner mitochondrial membrane potential and impairs mitochondrial oxidative phosphorylation, predominantly by affecting the Succinate (via Complex II)-linked electron transfer pathway. Lowering mt-tRF-LeuTAA impairs insulin secretion of rat and human pancreatic ß-cells. CONCLUSIONS: Our findings indicate that mt-tRF-LeuTAA interacts with electron transfer system complexes and is a pivotal regulator of mitochondrial oxidative phosphorylation and its coupling to insulin secretion.


Subject(s)
Insulin Secretion , Insulin-Secreting Cells , Mitochondria , Animals , Rats , Humans , Mitochondria/metabolism , Insulin-Secreting Cells/metabolism , RNA, Transfer/metabolism , RNA, Transfer/genetics , Male , Insulin/metabolism , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 2/metabolism , RNA, Mitochondrial/metabolism , RNA, Mitochondrial/genetics , Mice , Rats, Wistar , Electron Transport
2.
Cell Rep ; 40(2): 111069, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830789

ABSTRACT

tRNA-derived fragments (tRFs) are an emerging class of small non-coding RNAs with distinct cellular functions. Here, we studied the contribution of tRFs to the regulation of postnatal ß cell maturation, a critical process that may lead to diabetes susceptibility in adulthood. We identified three tRFs abundant in neonatal rat islets originating from 5' halves (tiRNA-5s) of histidine and glutamate tRNAs. Their inhibition in these islets reduced ß cell proliferation and insulin secretion. Mitochondrial respiration was also perturbed, fitting with the mitochondrial enrichment of nuclear-encoded tiRNA-5HisGTG and tiRNA-5GluCTC. Notably, tiRNA-5 inhibition reduced Mpc1, a mitochondrial pyruvate carrier whose knock down largely phenocopied tiRNA-5 inhibition. tiRNA-5HisGTG interactome revealed binding to Musashi-1, which was essential for the mitochondrial enrichment of tiRNA-5HisGTG. Finally, tiRNA-5s were dysregulated in the islets of diabetic and diabetes-prone animals. Altogether, tiRNA-5s represent a class of regulators of ß cell maturation, and their deregulation in neonatal islets may lead to diabetes susceptibility in adulthood.


Subject(s)
Insulin-Secreting Cells , RNA, Transfer , Animals , Cell Proliferation , Insulin Secretion , Insulin-Secreting Cells/metabolism , RNA/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Rats
3.
Sci Rep ; 11(1): 8800, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888791

ABSTRACT

Glucose-induced insulin secretion, a hallmark of mature ß-cells, is achieved after birth and is preceded by a phase of intense proliferation. These events occurring in the neonatal period are decisive for establishing an appropriate functional ß-cell mass that provides the required insulin throughout life. However, key regulators of gene expression involved in functional maturation of ß-cells remain to be elucidated. Here, we addressed this issue by mapping open chromatin regions in newborn versus adult rat islets using the ATAC-seq assay. We obtained a genome-wide picture of chromatin accessible sites (~ 100,000) among which 20% were differentially accessible during maturation. An enrichment analysis of transcription factor binding sites identified a group of transcription factors that could explain these changes. Among them, Scrt1 was found to act as a transcriptional repressor and to control ß-cell proliferation. Interestingly, Scrt1 expression was controlled by the transcriptional repressor RE-1 silencing transcription factor (REST) and was increased in an in vitro reprogramming system of pancreatic exocrine cells to ß-like cells. Overall, this study led to the identification of several known and unforeseen key transcriptional events occurring during ß-cell maturation. These findings will help defining new strategies to induce the functional maturation of surrogate insulin-producing cells.


Subject(s)
Cell Proliferation/physiology , Chromatin/metabolism , Gene Expression Regulation/physiology , Insulin-Secreting Cells/cytology , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , Humans , Rats
4.
Nat Commun ; 11(1): 5611, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154349

ABSTRACT

Fine-tuning of insulin release from pancreatic ß-cells is essential to maintain blood glucose homeostasis. Here, we report that insulin secretion is regulated by a circular RNA containing the lariat sequence of the second intron of the insulin gene. Silencing of this intronic circular RNA in pancreatic islets leads to a decrease in the expression of key components of the secretory machinery of ß-cells, resulting in impaired glucose- or KCl-induced insulin release and calcium signaling. The effect of the circular RNA is exerted at the transcriptional level and involves an interaction with the RNA-binding protein TAR DNA-binding protein 43 kDa (TDP-43). The level of this circularized intron is reduced in the islets of rodent diabetes models and of type 2 diabetic patients, possibly explaining their impaired secretory capacity. The study of this and other circular RNAs helps understanding ß-cell dysfunction under diabetes conditions, and the etiology of this common metabolic disorder.


Subject(s)
Insulin Secretion/genetics , Insulin/genetics , Introns , RNA, Circular/metabolism , Animals , Calcium Signaling , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , RNA, Circular/genetics , Rats
5.
Compr Physiol ; 10(3): 893-932, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32941685

ABSTRACT

The discovery that most mammalian genome sequences are transcribed to ribonucleic acids (RNA) has revolutionized our understanding of the mechanisms governing key cellular processes and of the causes of human diseases, including diabetes mellitus. Pancreatic islet cells were found to contain thousands of noncoding RNAs (ncRNAs), including micro-RNAs (miRNAs), PIWI-associated RNAs, small nucleolar RNAs, tRNA-derived fragments, long non-coding RNAs, and circular RNAs. While the involvement of miRNAs in islet function and in the etiology of diabetes is now well documented, there is emerging evidence indicating that other classes of ncRNAs are also participating in different aspects of islet physiology. The aim of this article will be to provide a comprehensive and updated view of the studies carried out in human samples and rodent models over the past 15 years on the role of ncRNAs in the control of α- and ß-cell development and function and to highlight the recent discoveries in the field. We not only describe the role of ncRNAs in the control of insulin and glucagon secretion but also address the contribution of these regulatory molecules in the proliferation and survival of islet cells under physiological and pathological conditions. It is now well established that most cells release part of their ncRNAs inside small extracellular vesicles, allowing the delivery of genetic material to neighboring or distantly located target cells. The role of these secreted RNAs in cell-to-cell communication between ß-cells and other metabolic tissues as well as their potential use as diabetes biomarkers will be discussed. © 2020 American Physiological Society. Compr Physiol 10:893-932, 2020.


Subject(s)
Diabetes Mellitus/genetics , Insulin-Secreting Cells/physiology , RNA, Untranslated/genetics , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Gene Expression Regulation , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology
6.
EBioMedicine ; 58: 102895, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32739864

ABSTRACT

BACKGROUND: Bariatric surgery is an effective treatment for type 2 diabetes. Early post-surgical enhancement of insulin secretion is key for diabetes remission. The full complement of mechanisms responsible for improved pancreatic beta cell functionality after bariatric surgery is still unclear. Our aim was to identify pathways, evident in the islet transcriptome, that characterize the adaptive response to bariatric surgery independently of body weight changes. METHODS: We performed entero-gastro-anastomosis (EGA) with pyloric ligature in leptin-deficient ob/ob mice as a surrogate of Roux-en-Y gastric bypass (RYGB) in humans. Multiple approaches such as determination of glucose tolerance, GLP-1 and insulin secretion, whole body insulin sensitivity, ex vivo glucose-stimulated insulin secretion (GSIS) and functional multicellular Ca2+-imaging, profiling of mRNA and of miRNA expression were utilized to identify significant biological processes involved in pancreatic islet recovery. FINDINGS: EGA resolved diabetes, increased pancreatic insulin content and GSIS despite a persistent increase in fat mass, systemic and intra-islet inflammation, and lipotoxicity. Surgery differentially regulated 193 genes in the islet, most of which were involved in the regulation of glucose metabolism, insulin secretion, calcium signaling or beta cell viability, and these were normalized alongside changes in glucose metabolism, intracellular Ca2+ dynamics and the threshold for GSIS. Furthermore, 27 islet miRNAs were differentially regulated, four of them hubs in a miRNA-gene interaction network and four others part of a blood signature of diabetes resolution in ob/ob mice and in humans. INTERPRETATION: Taken together, our data highlight novel miRNA-gene interactions in the pancreatic islet during the resolution of diabetes after bariatric surgery that form part of a blood signature of diabetes reversal. FUNDING: European Union's Horizon 2020 research and innovation programme via the Innovative Medicines Initiative 2 Joint Undertaking (RHAPSODY), INSERM, Société Francophone du Diabète, Institut Benjamin Delessert, Wellcome Trust Investigator Award (212625/Z/18/Z), MRC Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1), Diabetes UK (BDA/11/0004210, BDA/15/0005275, BDA 16/0005485) project grants, National Science Foundation (310030-188447), Fondation de l'Avenir.


Subject(s)
Diabetes Mellitus, Type 2/surgery , Gene Regulatory Networks , Insulin-Secreting Cells/chemistry , MicroRNAs/genetics , Obesity/surgery , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Gastric Bypass , Gene Expression Profiling , Gene Expression Regulation , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Male , Mice , Mice, Obese , Obesity/genetics , Obesity/metabolism
7.
Sci Rep ; 10(1): 8354, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32415214

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Sci Rep ; 10(1): 6413, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286361

ABSTRACT

Long non-coding RNAs (lncRNAs) contribute to diverse cellular functions and the dysregulation of their expression or function can contribute to diseases, including diabetes. The contributions of lncRNAs to ß-cell development, function and survival has been extensively studied in vitro. However, very little is currently known on the in vivo roles of lncRNAs in the regulation of glucose and insulin homeostasis. Here we investigated the impact of loss-of-function in mice of the lncRNA A830019P07Rik, hereafter P07Rik, which was previously reported to be associated with reduced plasma insulin levels. Compared with wild-type littermates, male and female P07Rik mutant mice did not show any defect in glycaemia and plasma insulin levels in both fed and fasted state. Furthermore, P07Rik mutant mice displayed similar glucose and insulin levels in response to an intra-peritoneal glucose tolerance test. Ex vivo, islets from mutant P07Rik released similar amount of insulin in response to increased glucose concentration as wildtype littermates. In contrast with previous reports, our characterization of P07Rik mouse mutants revealed that loss of function of this lncRNA does not affect glucose and insulin homeostasis in mice.


Subject(s)
Insulin Secretion/genetics , Insulin/metabolism , RNA, Long Noncoding/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Conserved Sequence/genetics , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Down-Regulation/genetics , Fasting/blood , Feeding Behavior , Female , Homeostasis , Insulin/blood , Islets of Langerhans/metabolism , Male , Mice, Obese , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Cell Metab ; 29(2): 348-361.e6, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30318337

ABSTRACT

Type 1 diabetes is an autoimmune disease initiated by the invasion of pancreatic islets by immune cells that selectively kill the ß cells. We found that rodent and human T lymphocytes release exosomes containing the microRNAs (miRNAs) miR-142-3p, miR-142-5p, and miR-155, which can be transferred in active form to ß cells favoring apoptosis. Inactivation of these miRNAs in recipient ß cells prevents exosome-mediated apoptosis and protects non-obese diabetic (NOD) mice from diabetes development. Islets from protected NOD mice display higher insulin levels, lower insulitis scores, and reduced inflammation. Looking at the mechanisms underlying exosome action, we found that T lymphocyte exosomes trigger apoptosis and the expression of genes involved in chemokine signaling, including Ccl2, Ccl7, and Cxcl10, exclusively in ß cells. The induction of these genes may promote the recruitment of immune cells and exacerbate ß cell death during the autoimmune attack. Our data point to exosomal-miRNA transfer as a communication mode between immune and insulin-secreting cells.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Exosomes/metabolism , Insulin-Secreting Cells/immunology , MicroRNAs/physiology , T-Lymphocytes/immunology , Adult , Animals , Female , Humans , Insulin-Secreting Cells/cytology , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Rats , Rats, Wistar , T-Lymphocytes/cytology
10.
Xenotransplantation ; 26(2): e12474, 2019 03.
Article in English | MEDLINE | ID: mdl-30461074

ABSTRACT

BACKGROUND: Aberrant microRNA (miRNA) expression levels are associated with various graft rejections. We used our humanized mouse model with transplanted human islets to identify miRNAs in islet grafts related to xenograft rejection and circulating miRNAs associated with xenograft rejection-mediated ß-cell loss. METHODS: Diabetic immunodeficient NOD.scid mice were transplanted with human islets and subsequently achieved stable normoglycemia. Lymphocytes from NOD mice were then adoptively transferred to the humanized mice to induce human ß-cell destruction. Islet graft and plasma were collected immediately once blood glucose reached >200 mg/dL. miRNAs in the islet grafts and in the plasma with or without adoptive lymphocyte transfer (ALT) were measured using NanoString nCounter® miRNA Expression Assay and qPCR. RESULTS: A set of immune-related miRNAs was significantly increased in human islet grafts of ALT-treated mice compared to control mice. Of these miRNAs, miR-150-5p was significantly increased in the circulation of ALT-treated mice at tissue collection and the increase was a result of immune activation rather than simply the presence of lymphocytes in circulation. Furthermore, miR-150-5p was significantly increased in human islet graft and circulation prior to the development of hyperglycemia in the ALT-treated mice. CONCLUSIONS: Our data demonstrated that immune-related miRNAs are associated with human islet xenograft rejection in mice. miR-150-5p is increased in human islet graft and in the circulation during islet xenograft rejection and ß-cell destruction prior to hyperglycemia and may be an early biomarker for islet xenograft rejection.


Subject(s)
Islets of Langerhans Transplantation/immunology , Lymphocytes/immunology , MicroRNAs/genetics , Transplantation, Heterologous , Animals , Disease Models, Animal , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , Heterografts/immunology , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD , MicroRNAs/immunology , Transplantation, Heterologous/methods
12.
Diabetes ; 67(11): 2254-2267, 2018 11.
Article in English | MEDLINE | ID: mdl-30115652

ABSTRACT

Pancreatic ß-cell expansion throughout the neonatal period is essential to generate the appropriate mass of insulin-secreting cells required to maintain blood glucose homeostasis later in life. Hence, defects in this process can predispose to diabetes development during adulthood. Global profiling of transcripts in pancreatic islets of newborn and adult rats revealed that the transcription factor E2F1 controls expression of the long noncoding RNA H19, which is profoundly downregulated during the postnatal period. H19 silencing decreased ß-cell expansion in newborns, whereas its re-expression promoted proliferation of ß-cells in adults via a mechanism involving the microRNA let-7 and the activation of Akt. The offspring of rats fed a low-protein diet during gestation and lactation display a small ß-cell mass and an increased risk of developing diabetes during adulthood. We found that the islets of newborn rats born to dams fed a low-protein diet express lower levels of H19 than those born to dams that did not eat a low-protein diet. Moreover, we observed that H19 expression increases in islets of obese mice under conditions of increased insulin demand. Our data suggest that the long noncoding RNA H19 plays an important role in postnatal ß-cell mass expansion in rats and contributes to the mechanisms compensating for insulin resistance in obesity.


Subject(s)
Cell Proliferation/physiology , Insulin-Secreting Cells/metabolism , RNA, Long Noncoding/metabolism , Animals , Cell Death/physiology , Cell Line , Gene Expression Profiling , Male , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics , Rats , Rats, Sprague-Dawley
13.
J Endocr Soc ; 2(7): 631-645, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29942927

ABSTRACT

An important feature of type 2 diabetes is a decrease in ß-cell mass. Therefore, it is essential to find new approaches to stimulate ß-cell proliferation. We have previously shown that heterozygous inactivation of the Na+/Ca2+ exchanger (isoform 1; NCX1), a protein responsible for Ca2+ extrusion from cells, increases ß-cell proliferation, mass, and function in mice. Here, we show that Ncx1 inactivation also increases ß-cell proliferation in 2-year-old mice and that NCX1 inhibition in adult mice by four small molecules of the benzoxyphenyl family stimulates ß-cell proliferation both in vitro and in vivo. NCX1 inhibition by small interfering RNA or small molecules activates the calcineurin/nuclear factor of activated T cells (NFAT) pathway and inhibits apoptosis induced by the immunosuppressors cyclosporine A (CsA) and tacrolimus in insulin-producing cell. Moreover, NCX1 inhibition increases the expression of ß-cell-specific genes, such as Ins1, Ins2, and Pdx1, and inactivates/downregulates the tumor suppressors retinoblastoma protein (pRb) and miR-193a and the cell cycle inhibitor p53. Our data show that Na+/Ca2+ exchange is a druggable target to stimulate ß-cell function and proliferation. Specific ß-cell inhibition of Na+/Ca2+ exchange by phenoxybenzamyl derivatives may represent an innovative approach to promote ß-cell regeneration in diabetes and improve the efficiency of pancreatic islet transplantation for the treatment of the disease.

14.
Mol Metab ; 9: 69-83, 2018 03.
Article in English | MEDLINE | ID: mdl-29396373

ABSTRACT

OBJECTIVE: There is strong evidence for an involvement of different classes of non-coding RNAs, including microRNAs and long non-coding RNAs, in the regulation of ß-cell activities and in diabetes development. Circular RNAs were recently discovered to constitute a substantial fraction of the mammalian transcriptome but the contribution of these non-coding RNAs in physiological and disease processes remains largely unknown. The goal of this study was to identify the circular RNAs expressed in pancreatic islets and to elucidate their possible role in the control of ß-cells functions. METHODS: We used a microarray approach to identify circular RNAs expressed in human islets and searched their orthologues in RNA sequencing data from mouse islets. We then measured the level of four selected circular RNAs in the islets of different Type 1 and Type 2 diabetes models and analyzed the role of these circular transcripts in the regulation of insulin secretion, ß-cell proliferation, and apoptosis. RESULTS: We identified thousands of circular RNAs expressed in human pancreatic islets, 497 of which were conserved in mouse islets. The level of two of these circular transcripts, circHIPK3 and ciRS-7/CDR1as, was found to be reduced in the islets of diabetic db/db mice. Mimicking this decrease in the islets of wild type animals resulted in impaired insulin secretion, reduced ß-cell proliferation, and survival. ciRS-7/CDR1as has been previously proposed to function by blocking miR-7. Transcriptomic analysis revealed that circHIPK3 acts by sequestering a group of microRNAs, including miR-124-3p and miR-338-3p, and by regulating the expression of key ß-cell genes, such as Slc2a2, Akt1, and Mtpn. CONCLUSIONS: Our findings point to circular RNAs as novel regulators of ß-cell activities and suggest an involvement of this novel class of non-coding RNAs in ß-cell dysfunction under diabetic conditions.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , RNA/genetics , Animals , Apoptosis , Cell Line , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Humans , Insulin Secretion , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , RNA/metabolism , RNA, Circular , Rats , Rats, Wistar
15.
Diabetes Obes Metab ; 19 Suppl 1: 137-146, 2017 09.
Article in English | MEDLINE | ID: mdl-28880477

ABSTRACT

Blood glucose homeostasis requires a constant communication between insulin-secreting and insulin-sensitive cells. A wide variety of circulating factors, including hormones, cytokines and chemokines work together to orchestrate the systemic response of metabolic organs to changes in the nutritional state. Failure in the coordination between these organs can lead to a rise in blood glucose levels and to the appearance of metabolic disorders such as diabetes mellitus. Exosomes are small extracellular vesicles (EVs) that are produced via the endosomal pathway and are released from the cells upon fusion of multivesicular bodies with the plasma membrane. There is emerging evidence indicating that these EVs play a central role in cell-to-cell communication. The interest in exosomes exploded when they were found to transport bioactive proteins, messenger RNA (mRNAs) and microRNA (miRNAs) that can be transferred in active form to adjacent cells or to distant organs. In this review, we will first outline the mechanisms governing the biogenesis, the cargo upload and the release of exosomes by donor cells as well as the uptake by recipient cells. We will then summarize the studies that support the novel concept that miRNAs and other exosomal cargo components are new important vehicles for metabolic organ cross-talk.


Subject(s)
Cell Communication , Exosomes/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Models, Biological , RNA, Messenger/metabolism , Animals , Autocrine Communication , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Endocytosis , Endosomes/metabolism , Endosomes/pathology , Endosomes/physiology , Exocytosis , Exosomes/pathology , Exosomes/physiology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , MicroRNAs/physiology , Organelle Biogenesis , Paracrine Communication , RNA, Messenger/physiology
16.
Diabetologia ; 60(10): 1977-1986, 2017 10.
Article in English | MEDLINE | ID: mdl-28711973

ABSTRACT

AIMS/HYPOTHESIS: P-element induced Wimpy testis (PIWI)-interacting RNAs (piRNAs) are small non-coding RNAs that interact with PIWI proteins and guide them to silence transposable elements. They are abundantly expressed in germline cells and play key roles in spermatogenesis. There is mounting evidence that piRNAs are also present in somatic cells, where they may accomplish additional regulatory tasks. The aim of this study was to identify the piRNAs expressed in pancreatic islets and to determine whether they are involved in the control of beta cell activities. METHODS: piRNA profiling of rat pancreatic islets was performed by microarray analysis. The functions of piRNAs were investigated by silencing the two main Piwi genes or by modulating the level of selected piRNAs in islet cells. RESULTS: We detected about 18,000 piRNAs in rat pancreatic islets, many of which were differentially expressed throughout islet postnatal development. Moreover, we identified changes in the level of several piRNAs in the islets of Goto-Kakizaki rats, a well-established animal model of type 2 diabetes. Silencing of Piwil2 or Piwil4 genes in adult rat islets caused a reduction in the level of several piRNAs and resulted in defective insulin secretion and increased resistance of the cells to cytokine-induced cell death. Furthermore, overexpression in the islets of control animals of two piRNAs that are upregulated in diabetic rats led to a selective defect in glucose-induced insulin release. CONCLUSIONS/INTERPRETATION: Our results provide evidence for a role of PIWI proteins and their associated piRNAs in the control of beta cell functions, and suggest a possible involvement in the development of type 2 diabetes. DATA AVAILABILITY: Data have been deposited in Gene Expression Omnibus repository under the accession number GSE93792. Data can be accessed via the following link: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=ojklueugdzehpkv&acc=GSE93792.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , RNA, Small Interfering/metabolism , Animals , Cell Proliferation/physiology , Diabetes Mellitus, Type 2/metabolism , Female , Gene Expression Profiling , Insulin Secretion , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar
17.
Diabetologia ; 60(10): 2011-2020, 2017 10.
Article in English | MEDLINE | ID: mdl-28674733

ABSTRACT

AIMS/HYPOTHESIS: Evidence continues to emerge detailing a fine-tuning of the regulation of metabolic processes and energy homeostasis by cell-autonomous circadian clocks. Pancreatic beta cell functional maturation occurs after birth and implies transcriptional changes triggered by a shift in the nutritional supply that occurs at weaning, enabling the adaptation of insulin secretion. So far, the developmental timing and exact mechanisms involved in the initiation of the circadian clock in the growing pancreatic islets have never been addressed. METHODS: Circadian gene expression was measured by quantitative RT-PCR in islets of rats at different postnatal ages up to 3 months, and by in vitro bioluminescence recording in newborn (10-day-old) and adult (3-month-old) islets. The effect of the microRNAs miR-17-5p and miR-29b-3p on the expression of target circadian genes was assessed in newborn rat islets transfected with microRNA antisense or mimic oligonucleotides, and luciferase reporter assays were performed on the rat insulin-secreting cell line INS832/13 to determine a direct effect. The global regulatory network between microRNAs and circadian genes was computationally predicted. RESULTS: We found up to a sixfold-change in the 24 h transcriptional oscillations and overall expression of Clock, Npas2, Bmal1, Bmal2, Rev-erbα, Per1, Per2, Per3 and Cry2 between newborn and adult rat islets. Synchronisation of the clock machinery in cultured islet cells revealed a delayed cell-autonomous rhythmicity of about 1.5 h in newborn compared with adult rats. Computational predictions unveiled the existence of a complex regulatory network linking over 40 microRNAs displaying modifications in their expression profiles during postnatal beta cell maturation and key core-clock genes. In agreement with these computational predictions, we demonstrated that miR-17-5p and miR-29b-3p directly regulated circadian gene expression in the maturing islet cells of 10-day-old rats. CONCLUSIONS/INTERPRETATION: These data show that the circadian clock is not fully operational in newborn islets and that microRNAs potently contribute to its regulation during postnatal beta cell maturation. Defects in this process may have long-term consequences on circadian physiology and pancreatic islet function, favouring the manifestation of metabolic diseases such as diabetes.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Gene Expression Regulation/physiology , Islets of Langerhans/metabolism , MicroRNAs/metabolism , Animals , Animals, Newborn , Circadian Rhythm Signaling Peptides and Proteins/genetics , Female , Male , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley
18.
Biochim Biophys Acta ; 1861(12 Pt B): 2121-2129, 2016 12.
Article in English | MEDLINE | ID: mdl-27178175

ABSTRACT

MicroRNAs are key regulators of ß-cell physiology. They participate to the differentiation of insulin-producing cells and are instrumental for the acquisition of their unique secretory properties. Moreover, they contribute to the adaptation of ß-cells to conditions of increased insulin demand and, if expressed at inappropriate levels, certain microRNAs cause ß-cell dysfunction and promote the development of different forms of diabetes mellitus. While these functions are increasingly better understood, additional tasks for these small non-coding RNAs have been recently unveiled. Thus, microRNAs are emerging as signaling molecules of a novel exosome-mediated cell-to-cell communication mode permitting a coordinated response of the ß-cells to inflammatory conditions and to modifications in the insulin demand. These discoveries raise a number of important issues that once addressed promise to shed new light on the molecular mechanism governing the functions of the ß-cells under normal and disease states. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.


Subject(s)
Cell Communication/physiology , Cell Differentiation/physiology , Inflammation/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Animals , Exosomes/metabolism , Exosomes/physiology , Humans , Inflammation/pathology , Insulin-Secreting Cells/physiology
19.
Diabetologia ; 59(5): 1049-58, 2016 May.
Article in English | MEDLINE | ID: mdl-26852333

ABSTRACT

AIMS/HYPOTHESIS: The crosstalk between skeletal muscle (SkM) and beta cells plays a role in diabetes aetiology. In this study, we have investigated whether SkM-released exosome-like vesicles (ELVs) can be taken up by pancreatic beta cells and can deliver functional cargoes. METHODS: Mice were fed for 16 weeks with standard chow diet (SCD) or with standard diet enriched with 20% palmitate (HPD) and ELVs were purified from quadriceps muscle. Fluorescent ELVs from HPD or SCD quadriceps were injected i.v. or intramuscularly (i.m.) into mice to determine their biodistributions. Micro (mi)RNA quantification in ELVs was determined using quantitative real-time RT-PCR (qRT-PCR)-based TaqMan low-density arrays. Microarray analyses were performed to determine whether standard diet ELVs (SD-ELVs) and high palmitate diet ELVs (HPD-ELVs) induced specific transcriptional signatures in MIN6B1 cells. RESULTS: In vivo, muscle ELVs were taken up by pancreas, 24 h post-injection. In vitro, both SD-ELVs and HPD-ELVs transferred proteins and miRNAs to MIN6B1 cells and modulated gene expressions whereas only HPD-ELVs induced proliferation of MIN6B1 cells and isolated islets. Bioinformatic analyses suggested that transferred HPD-ELV miRNAs may participate in these effects. To validate this, we demonstrated that miR-16, which is overexpressed in HPD-ELVs, was transferred to MIN6B1 cells and regulated Ptch1, involved in pancreas development. In vivo, islets from HPD mice showed increased size and altered expression of genes involved in development, including Ptch1, suggesting that the effect of palm oil on islet size in vivo was reproduced in vitro by treating beta cells with HPD-ELVs. CONCLUSIONS/INTERPRETATION: Our data suggest that muscle ELVs might have an endocrine effect and could participate in adaptations in beta cell mass during insulin resistance.


Subject(s)
Exosomes/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Muscle, Skeletal/metabolism , Animals , Cell Line , Male , Mice , MicroRNAs/metabolism , Muscle Fibers, Skeletal/metabolism
20.
Diabetologia ; 59(1): 161-169, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26474776

ABSTRACT

AIMS/HYPOTHESIS: Ageing can lead to reduced insulin sensitivity and loss of pancreatic beta cell function, predisposing individuals to the development of diabetes. The aim of this study was to assess the contribution of microRNAs (miRNAs) to age-associated beta cell dysfunction. METHODS: The global mRNA and miRNA profiles of 3- and 12-month-old rat islets were collected by microarray. The functional impact of age-associated differences in miRNA expression was investigated by mimicking the observed changes in primary beta cells from young animals. RESULTS: Beta cells from 12-month-old rats retained normal insulin content and secretion, but failed to proliferate in response to mitotic stimuli. The islets of these animals displayed modifications at the level of several miRNAs, including upregulation of miR-34a, miR-124a and miR-383, and downregulation of miR-130b and miR-181a. Computational analysis of the transcriptomic modifications observed in the islets of 12-month-old rats revealed that the differentially expressed genes were enriched for miR-34a and miR-181a targets. Indeed, the induction of miR-34a and reduction of miR-181a in the islets of young animals mimicked the impaired beta cell proliferation observed in old animals. mRNA coding for alpha-type platelet-derived growth factor receptor, which is critical for compensatory beta cell mass expansion, is directly inhibited by miR34a and is likely to be at least partly responsible for the effects of this miRNA. CONCLUSIONS/INTERPRETATION: Changes in the level of specific miRNAs that occur during ageing affect the proliferative capacity of beta cells. This might reduce their ability to expand under conditions of increased insulin demand, favouring the development of type 2 diabetes.


Subject(s)
Aging , Gene Expression Regulation , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , MicroRNAs/metabolism , Animals , Apoptosis , Cell Proliferation , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Humans , Insulin/metabolism , Insulin Resistance , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transcriptome , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...