Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Exp Physiol ; 106(7): 1482-1497, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33913203

RESUMEN

NEW FINDINGS: What is the central question of this study? Type 1 diabetes mellitus (T1D) leads to hyperglycaemia owing to pancreatic ß-cell destruction by the immune system. Physical exercise has been shown to have potentially beneficial protective roles against cytokine-induced pancreatic ß-cell death, but its benefits are yet to be proved and should be understood better, especially in the islet environment. What is the main finding and its importance? Physical exercise protects against ß-cell loss in a well-described animal model for T1D, induced by multiple low doses of streptozotocin. This seems to be related to reduced cytokine-induced ß-cell death and increased islet cell proliferation. Contributions of islet neogenesis and/or transdifferentiation of pancreatic non-ß-cells into ß-cells cannot be excluded. ABSTRACT: Physical exercise has beneficial effects on pancreatic ß-cell function and survival in a pro-inflammatory environment. Although these effects have been linked to decreased islet inflammation and modulation of pro-apoptotic pathways, little is known about the islet microenvironment. Our aim was to evaluate the effects of physical exercise in islet histomorphology in a mouse model of type 1 diabetes mellitus induced by multiple low doses of streptozotocin. As expected, induction of type 1 diabetes mellitus led to ß-cell loss and, consequently, decreased islet area. Interestingly, although the decrease in islet area was not prevented by physical exercise, this was not the case for the decrease in ß-cell mass. This was probably related to induction of ß-cell regeneration, because we observed increased proliferation and regeneration markers, such as Ki67 and Pcna, in islets of trained mice. These were found in the central and peripheral regions of the islets. An increase in the percentage of α- and δ-cells in these conditions, combined with an increase in proliferation and Pax4 labelling in peripheral regions, suggest that ß-cell regeneration might also occur by transdifferentiation. This agrees with the presence of cells double stained for insulin and glucagon only in islets of diabetic trained mice. In addition, this group had more extra-islet insulin-positive cells and islets associated with ducts than diabetic mice. Physical exercise also decreased nuclear factor-κB activation in islet cells of diabetic trained compared with diabetic untrained mice, indicating a decrease in pro-inflammatory cytokine-induced ß-cell death. Taken together, these findings indicate that preservation of ß-cell mass induced by physical exercise involves an increase in ß-cell replication and decrease in ß-cell death, together with islet neogenesis and islet cell transdifferentiation.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Glucagón/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones
2.
Diabetologia ; 59(3): 512-21, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26634571

RESUMEN

AIMS/HYPOTHESIS: Activation of the transcription factor nuclear factor (NF)-κB by proinflammatory cytokines plays an important role in beta cell demise in type 1 diabetes. Two main signalling pathways are known to activate NF-κB, namely the canonical and the non-canonical pathways. Up to now, studies on the role of NF-κB activation in beta cells have focused on the canonical pathway. The aim of this study was to investigate whether cytokines activate the non-canonical pathway in beta cells, how this pathway is regulated and the consequences of its activation on beta cell fate. METHODS: NF-κB signalling was analysed by immunoblotting, promoter reporter assays and real-time RT-PCR, after knockdown or overexpression of key genes/proteins. INS-1E cells, FACS-purified rat beta cells and the human beta cell line EndoC-ßH1 exposed to cytokines were used as models. RESULTS: IL-1ß plus IFN-γ induced stabilisation of NF-κB-inducing kinase and increased the expression and cleavage of p100 protein, culminating in the nuclear translocation of p52, the hallmark of the non-canonical signalling. This activation relied on different crosstalks between the canonical and non-canonical pathways, some of which were beta cell specific. Importantly, cytokine-mediated activation of the non-canonical pathway controlled the expression of 'late' NF-κB-dependent genes, regulating both pro-apoptotic and inflammatory responses, which are implicated in beta cell loss in early type 1 diabetes. CONCLUSIONS/INTERPRETATION: The atypical activation of the non-canonical NF-κB pathway by proinflammatory cytokines constitutes a novel 'feed-forward' mechanism that contributes to the particularly pro-apoptotic effect of NF-κB in beta cells.


Asunto(s)
Citocinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , FN-kappa B/metabolismo , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Inmunoprecipitación , Masculino , Óxido Nítrico/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
3.
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
4.
Proc Natl Acad Sci U S A ; 108(37): E681-8, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21705657

RESUMEN

Type 1 diabetes (T1D) is a complex disease characterized by the loss of insulin-secreting ß-cells. Although the disease has a strong genetic component, and several loci are known to increase T1D susceptibility risk, only few causal genes have currently been identified. To identify disease-causing genes in T1D, we performed an in silico "phenome-interactome analysis" on a genome-wide linkage scan dataset. This method prioritizes candidates according to their physical interactions at the protein level with other proteins involved in diabetes. A total of 11 genes were predicted to be likely disease genes in T1D, including the INS gene. An unexpected top-scoring candidate gene was huntingtin-interacting protein (HIP)-14/ZDHHC17. Immunohistochemical analysis of pancreatic sections demonstrated that HIP14 is almost exclusively expressed in insulin-positive cells in islets of Langerhans. RNAi knockdown experiments established that HIP14 is an antiapoptotic protein required for ß-cell survival and glucose-stimulated insulin secretion. Proinflammatory cytokines (IL-1ß and IFN-γ) that mediate ß-cell dysfunction in T1D down-regulated HIP14 expression in insulin-secreting INS-1 cells and in isolated rat and human islets. Overexpression of HIP14 was associated with a decrease in IL-1ß-induced NF-κB activity and protection against IL-1ß-mediated apoptosis. Our study demonstrates that the current network biology approach is a valid method to identify genes of importance for T1D and may therefore embody the basis for more rational and targeted therapeutic approaches.


Asunto(s)
Apoptosis , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adolescente , Adulto , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Niño , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/genética , Femenino , Predisposición Genética a la Enfermedad , Glucosa/farmacología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Interleucina-1beta/farmacología , Masculino , Ratones , Persona de Mediana Edad , FN-kappa B/metabolismo , Polimorfismo de Nucleótido Simple/genética , Unión Proteica/efectos de los fármacos , Ratas , Factores de Transcripción/metabolismo , Adulto Joven
5.
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
6.
Hum Mol Genet ; 19(1): 135-46, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19825843

RESUMEN

beta-Cell destruction in type 1 diabetes (T1D) is at least in part consequence of a 'dialog' between beta-cells and immune system. This dialog may be affected by the individual's genetic background. We presently evaluated whether modulation of MDA5 and PTPN2, two candidate genes for T1D, affects beta-cell responses to double-stranded RNA (dsRNA), a by-product of viral replication. These genes were selected following comparison between known candidate genes for T1D and genes expressed in pancreatic beta-cells, as identified in previous array analysis. INS-1E cells and primary fluorescence-activated cell sorting-purified rat beta-cells were transfected with small interference RNAs (siRNAs) targeting MDA5 or PTPN2 and subsequently exposed to intracellular synthetic dsRNA (polyinosinic-polycitidilic acid-PIC). Real-time RT-PCR, western blot and viability assays were performed to characterize gene/protein expression and viability. PIC increased MDA5 and PTPN2 mRNA expression, which was inhibited by the specific siRNAs. PIC triggered apoptosis in INS-1E and primary beta-cells and this was augmented by PTPN2 knockdown (KD), although inhibition of MDA5 did not modify PIC-induced apoptosis. In contrast, MDA5 silencing decreased PIC-induced cytokine and chemokine expression, although inhibition of PTPN2 induced minor or no changes in these inflammatory mediators. These findings indicate that changes in MDA5 and PTPN2 expression modify beta-cell responses to dsRNA. MDA5 regulates inflammatory signals, whereas PTPN2 may function as a defence mechanism against pro-apoptotic signals generated by dsRNA. These two candidate genes for T1D may thus modulate beta-cell apoptosis and/or local release of inflammatory mediators in the course of a viral infection by acting, at least in part, at the pancreatic beta-cell level.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , ARN Bicatenario/farmacología , Virus/metabolismo , Animales , Apoptosis/efectos de los fármacos , Quimiocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Interferón beta/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , FN-kappa B/metabolismo , Poli I-C/farmacología , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar
7.
J Biol Chem ; 285(26): 19910-20, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20421300

RESUMEN

Type 1 diabetes is an autoimmune disorder characterized by chronic inflammation and pancreatic beta-cell loss. Here, we demonstrate that the proinflammatory cytokine interleukin-1beta, combined with interferon-gamma, induces the expression of the Bcl-2 homology 3 (BH3)-only activator PUMA (p53 up-regulated modulator of apoptosis) in beta-cells. Transcriptional activation of PUMA is regulated by nuclear factor-kappaB and endoplasmic reticulum stress but is independent of p53. PUMA activation leads to mitochondrial Bax translocation, cytochrome c release, and caspase-3 cleavage resulting in beta-cell demise. The antiapoptotic Bcl-XL protein is localized mainly at the mitochondria of the beta-cells and antagonizes PUMA action, but Bcl-XL is inactivated by the BH3-only sensitizer DP5/Hrk in cytokine-exposed beta-cells. Moreover, a pharmacological mimic of the BH3-only sensitizer Bad, which inhibits Bcl-XL and Bcl-2, induces PUMA-dependent beta-cell death and potentiates cytokine-induced apoptosis. Our data support a hierarchical activation of BH3-only proteins controlling the intrinsic pathway of beta-cell apoptosis in the context of inflammation and type 1 diabetes.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Citocinas/farmacología , Retículo Endoplásmico/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Sitios de Unión/genética , Compuestos de Bifenilo/farmacología , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Mutación , FN-kappa B/metabolismo , Nitrofenoles/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
8.
Cells ; 10(12)2021 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-34943836

RESUMEN

A high caloric intake, rich in saturated fats, greatly contributes to the development of obesity, which is the leading risk factor for type 2 diabetes (T2D). A persistent caloric surplus increases plasma levels of fatty acids (FAs), especially saturated ones, which were shown to negatively impact pancreatic ß-cell function and survival in a process called lipotoxicity. Lipotoxicity in ß-cells activates different stress pathways, culminating in ß-cells dysfunction and death. Among all stresses, endoplasmic reticulum (ER) stress and oxidative stress have been shown to be strongly correlated. One main source of oxidative stress in pancreatic ß-cells appears to be the reactive oxygen species producer NADPH oxidase (NOX) enzyme, which has a role in the glucose-stimulated insulin secretion and in the ß-cell demise during both T1 and T2D. In this review, we focus on the acute and chronic effects of FAs and the lipotoxicity-induced ß-cell failure during T2D development, with special emphasis on the oxidative stress induced by NOX, the ER stress, and the crosstalk between NOX and ER stress.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/patología , Lípidos/toxicidad , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Lípidos/química , Estrés Oxidativo/efectos de los fármacos
9.
Antioxidants (Basel) ; 10(8)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34439552

RESUMEN

In type 1 diabetes (T1D) development, proinflammatory cytokines (PIC) released by immune cells lead to increased reactive oxygen species (ROS) production in ß-cells. Nonetheless, the temporality of the events triggered and the role of different ROS sources remain unclear. Isolated islets from C57BL/6J wild-type (WT), NOX1 KO and NOX2 KO mice were exposed to a PIC combination. We show that cytokines increase O2•- production after 2 h in WT and NOX1 KO but not in NOX2 KO islets. Using transgenic mice constitutively expressing a genetically encoded compartment specific H2O2 sensor, we show, for the first time, a transient increase of cytosolic/nuclear H2O2 in islet cells between 4 and 5 h during cytokine exposure. The H2O2 increase coincides with the intracellular NAD(P)H decrease and is absent in NOX2 KO islets. NOX2 KO confers better glucose tolerance and protects against cytokine-induced islet secretory dysfunction and death. However, NOX2 absence does not counteract the cytokine effects in ER Ca2+ depletion, Store-Operated Calcium Entry (SOCE) increase and ER stress. Instead, the activation of ER stress precedes H2O2 production. As early NOX2-driven ROS production impacts ß-cells' function and survival during insulitis, NOX2 might be a potential target for designing therapies against early ß-cell dysfunction in the context of T1D onset.

10.
Free Radic Biol Med ; 162: 1-13, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33249137

RESUMEN

Modern lifestyles, including lack of physical activity and poor nutritional habits, are driving the rapidly increasing prevalence of obesity and type 2 diabetes. Increased levels of free fatty acids (FFAs), particularly saturated FFAs, in obese individuals have been linked to pancreatic ß-cell failure. This process, termed lipotoxicity, involves activation of several stress responses, including ER stress and oxidative stress. However, the molecular underpinnings and causal relationships between the disparate stress responses remain unclear. Here we employed transgenic mice, expressing a genetically-encoded cytosolic H2O2 sensor, roGFP2-Orp1, to monitor dynamic changes in H2O2 levels in pancreatic islets in response to chronic palmitate exposure. We identified a transient increase in H2O2 levels from 4 to 8 h after palmitate addition, which was mirrored by a concomitant decrease in cellular NAD(P)H levels. Intriguingly, islets isolated from NOX2 knock-out mice displayed no H2O2 transient upon chronic palmitate treatment. Furthermore, NOX2 knockout rescued palmitate-dependent impairment of insulin secretion, calcium homeostasis and viability. Chemical inhibition of NOX activity protected islets from palmitate-induced impairment in insulin secretion, however had no detectable impact upon the induction of ER stress. In summary, our results reveal that transient NOX2-dependent H2O2 production is a likely cause of early palmitate-dependent lipotoxic effects.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Peróxido de Hidrógeno , Insulina , Ratones , NADPH Oxidasa 2/genética , Palmitatos/toxicidad
11.
Front Endocrinol (Lausanne) ; 11: 599165, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324349

RESUMEN

ARHGAP21 is a RhoGAP protein implicated in the modulation of insulin secretion and energy metabolism. ARHGAP21 transient-inhibition increase glucose-stimulated insulin secretion (GSIS) in neonatal islets; however, ARHGAP21 heterozygote mice have a reduced insulin secretion. These discrepancies are not totally understood, and it might be related to functional maturation of beta cells and peripheral sensitivity. Here, we investigated the real ARHGAP21 role in the insulin secretion process using an adult mouse model of acute ARHGAP21 inhibition, induced by antisense. After ARHGAP21 knockdown induction by antisense injection in 60-day old male mice, we investigated glucose and insulin tolerance test, glucose-induced insulin secretion, glucose-induced intracellular calcium dynamics, and gene expression. Our results showed that ARHGAP21 acts negatively in the GSIS of adult islet. This effect seems to be due to the modulation of important points of insulin secretion process, such as the energy metabolism (PGC1α), Ca2+ signalization (SYTVII), granule-extrusion (SNAP25), and cell-cell interaction (CX36). Therefore, based on these finds, ARHGAP21 may be an important target in Diabetes Mellitus (DM) treatment.


Asunto(s)
Proteínas Activadoras de GTPasa/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Hiperinsulinismo/prevención & control , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Animales , Homeostasis , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Edulcorantes/farmacología
12.
Biochim Biophys Acta ; 1768(9): 2222-34, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17626783

RESUMEN

We have explored the threshold of tolerance of three unrelated cell types to treatments with potential cytoprotective peptides bound to Tat(48-57) and Antp(43-58) cell-permeable peptide carriers. Both Tat(48-57) and Antp(43-58) are well known for their good efficacy at crossing membranes of different cell types, their overall low toxicity, and their absence of leakage once internalised. Here, we show that concentrations of up to 100 microM of Tat(48-57) were essentially harmless in all cells tested, whereas Antp(43-58) was significantly more toxic. Moreover, all peptides bound to Tat(48-57) and Antp(43-58) triggered significant and length-dependent cytotoxicity when used at concentrations above 10 microM in all but one cell types (208F rat fibroblasts), irrespective of the sequence of the cargo. Absence of cytotoxicity in 208F fibroblasts correlated with poor intracellular peptide uptake, as monitored by confocal laser scanning fluorescence microscopy. Our data further suggest that the onset of cytotoxicity correlates with the activation of two intracellular stress signalling pathways, namely those involving JNK, and to a lesser extent p38 mitogen-activated protein kinases. These responses are of particular concern for cells that are especially sensitive to the activation of stress kinases. Collectively, these results indicate that in order to avoid unwanted and unspecific cytotoxicity, effector molecules bound to Tat(48-57) should be designed with the shortest possible sequence and the highest possible affinity for their binding partners or targets, so that concentrations below 10 microM can be successfully applied to cells without harm. Considering that cytotoxicity associated to Tat(48-57)- and Antp(43-58) bound peptide conjugates was not restricted to a particular type of cells, our data provide a general framework for the design of cell-penetrating peptides that may apply to broader uses of intracellular peptide and drug delivery.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Péptidos/administración & dosificación , Péptidos/farmacocinética , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Tasa de Depuración Metabólica , Peso Molecular , Péptidos/química , Ratas
13.
Diabetes ; 56(4): 1069-77, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17395747

RESUMEN

Endoplasmic reticulum stress-mediated apoptosis may play an important role in the destruction of pancreatic beta-cells, thus contributing to the development of type 1 and type 2 diabetes. One of the regulators of endoplasmic reticulum stress-mediated cell death is the CCAAT/enhancer binding protein (C/EBP) homologous protein (Chop). We presently studied the molecular regulation of Chop expression in insulin-producing cells (INS-1E) in response to three pro-apoptotic and endoplasmic reticulum stress-inducing agents, namely the cytokines interleukin-1beta + interferon-gamma, the free fatty acid palmitate, and the sarcoendoplasmic reticulum pump Ca(2+) ATPase blocker cyclopiazonic acid (CPA). Detailed mutagenesis studies of the Chop promoter showed differential regulation of Chop transcription by CPA, cytokines, and palmitate. Whereas palmitate- and cytokine-induced Chop expression was mediated via a C/EBP-activating transcription factor (ATF) composite and AP-1 binding sites, CPA induction required the C/EBP-ATF site and the endoplasmic reticulum stress response element. Cytokines, palmitate, and CPA induced eIF2alpha phosphorylation in INS-1E cells leading to activation of the transcription factor ATF4. Chop transcription in response to cytokines and palmitate depends on the binding of ATF4 and AP-1 to the Chop promoter, but distinct AP-1 dimers were formed by cytokines and palmitate. These results suggest a differential response of beta-cells to diverse endoplasmic reticulum stress inducers, leading to a differential regulation of Chop transcription.


Asunto(s)
Retículo Endoplásmico/fisiología , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/fisiología , Factor de Transcripción CHOP/genética , Transcripción Genética , Animales , Línea Celular , Cartilla de ADN , Genes Reporteros , Insulinoma , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Ratas , Mapeo Restrictivo , Transfección
14.
J Nutr Biochem ; 19(4): 216-28, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17618105

RESUMEN

Abscisic acid (ABA) is a natural phytohormone and peroxisome proliferator-activated receptor gamma (PPARgamma) agonist that significantly improves insulin sensitivity in db/db mice. Although it has become clear that obesity is associated with macrophage infiltration into white adipose tissue (WAT), the phenotype of adipose tissue macrophages (ATMs) and the mechanisms by which insulin-sensitizing compounds modulate their infiltration remain unknown. We used a loss-of-function approach to investigate whether ABA ameliorates insulin resistance through a mechanism dependent on immune cell PPARgamma. We characterized two phenotypically distinct ATM subsets in db/db mice based on their surface expression of F4/80. F4/80(hi) ATMs were more abundant and expressed greater concentrations of chemokine receptor (CCR) 2 and CCR5 when compared to F4/80(lo) ATMs. ABA significantly decreased CCR2(+) F4/80(hi) infiltration into WAT and suppressed monocyte chemoattractant protein-1 (MCP-1) expression in WAT and plasma. Furthermore, the deficiency of PPARgamma in immune cells, including macrophages, impaired the ability of ABA to suppress the infiltration of F4/80(hi) ATMs into WAT, to repress WAT MCP-1 expression and to improve glucose tolerance. We provide molecular evidence in vivo demonstrating that ABA improves insulin sensitivity and obesity-related inflammation by inhibiting MCP-1 expression and F4/80(hi) ATM infiltration through a PPARgamma-dependent mechanism.


Asunto(s)
Ácido Abscísico/farmacología , Tejido Adiposo Blanco/inmunología , Quimiocina CCL2/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina , Macrófagos/inmunología , PPAR gamma/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal , Quimiocina CCL2/genética , Inflamación/inmunología , Hígado/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos , Obesidad/metabolismo , PPAR gamma/deficiencia , Fenotipo , Triglicéridos/sangre
15.
J Mol Endocrinol ; 61(2): F1-F6, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29728424

RESUMEN

The prevalence of diabetes has reached 8.8% in worldwide population and is predicted to increase up to 10.4% by 2040. Thus, there is an urgent need for the development of means to treat or prevent this major disease. Due to its role in inflammatory responses, several studies demonstrated the importance of the transcription factor nuclear factor-κB (NF-κB) in both type 1 diabetes (T1D) and type 2 diabetes (T2D). The two major NF-κB pathways are the canonical and the non-canonical. The later pathway is activated by the NF-κB-inducing kinase (NIK) that triggers p100 processing into p52, which forms with RelB its main dimer. Cytokines mediating the activation of this pathway are present in the serum of T1D and T2D patients. Conversely, limited information is available regarding the role of the alternative pathway on diabetes development and ß-cell fate. In the present review, we will briefly describe the involvement of NF-κB on diabetes pathology and discuss new studies indicating an important role for the non-canonical NF-κB activation in ß-cell function and survival. The non-canonical NF-κB pathway is emerging as a novel potential target for the development of therapeutic strategies to treat or prevent diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , FN-kappa B/metabolismo , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Transducción de Señal/fisiología
16.
J Mol Endocrinol ; 61(1): 25-36, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29632026

RESUMEN

Type 1 diabetes is caused by an autoimmune assault that induces progressive beta-cell dysfunction and dead. Pro-inflammatory cytokines, such as interleukin 1 beta (IL1B), tumor necrosis factor (TNF) and interferon gamma (IFNG) contribute for beta-cell death, which involves the activation of the nuclear factor kappa B (NFκB) and c- Jun N-terminal kinase (JNK). Prolactin (PRL), a physiological mediator for beta-cell proliferation, was shown to protect beta cells against cytokines pro-apoptotic effects. We presently investigated the mechanisms involved in the protective effects of prolactin against cytokine-induced beta-cell death. The findings obtained indicate that STAT3 activation is involved in the anti-apoptotic role of PRL in rat beta cells. PRL prevents the activation of JNK via AKT and promotes a shift from expression of pro- to anti-apoptotic proteins downstream of the JNK cascade. Furthermore, PRL partially prevents the activation of NFκB and the transcription of its target genes IkBa, Fas, Mcp1, A20 and Cxcl10 and also decreases NO production. On the other hand, the pro-survival effects of PRL do not involve modulation of cytokine-induced endoplasmic reticulum stress. These results suggest that the beneficial effects of PRL in beta cells involve augmentation of anti-apoptotic mechanisms and, at the same time, reduction of pro-apoptotic effectors, rendering beta cells better prepared to deal with inflammatory insults. The better understanding of the pro-survival mechanisms modulated by PRL in beta cells can provide tools to prevent cell demise during an autoimmune attack or following islet transplantation.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Prolactina/farmacología , Animales , Western Blotting , Células Cultivadas , Femenino , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
17.
Mol Endocrinol ; 20(8): 1867-79, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16556731

RESUMEN

Cytokines, such as IL-1beta and TNF-alpha, contribute to pancreatic beta-cell death in type 1 diabetes mellitus. The transcription factor nuclear factor-kappaB (NF-kappaB) mediates cytokine-induced beta-cell apoptosis. Paradoxically, NF-kappaB has mostly antiapoptotic effects in other cell types. The cellular actions of NF-kappaB depend on the cell type, the nature and duration of the stimulus, the periodicity, and the degree of activity of the particular dimers involved. To clarify the reasons behind the proapoptotic effects of NF-kappaB in pancreatic beta-cells, we compared the pattern of cytokine-induced NF-kappaB activation between rat insulin-producing cells (INS-1E cells) and fibroblasts (208F cells). NF-kappaB activation was induced in INS-1E cells and in 208F cells after exposure to cytokines, but apoptosis was induced only in INS-1E cells, with a more pronounced proapoptotic effect of IL-1beta than of TNF-alpha. NF-kappaB activation in IL-1beta-exposed INS-1E cells was earlier and more marked as compared with TNF-alpha-exposed INS-1E cells or IL-1beta-exposed 208F cells. Both cytokines induced a prolonged (up to 48 h) and stable NF-kappaB activation in INS-1E cells, whereas IL-1beta induced an oscillatory NF-kappaB activation in 208F cells. p65/p65 and p65/p50 were the predominant NF-kappaB dimers in IL-1beta-exposed INS-1E cells and 208F cells, respectively. IL-1beta induced a differential usage of cis-elements in the inducible nitric oxide synthase promoter region in the two cell-lines and an increase in ERK1/2 activity in INS-1E cells but not in 208F cells. Cytokine-induced expression of IkappaB isoforms and other NF-kappaB target genes (Fas, MCP-1, and inducible nitric oxide synthase) was severalfold higher in INS-1E cells than in 208F cells. These results suggest that cytokine-induced NF-kappaB activation in insulin-producing cells is more rapid, marked, and sustained than in fibroblasts, which correlates with a more pronounced activation of downstream genes and a proapoptotic outcome.


Asunto(s)
Apoptosis/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , FN-kappa B/metabolismo , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Proteínas I-kappa B/metabolismo , Cinética , FN-kappa B/química , FN-kappa B/fisiología , Óxido Nítrico Sintasa de Tipo II/genética , Regiones Promotoras Genéticas , Desnaturalización Proteica , Isoformas de Proteínas/metabolismo , Ratas , Elementos de Respuesta/genética , Factores de Tiempo
18.
Diabetes ; 54(2): 452-61, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15677503

RESUMEN

Cytokines and free radicals are mediators of beta-cell death in type 1 diabetes. Under in vitro conditions, interleukin-1beta (IL-1beta) + gamma-interferon (IFN-gamma) induce nitric oxide (NO) production and apoptosis in rodent and human pancreatic beta-cells. We have previously shown, by microarray analysis of primary beta-cells, that IL-1beta + IFN-gamma decrease expression of the mRNA encoding for the sarcoendoplasmic reticulum pump Ca(2+) ATPase 2b (SERCA2b) while inducing expression of the endoplasmic reticulum stress-related and proapoptotic gene CHOP (C/EBP [CCAAT/enhancer binding protein] homologous protein). In the present study we show that cytokine-induced apoptosis and necrosis in primary rat beta-cells and INS-1E cells largely depends on NO production. IL-1beta + IFN-gamma, via NO synthesis, markedly decreased SERCA2b protein expression and depleted ER Ca(2+) stores. Of note, beta-cells showed marked sensitivity to apoptosis induced by SERCA blockers, as compared with fibroblasts. Cytokine-induced ER Ca(2+) depletion was paralleled by an NO-dependent induction of CHOP protein and activation of diverse components of the ER stress response, including activation of inositol-requiring ER-to-nucleus signal kinase 1alpha (IRE1alpha) and PRK (RNA-dependent protein kinase)-like ER kinase (PERK)/activating transcription factor 4 (ATF4), but not ATF6. In contrast, the ER stress-inducing agent thapsigargin triggered these four pathways in parallel. In conclusion, our results suggest that the IL-1beta + IFN-gamma-induced decrease in SERCA2b expression, with subsequent depletion of ER Ca(2+) and activation of the ER stress pathway, is a potential contributory mechanism to beta-cell death.


Asunto(s)
ATPasas Transportadoras de Calcio/genética , Calcio/fisiología , Citocinas/farmacología , Retículo Endoplásmico/enzimología , Islotes Pancreáticos/fisiología , Estrés Oxidativo/fisiología , Retículo Sarcoplasmático/enzimología , Animales , Secuencia de Bases , Cartilla de ADN , Retículo Endoplásmico/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/enzimología , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Retículo Sarcoplasmático/efectos de los fármacos , Tapsigargina/farmacología
19.
J Mol Endocrinol ; 57(1): R1-R17, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27067637

RESUMEN

Insulin-secreting pancreatic ß-cells are extremely dependent on their endoplasmic reticulum (ER) to cope with the oscillatory requirement of secreted insulin to maintain normoglycemia. Insulin translation and folding rely greatly on the unfolded protein response (UPR), an array of three main signaling pathways designed to maintain ER homeostasis and limit ER stress. However, prolonged or excessive UPR activation triggers alternative molecular pathways that can lead to ß-cell dysfunction and apoptosis. An increasing number of studies suggest a role of these pro-apoptotic UPR pathways in the downfall of ß-cells observed in diabetic patients. Particularly, the past few years highlighted a cross talk between the UPR and inflammation in the context of both type 1 (T1D) and type 2 diabetes (T2D). In this article, we describe the recent advances in research regarding the interplay between ER stress, the UPR, and inflammation in the context of ß-cell apoptosis leading to diabetes.


Asunto(s)
Estrés del Retículo Endoplásmico , Inflamación/metabolismo , Islotes Pancreáticos/metabolismo , Respuesta de Proteína Desplegada , Animales , Citocinas/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Susceptibilidad a Enfermedades , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Inflamasomas/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Chaperonas Moleculares/metabolismo , Terapia Molecular Dirigida , FN-kappa B/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada/efectos de los fármacos
20.
Mol Endocrinol ; 30(1): 48-61, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26652732

RESUMEN

Activation of the transcription factor nuclear factor kappa B (NFkB) contributes to ß-cell death in type 1 diabetes (T1D). Genome-wide association studies have identified the gene TNF-induced protein 3 (TNFAIP3), encoding for the zinc finger protein A20, as a susceptibility locus for T1D. A20 restricts NF-κB signaling and has strong antiapoptotic activities in ß-cells. Although the role of A20 on NF-κB inhibition is well characterized, its other antiapoptotic functions are largely unknown. By studying INS-1E cells and rat dispersed islet cells knocked down or overexpressing A20 and islets isolated from the ß-cell-specific A20 knockout mice, we presently demonstrate that A20 has broader effects in ß-cells that are not restricted to inhibition of NF-κB. These involves, suppression of the proapoptotic mitogen-activated protein kinase c-Jun N-terminal kinase (JNK), activation of survival signaling via v-akt murine thymoma viral oncogene homolog (Akt) and consequently inhibition of the intrinsic apoptotic pathway. Finally, in a cohort of T1D children, we observed that the risk allele of the rs2327832 single nucleotide polymorphism of TNFAIP3 predicted lower C-peptide and higher hemoglobin A1c (HbA1c) levels 12 months after disease onset, indicating reduced residual ß-cell function and impaired glycemic control. In conclusion, our results indicate a critical role for A20 in the regulation of ß-cell survival and unveil novel mechanisms by which A20 controls ß-cell fate. Moreover, we identify the single nucleotide polymorphism rs2327832 of TNFAIP3 as a possible prognostic marker for diabetes outcome in children with T1D.


Asunto(s)
Apoptosis/fisiología , Cisteína Endopeptidasas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Niño , Cisteína Endopeptidasas/genética , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Células Secretoras de Insulina/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Polimorfismo de Nucleótido Simple , Ratas , Transducción de Señal/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA