Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Diabetologia ; 66(7): 1306-1321, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36995380

RESUMEN

AIMS/HYPOTHESIS: Wolfram syndrome is a rare autosomal recessive disorder caused by pathogenic variants in the WFS1 gene. It is characterised by insulin-dependent diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing loss and neurodegeneration. Considering the unmet treatment need for this orphan disease, this study aimed to evaluate the therapeutic potential of glucagon-like peptide 1 receptor (GLP-1R) agonists under wolframin (WFS1) deficiency with a particular focus on human beta cells and neurons. METHODS: The effect of the GLP-1R agonists dulaglutide and exenatide was examined in Wfs1 knockout mice and in an array of human preclinical models of Wolfram syndrome, including WFS1-deficient human beta cells, human induced pluripotent stem cell (iPSC)-derived beta-like cells and neurons from control individuals and individuals affected by Wolfram syndrome, and humanised mice. RESULTS: Our study shows that the long-lasting GLP-1R agonist dulaglutide reverses impaired glucose tolerance in WFS1-deficient mice, and that exenatide and dulaglutide improve beta cell function and prevent apoptosis in different human WFS1-deficient models including iPSC-derived beta cells from people with Wolfram syndrome. Exenatide improved mitochondrial function, reduced oxidative stress and prevented apoptosis in Wolfram syndrome iPSC-derived neural precursors and cerebellar neurons. CONCLUSIONS/INTERPRETATION: Our study provides novel evidence for the beneficial effect of GLP-1R agonists on WFS1-deficient human pancreatic beta cells and neurons, suggesting that these drugs may be considered as a treatment for individuals with Wolfram syndrome.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Secretoras de Insulina , Atrofia Óptica , Síndrome de Wolfram , Humanos , Animales , Ratones , Síndrome de Wolfram/tratamiento farmacológico , Síndrome de Wolfram/genética , Exenatida/uso terapéutico , Atrofia Óptica/patología , Células Secretoras de Insulina/patología , Ratones Noqueados
2.
Int J Mol Sci ; 22(2)2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33419045

RESUMEN

The global rise in type 2 diabetes results from a combination of genetic predisposition with environmental assaults that negatively affect insulin action in peripheral tissues and impair pancreatic ß-cell function and survival. Nongenetic heritability of metabolic traits may be an important contributor to the diabetes epidemic. Transfer RNAs (tRNAs) are noncoding RNA molecules that play a crucial role in protein synthesis. tRNAs also have noncanonical functions through which they control a variety of biological processes. Genetic and environmental effects on tRNAs have emerged as novel contributors to the pathogenesis of diabetes. Indeed, altered tRNA aminoacylation, modification, and fragmentation are associated with ß-cell failure, obesity, and insulin resistance. Moreover, diet-induced tRNA fragments have been linked with intergenerational inheritance of metabolic traits. Here, we provide a comprehensive review of how perturbations in tRNA biology play a role in the pathogenesis of monogenic and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad/genética , Biosíntesis de Proteínas/genética , ARN de Transferencia/genética , Aminoacilación de ARN de Transferencia/genética , Animales , Diabetes Mellitus Tipo 2/metabolismo , Interacción Gen-Ambiente , Humanos , Células Secretoras de Insulina/metabolismo , Procesamiento Postranscripcional del ARN/genética , ARN de Transferencia/metabolismo
3.
BMC Genomics ; 21(1): 590, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32847508

RESUMEN

BACKGROUND: Prolonged exposure to elevated free fatty acids induces ß-cell failure (lipotoxicity) and contributes to the pathogenesis of type 2 diabetes. In vitro exposure of ß-cells to the saturated free fatty acid palmitate is a valuable model of lipotoxicity, reproducing features of ß-cell failure observed in type 2 diabetes. In order to map the ß-cell response to lipotoxicity, we combined RNA-sequencing of palmitate-treated human islets with iTRAQ proteomics of insulin-secreting INS-1E cells following a time course exposure to palmitate. RESULTS: Crossing transcriptome and proteome of palmitate-treated ß-cells revealed 85 upregulated and 122 downregulated genes at both transcript and protein level. Pathway analysis identified lipid metabolism, oxidative stress, amino-acid metabolism and cell cycle pathways among the most enriched palmitate-modified pathways. Palmitate induced gene expression changes compatible with increased free fatty acid mitochondrial import and ß-oxidation, decreased lipogenesis and modified cholesterol transport. Palmitate modified genes regulating endoplasmic reticulum (ER) function, ER-to-Golgi transport and ER stress pathways. Furthermore, palmitate modulated cAMP/protein kinase A (PKA) signaling, inhibiting expression of PKA anchoring proteins and downregulating the GLP-1 receptor. SLC7 family amino-acid transporters were upregulated in response to palmitate but this induction did not contribute to ß-cell demise. To unravel critical mediators of lipotoxicity upstream of the palmitate-modified genes, we identified overrepresented transcription factor binding sites and performed network inference analysis. These identified LXR, PPARα, FOXO1 and BACH1 as key transcription factors orchestrating the metabolic and oxidative stress responses to palmitate. CONCLUSIONS: This is the first study to combine transcriptomic and sensitive time course proteomic profiling of palmitate-exposed ß-cells. Our results provide comprehensive insight into gene and protein expression changes, corroborating and expanding beyond previous findings. The identification of critical drivers and pathways of the ß-cell lipotoxic response points to novel therapeutic targets for type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Apoptosis , Humanos , Palmitatos/toxicidad , Proteoma , Proteómica , Transcriptoma
4.
Nucleic Acids Res ; 46(19): 10302-10318, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30247717

RESUMEN

Transfer RNAs (tRNAs) are non-coding RNA molecules essential for protein synthesis. Post-transcriptionally they are heavily modified to improve their function, folding and stability. Intronic polymorphisms in CDKAL1, a tRNA methylthiotransferase, are associated with increased type 2 diabetes risk. Loss-of-function mutations in TRMT10A, a tRNA methyltransferase, are a monogenic cause of early onset diabetes and microcephaly. Here we confirm the role of TRMT10A as a guanosine 9 tRNA methyltransferase, and identify tRNAGln and tRNAiMeth as two of its targets. Using RNA interference and induced pluripotent stem cell-derived pancreatic ß-like cells from healthy controls and TRMT10A-deficient patients we demonstrate that TRMT10A deficiency induces oxidative stress and triggers the intrinsic pathway of apoptosis in ß-cells. We show that tRNA guanosine 9 hypomethylation leads to tRNAGln fragmentation and that 5'-tRNAGln fragments mediate TRMT10A deficiency-induced ß-cell death. This study unmasks tRNA hypomethylation and fragmentation as a hitherto unknown mechanism of pancreatic ß-cell demise relevant to monogenic and polygenic forms of diabetes.


Asunto(s)
Metilación de ADN , Diabetes Mellitus/genética , Células Secretoras de Insulina/metabolismo , Metiltransferasas/genética , ARN de Transferencia/metabolismo , Anciano , Animales , Apoptosis/genética , Muerte Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Fragmentación del ADN , Diabetes Mellitus/metabolismo , Ligamiento Genético , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Células Secretoras de Insulina/fisiología , Metiltransferasas/deficiencia , Metiltransferasas/metabolismo , Persona de Mediana Edad , Mutación , Ratas
5.
Hum Mol Genet ; 24(8): 2274-86, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25552656

RESUMEN

Friedreich's ataxia (FRDA) is a neurodegenerative disorder associated with cardiomyopathy and diabetes. Effective therapies for FRDA are an urgent unmet need; there are currently no options to prevent or treat this orphan disease. FRDA is caused by reduced expression of the mitochondrial protein frataxin. We have previously demonstrated that pancreatic ß-cell dysfunction and death cause diabetes in FRDA. This is secondary to mitochondrial dysfunction and apoptosis but the underlying molecular mechanisms are not known. Here we show that ß-cell demise in frataxin deficiency is the consequence of oxidative stress-mediated activation of the intrinsic pathway of apoptosis. The pro-apoptotic Bcl-2 family members Bad, DP5 and Bim are the key mediators of frataxin deficiency-induced ß-cell death. Importantly, the intrinsic pathway of apoptosis is also activated in FRDA patients' induced pluripotent stem cell-derived neurons. Interestingly, cAMP induction normalizes mitochondrial oxidative status and fully prevents activation of the intrinsic pathway of apoptosis in frataxin-deficient ß-cells and neurons. This preclinical study suggests that incretin analogs hold potential to prevent/delay both diabetes and neurodegeneration in FRDA.


Asunto(s)
Apoptosis , Ataxia de Friedreich/fisiopatología , Células Secretoras de Insulina/citología , Neuronas/citología , Animales , Línea Celular , Diabetes Mellitus/etiología , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Femenino , Ataxia de Friedreich/complicaciones , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Estrés Oxidativo , Ratas , Ratas Wistar , Frataxina
6.
EMBO J ; 31(6): 1405-26, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22293752

RESUMEN

In addition to genetic predisposition, environmental and lifestyle factors contribute to the pathogenesis of type 2 diabetes (T2D). Epigenetic changes may provide the link for translating environmental exposures into pathological mechanisms. In this study, we performed the first comprehensive DNA methylation profiling in pancreatic islets from T2D and non-diabetic donors. We uncovered 276 CpG loci affiliated to promoters of 254 genes displaying significant differential DNA methylation in diabetic islets. These methylation changes were not present in blood cells from T2D individuals nor were they experimentally induced in non-diabetic islets by exposure to high glucose. For a subgroup of the differentially methylated genes, concordant transcriptional changes were present. Functional annotation of the aberrantly methylated genes and RNAi experiments highlighted pathways implicated in ß-cell survival and function; some are implicated in cellular dysfunction while others facilitate adaptation to stressors. Together, our findings offer new insights into the intricate mechanisms of T2D pathogenesis, underscore the important involvement of epigenetic dysregulation in diabetic islets and may advance our understanding of T2D aetiology.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Anciano , Animales , Línea Celular , Islas de CpG , Dermatoglifia del ADN/métodos , Epigénesis Genética , Sitios Genéticos , Glucosa/metabolismo , Humanos , Regiones Promotoras Genéticas , Ratas , Transcripción Genética
7.
PLoS Genet ; 9(10): e1003888, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204302

RESUMEN

We describe a new syndrome of young onset diabetes, short stature and microcephaly with intellectual disability in a large consanguineous family with three affected children. Linkage analysis and whole exome sequencing were used to identify the causal nonsense mutation, which changed an arginine codon into a stop at position 127 of the tRNA methyltransferase homolog gene TRMT10A (also called RG9MTD2). TRMT10A mRNA and protein were absent in lymphoblasts from the affected siblings. TRMT10A is ubiquitously expressed but enriched in brain and pancreatic islets, consistent with the tissues affected in this syndrome. In situ hybridization studies showed that TRMT10A is expressed in human embryonic and fetal brain. TRMT10A is the mammalian ortholog of S. cerevisiae TRM10, previously shown to catalyze the methylation of guanine 9 (m(1)G9) in several tRNAs. Consistent with this putative function, in silico topology prediction indicated that TRMT10A has predominant nuclear localization, which we experimentally confirmed by immunofluorescence and confocal microscopy. TRMT10A localizes to the nucleolus of ß- and non-ß-cells, where tRNA modifications occur. TRMT10A silencing induces rat and human ß-cell apoptosis. Taken together, we propose that TRMT10A deficiency negatively affects ß-cell mass and the pool of neurons in the developing brain. This is the first study describing the impact of TRMT10A deficiency in mammals, highlighting a role in the pathogenesis of microcephaly and early onset diabetes. In light of the recent report that the type 2 diabetes candidate gene CDKAL1 is a tRNA methylthiotransferase, the findings in this family suggest broader relevance of tRNA methyltransferases in the pathogenesis of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Discapacidad Intelectual/genética , Metiltransferasas/genética , Microcefalia/genética , ARNt Metiltransferasas/genética , Adulto , Edad de Inicio , Animales , Apoptosis/genética , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Ligamiento Genético , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Masculino , Microcefalia/complicaciones , Microcefalia/patología , Mutación , Linaje , Ratas , Proteínas de Saccharomyces cerevisiae/genética , ARNt Metiltransferasas/deficiencia
8.
Diabetologia ; 58(10): 2307-16, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26099855

RESUMEN

AIMS/HYPOTHESIS: Proinflammatory cytokines contribute to beta cell damage in type 1 diabetes in part through activation of endoplasmic reticulum (ER) stress. In rat beta cells, cytokine-induced ER stress involves NO production and consequent inhibition of the ER Ca(2+) transporting ATPase sarco/endoplasmic reticulum Ca(2+) pump 2 (SERCA2B). However, the mechanisms by which cytokines induce ER stress and apoptosis in mouse and human pancreatic beta cells remain unclear. The purpose of this study is to elucidate the role of ER stress on cytokine-induced beta cell apoptosis in these three species and thus solve ongoing controversies in the field. METHODS: Rat and mouse insulin-producing cells, human pancreatic islets and human EndoC-ßH1 cells were exposed to the cytokines IL-1ß, TNF-α and IFN-γ, with or without NO inhibition. A global comparison of cytokine-modulated gene expression in human, mouse and rat beta cells was also performed. The chemical chaperone tauroursodeoxycholic acid (TUDCA) and suppression of C/EBP homologous protein (CHOP) were used to assess the role of ER stress in cytokine-induced apoptosis of human beta cells. RESULTS: NO plays a key role in cytokine-induced ER stress in rat islets, but not in mouse or human islets. Bioinformatics analysis indicated greater similarity between human and mouse than between human and rat global gene expression after cytokine exposure. The chemical chaperone TUDCA and suppression of CHOP or c-Jun N-terminal kinase (JNK) protected human beta cells against cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION: These observations clarify previous results that were discrepant owing to the use of islets from different species, and confirm that cytokine-induced ER stress contributes to human beta cell death, at least in part via JNK activation.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Ratas , Ratas Wistar , Ácido Tauroquenodesoxicólico/farmacología , Factor de Transcripción CHOP/farmacología , omega-N-Metilarginina/farmacología
9.
PLoS Genet ; 8(3): e1002552, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22412385

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic beta cells are killed by infiltrating immune cells and by cytokines released by these cells. Signaling events occurring in the pancreatic beta cells are decisive for their survival or death in diabetes. We have used RNA sequencing (RNA-seq) to identify transcripts, including splice variants, expressed in human islets of Langerhans under control conditions or following exposure to the pro-inflammatory cytokines interleukin-1ß (IL-1ß) and interferon-γ (IFN-γ). Based on this unique dataset, we examined whether putative candidate genes for T1D, previously identified by GWAS, are expressed in human islets. A total of 29,776 transcripts were identified as expressed in human islets. Expression of around 20% of these transcripts was modified by pro-inflammatory cytokines, including apoptosis- and inflammation-related genes. Chemokines were among the transcripts most modified by cytokines, a finding confirmed at the protein level by ELISA. Interestingly, 35% of the genes expressed in human islets undergo alternative splicing as annotated in RefSeq, and cytokines caused substantial changes in spliced transcripts. Nova1, previously considered a brain-specific regulator of mRNA splicing, is expressed in islets and its knockdown modified splicing. 25/41 of the candidate genes for T1D are expressed in islets, and cytokines modified expression of several of these transcripts. The present study doubles the number of known genes expressed in human islets and shows that cytokines modify alternative splicing in human islet cells. Importantly, it indicates that more than half of the known T1D candidate genes are expressed in human islets. This, and the production of a large number of chemokines and cytokines by cytokine-exposed islets, reinforces the concept of a dialog between pancreatic islets and the immune system in T1D. This dialog is modulated by candidate genes for the disease at both the immune system and beta cell level.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Interferón gamma , Interleucina-1beta , Islotes Pancreáticos , Transducción de Señal , Adulto , Anciano , Anciano de 80 o más Años , Empalme Alternativo/genética , Animales , Apoptosis , Línea Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Regulación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Sistema Inmunológico , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Ratas , Ratas Wistar , Análisis de Secuencia de ARN , Transcriptoma/genética
10.
J Neurochem ; 126 Suppl 1: 94-102, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23859345

RESUMEN

Diabetes is a common metabolic disorder in patients with Friedreich ataxia. In this Supplement article, we review the clinical data on diabetes in Friedreich ataxia, and the experimental data from rodent and in vitro models of the disease. Increased body adiposity and insulin resistance are frequently present in Friedreich ataxia, but pancreatic ß cell dysfunction and death are a conditio sine qua non for the loss of glucose tolerance and development of diabetes. The loss of frataxin function in mitochondria accounts for these pathogenic processes in Friedreich ataxia. Mitochondria are essential for the sensing of nutrients by the ß cell and for the generation of signals that trigger and amplify insulin secretion, known as stimulus-secretion coupling. Moreover, in the intrinsic pathway of apoptosis, pro-apoptotic signals converge on mitochondria, resulting in mitochondrial Bax translocation, membrane permeabilization, cytochrome c release and caspase cleavage. How and at which level frataxin deficiency impacts on these processes in ß cells is only partially understood. A better understanding of the molecular mechanisms mediating ß cell demise in Friedreich ataxia will pave the way for new therapeutic approaches.


Asunto(s)
Diabetes Mellitus/etiología , Ataxia de Friedreich/complicaciones , Animales , Supervivencia Celular/fisiología , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Ataxia de Friedreich/patología , Humanos , Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Proteínas de Unión a Hierro/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/complicaciones , Páncreas/patología , Riesgo , Frataxina
11.
Ann Neurol ; 72(6): 971-82, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23280845

RESUMEN

OBJECTIVE: Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused in almost all cases by homozygosity for a GAA trinucleotide repeat expansion in the frataxin gene. Frataxin is a mitochondrial protein involved in iron homeostasis. FRDA patients have a high prevalence of diabetes, the pathogenesis of which is not known. We aimed to evaluate the relative contribution of insulin resistance and ß-cell failure and the pathogenic mechanisms involved in FRDA diabetes. METHODS: Forty-one FRDA patients, 26 heterozygous carriers of a GAA expansion, and 53 controls underwent oral and intravenous glucose tolerance tests. ß-Cell proportion was quantified in postmortem pancreas sections from 9 unrelated FRDA patients. Using an in vitro disease model, we studied how frataxin deficiency affects ß-cell function and survival. RESULTS: FRDA patients had increased abdominal fat and were insulin resistant. This was not compensated for by increased insulin secretion, resulting in a markedly reduced disposition index, indicative of pancreatic ß-cell failure. Loss of glucose tolerance was driven by ß-cell dysfunction, which correlated with abdominal fatness. In postmortem pancreas sections, pancreatic islets of FRDA patients had a lower ß-cell content. RNA interference-mediated frataxin knockdown impaired glucose-stimulated insulin secretion and induced apoptosis in rat ß cells and human islets. Frataxin deficiency sensitized ß cells to oleate-induced and endoplasmic reticulum stress-induced apoptosis, which could be prevented by the incretins glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide. INTERPRETATION: Pancreatic ß-cell dysfunction is central to diabetes development in FRDA as a result of mitochondrial dysfunction and higher sensitivity to metabolic and endoplasmic reticulum stress-induced ß-cell death.


Asunto(s)
Diabetes Mellitus/etiología , Diabetes Mellitus/patología , Ataxia de Friedreich/complicaciones , Células Secretoras de Insulina/fisiología , Proteínas de Unión a Hierro/genética , Expansión de Repetición de Trinucleótido/genética , Tejido Adiposo/metabolismo , Adulto , Animales , Distribución de la Grasa Corporal , Metabolismo Energético/genética , Salud de la Familia , Femenino , Citometría de Flujo , Ataxia de Friedreich/genética , Prueba de Tolerancia a la Glucosa , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Resistencia a la Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Modelos Lineales , Masculino , Persona de Mediana Edad , Ratas , Frataxina
12.
Adv Sci (Weinh) ; 10(25): e2300063, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37382191

RESUMEN

Type 1 diabetes (T1D) is a complex autoimmune disease that develops in genetically susceptible individuals. Most T1D-associated single nucleotide polymorphisms (SNPs) are located in non-coding regions of the human genome. Interestingly, SNPs in long non-coding RNAs (lncRNAs) may result in the disruption of their secondary structure, affecting their function, and in turn, the expression of potentially pathogenic pathways. In the present work, the function of a virus-induced T1D-associated lncRNA named ARGI (Antiviral Response Gene Inducer) is characterized. Upon a viral insult, ARGI is upregulated in the nuclei of pancreatic ß cells and binds to CTCF to interact with the promoter and enhancer regions of IFNß and interferon-stimulated genes, promoting their transcriptional activation in an allele-specific manner. The presence of the T1D risk allele in ARGI induces a change in its secondary structure. Interestingly, the T1D risk genotype induces hyperactivation of type I IFN response in pancreatic ß cells, an expression signature that is present in the pancreas of T1D patients. These data shed light on the molecular mechanisms by which T1D-related SNPs in lncRNAs influence pathogenesis at the pancreatic ß cell level and opens the door for the development of therapeutic strategies based on lncRNA modulation to delay or avoid pancreatic ß cell inflammation in T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Activación Transcripcional/genética , Inflamación/metabolismo
13.
J Biol Chem ; 286(2): 929-41, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-20980260

RESUMEN

Cytokines produced by islet-infiltrating immune cells induce ß-cell apoptosis in type 1 diabetes. The IFN-γ-regulated transcription factors STAT1/IRF-1 have apparently divergent effects on ß-cells. Thus, STAT1 promotes apoptosis and inflammation, whereas IRF-1 down-regulates inflammatory mediators. To understand the molecular basis for these differential outcomes within a single signal transduction pathway, we presently characterized the gene networks regulated by STAT1 and IRF-1 in ß-cells. This was done by using siRNA approaches coupled to microarray analysis of insulin-producing cells exposed or not to IL-1ß and IFN-γ. Relevant microarray findings were further studied in INS-1E cells and primary rat ß-cells. STAT1, but not IRF-1, mediates the cytokine-induced loss of the differentiated ß-cell phenotype, as indicated by decreased insulin, Pdx1, MafA, and Glut2. Furthermore, STAT1 regulates cytokine-induced apoptosis via up-regulation of the proapoptotic protein DP5. STAT1 and IRF-1 have opposite effects on cytokine-induced chemokine production, with IRF-1 exerting negative feedback inhibition on STAT1 and downstream chemokine expression. The present study elucidates the transcriptional networks through which the IFN-γ/STAT1/IRF-1 axis controls ß-cell function/differentiation, demise, and islet inflammation.


Asunto(s)
Apoptosis/inmunología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Pancreatitis/inmunología , Pancreatitis/patología , Factor de Transcripción STAT1/inmunología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Retroalimentación Fisiológica/fisiología , Técnicas de Silenciamiento del Gen , Factor 1 Regulador del Interferón/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-1beta/farmacología , Masculino , Neuropéptidos/genética , Neuropéptidos/inmunología , ARN Interferente Pequeño , Ratas , Ratas Wistar , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transcripción Genética/inmunología
14.
Front Cell Dev Biol ; 10: 967765, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36060810

RESUMEN

In vitro differentiation of human induced pluripotent stem cells (iPSCs) into beta cells represents an important cell source for diabetes research. Here, we fully characterized iPSC-derived beta cell function in vitro and in vivo in humanized mice. Using a 7-stage protocol, human iPSCs were differentiated into islet-like aggregates with a yield of insulin-positive beta cells comparable to that of human islets. The last three stages of differentiation were conducted with two different 3D culture systems, rotating suspension or static microwells. In the latter, homogeneously small-sized islet-like aggregates were obtained, while in rotating suspension size was heterogeneous and aggregates often clumped. In vitro function was assessed by glucose-stimulated insulin secretion, NAD(P)H and calcium fluctuations. Stage 7 aggregates slightly increased insulin release in response to glucose in vitro. Aggregates were transplanted under the kidney capsule of NOD-SCID mice to allow for further in vivo beta cell maturation. In transplanted mice, grafts showed glucose-responsiveness and maintained normoglycemia after streptozotocin injection. In situ kidney perfusion assays showed modulation of human insulin secretion in response to different secretagogues. In conclusion, iPSCs differentiated with equal efficiency into beta cells in microwells compared to rotating suspension, but the former had a higher experimental success rate. In vitro differentiation generated aggregates lacking fully mature beta cell function. In vivo, beta cells acquired the functional characteristics typical of human islets. With this technology an unlimited supply of islet-like organoids can be generated from human iPSCs that will be instrumental to study beta cell biology and dysfunction in diabetes.

15.
Front Pharmacol ; 12: 768023, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34992533

RESUMEN

Rare diseases are life-threatening or chronically debilitating low-prevalent disorders caused by pathogenic mutations or particular environmental insults. Due to their high complexity and low frequency, important gaps still exist in their prevention, diagnosis, and treatment. Since new drug discovery is a very costly and time-consuming process, leading pharmaceutical companies show relatively low interest in orphan drug research and development due to the high cost of investments compared to the low market return of the product. Drug repurposing-based approaches appear then as cost- and time-saving strategies for the development of therapeutic opportunities for rare diseases. In this article, we discuss the scientific, regulatory, and economic aspects of the development of repurposed drugs for the treatment of rare neurodegenerative disorders with a particular focus on Huntington's disease, Friedreich's ataxia, Wolfram syndrome, and amyotrophic lateral sclerosis. The role of academia, pharmaceutical companies, patient associations, and foundations in the identification of candidate compounds and their preclinical and clinical evaluation will also be discussed.

16.
Int Rev Cell Mol Biol ; 359: 139-256, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33832649

RESUMEN

Monogenetic forms of diabetes represent 1%-5% of all diabetes cases and are caused by mutations in a single gene. These mutations, that affect genes involved in pancreatic ß-cell development, function and survival, or insulin regulation, may be dominant or recessive, inherited or de novo. Most patients with monogenic diabetes are very commonly misdiagnosed as having type 1 or type 2 diabetes. The severity of their symptoms depends on the nature of the mutation, the function of the affected gene and, in some cases, the influence of additional genetic or environmental factors that modulate severity and penetrance. In some patients, diabetes is accompanied by other syndromic features such as deafness, blindness, microcephaly, liver and intestinal defects, among others. The age of diabetes onset may also vary from neonatal until early adulthood manifestations. Since the different mutations result in diverse clinical presentations, patients usually need different treatments that range from just diet and exercise, to the requirement of exogenous insulin or other hypoglycemic drugs, e.g., sulfonylureas or glucagon-like peptide 1 analogs to control their glycemia. As a consequence, awareness and correct diagnosis are crucial for the proper management and treatment of monogenic diabetes patients. In this chapter, we describe mutations causing different monogenic forms of diabetes associated with inadequate pancreas development or impaired ß-cell function and survival, and discuss the molecular mechanisms involved in ß-cell demise.


Asunto(s)
Diabetes Mellitus/genética , Diabetes Mellitus/patología , Células Secretoras de Insulina/patología , Animales , Muerte Celular , Predisposición Genética a la Enfermedad , Humanos , Células Secretoras de Insulina/metabolismo , Mutación/genética , Factores de Transcripción/metabolismo
17.
Islets ; 13(3-4): 51-65, 2021 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-34241569

RESUMEN

Exposure of human pancreatic beta cells to pro-inflammatory cytokines or metabolic stressors is used to model events related to type 1 and type 2 diabetes, respectively. Quantitative real-time PCR is commonly used to quantify changes in gene expression. The selection of the most adequate reference gene(s) for gene expression normalization is an important pre-requisite to obtain accurate and reliable results. There are no universally applicable reference genes, and the human beta cell expression of commonly used reference genes can be altered by different stressors. Here we aimed to identify the most stably expressed genes in human beta cells to normalize quantitative real-time PCR gene expression.We used comprehensive RNA-sequencing data from the human pancreatic beta cell line EndoC-ßH1, human islets exposed to cytokines or the free fatty acid palmitate in order to identify the most stably expressed genes. Genes were filtered based on their level of significance (adjusted P-value >0.05), fold-change (|fold-change| <1.5) and a coefficient of variation <10%. Candidate reference genes were validated by quantitative real-time PCR in independent samples.We identified a total of 264 genes stably expressed in EndoC-ßH1 cells and human islets following cytokines - or palmitate-induced stress, displaying a low coefficient of variation. Validation by quantitative real-time PCR of the top five genes ARF1, CWC15, RAB7A, SIAH1 and VAPA corroborated their expression stability under most of the tested conditions. Further validation in independent samples indicated that the geometric mean of ACTB and VAPA expression can be used as a reliable normalizing factor in human beta cells.


Asunto(s)
Genómica/métodos , Células Secretoras de Insulina , Humanos , Células Secretoras de Insulina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Eur J Endocrinol ; 184(3): 455-468, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33486469

RESUMEN

OBJECTIVE: DNAJC3, also known as P58IPK, is an Hsp40 family member that interacts with and inhibits PKR-like ER-localized eIF2α kinase (PERK). Dnajc3 deficiency in mice causes pancreatic ß-cell loss and diabetes. Loss-of-function mutations in DNAJC3 cause early-onset diabetes and multisystemic neurodegeneration. The aim of our study was to investigate the genetic cause of early-onset syndromic diabetes in two unrelated patients, and elucidate the mechanisms of ß-cell failure in this syndrome. METHODS: Whole exome sequencing was performed and identified variants were confirmed by Sanger sequencing. DNAJC3 was silenced by RNAi in INS-1E cells, primary rat ß-cells, human islets, and induced pluripotent stem cell-derived ß-cells. ß-cell function and apoptosis were assessed, and potential mediators of apoptosis examined. RESULTS: The two patients presented with juvenile-onset diabetes, short stature, hypothyroidism, neurodegeneration, facial dysmorphism, hypoacusis, microcephaly and skeletal bone deformities. They were heterozygous compound and homozygous for novel loss-of-function mutations in DNAJC3. DNAJC3 silencing did not impair insulin content or secretion. Instead, the knockdown induced rat and human ß-cell apoptosis and further sensitized cells to endoplasmic reticulum stress, triggering mitochondrial apoptosis via the pro-apoptototic Bcl-2 proteins BIM and PUMA. CONCLUSIONS: This report confirms previously described features and expands the clinical spectrum of syndromic DNAJC3 diabetes, one of the five monogenic forms of diabetes pertaining to the PERK pathway of the endoplasmic reticulum stress response. DNAJC3 deficiency may lead to ß-cell loss through BIM- and PUMA-dependent activation of the mitochondrial pathway of apoptosis.


Asunto(s)
Apoptosis/genética , Diabetes Mellitus Tipo 1/genética , Proteínas del Choque Térmico HSP40/genética , Células Secretoras de Insulina/fisiología , Mitocondrias/metabolismo , Adolescente , Adulto , Factores de Edad , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/metabolismo , Mutación con Pérdida de Función , Masculino , Ratones , Mitocondrias/patología , Linaje , Ratas , Síndrome
19.
Methods Mol Biol ; 2128: 55-67, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180185

RESUMEN

Diabetes is a major public health problem: it is estimated that 420 million people are affected globally. Monogenic forms of diabetes are less common, but variants in monogenic diabetes genes have been shown to contribute to type 2 diabetes risk. In vitro and in vivo models of monogenic forms of diabetes related to the endoplasmic reticulum (ER) stress response provided compelling evidence on the role of ER stress and dysregulated ER stress signaling on ß cell demise in type 1 and type 2 diabetes. In this chapter, we describe the genetics, background, and phenotype of ER stress-related monogenic diabetes mouse models, and we comment on their advantages and disadvantages. We conclude that these mouse models are very useful tools for monogenic diabetes molecular pathogenesis studies, although there is a variability on the methodology that is used. Regarding the use of these models for therapeutic testing of ER stress modulators, a specific consideration should be given to the fact that they recapitulate some, but not all, the phenotypic characteristics of the human disease.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Intolerancia a la Glucosa/fisiopatología , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Mutantes , Fenotipo , Transducción de Señal
20.
JCI Insight ; 5(2)2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-31877117

RESUMEN

Friedreich ataxia is an autosomal recessive neurodegenerative disease associated with a high diabetes prevalence. No treatment is available to prevent or delay disease progression. Friedreich ataxia is caused by intronic GAA trinucleotide repeat expansions in the frataxin-encoding FXN gene that reduce frataxin expression, impair iron-sulfur cluster biogenesis, cause oxidative stress, and result in mitochondrial dysfunction and apoptosis. Here we examined the metabolic, neuroprotective, and frataxin-inducing effects of glucagon-like peptide-1 (GLP-1) analogs in in vivo and in vitro models and in patients with Friedreich ataxia. The GLP-1 analog exenatide improved glucose homeostasis of frataxin-deficient mice through enhanced insulin content and secretion in pancreatic ß cells. Exenatide induced frataxin and iron-sulfur cluster-containing proteins in ß cells and brain and was protective to sensory neurons in dorsal root ganglia. GLP-1 analogs also induced frataxin expression, reduced oxidative stress, and improved mitochondrial function in Friedreich ataxia patients' induced pluripotent stem cell-derived ß cells and sensory neurons. The frataxin-inducing effect of exenatide was confirmed in a pilot trial in Friedreich ataxia patients, showing modest frataxin induction in platelets over a 5-week treatment course. Taken together, GLP-1 analogs improve mitochondrial function in frataxin-deficient cells and induce frataxin expression. Our findings identify incretin receptors as a therapeutic target in Friedreich ataxia.


Asunto(s)
Exenatida/farmacología , Ataxia de Friedreich/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Mitocondrias/metabolismo , Adolescente , Adulto , Anciano , Animales , Encéfalo/patología , Cerebelo/patología , Modelos Animales de Enfermedad , Exenatida/uso terapéutico , Femenino , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Ganglios Espinales/patología , Técnicas de Sustitución del Gen , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Hierro/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Expansión de Repetición de Trinucleótido , Adulto Joven , Frataxina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA