Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
J Clin Invest ; 131(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34343133

RESUMEN

Decreased skeletal muscle strength and mitochondrial dysfunction are characteristic of diabetes. The actions of insulin and IGF-1 through the insulin receptor (IR) and IGF-1 receptor (IGF1R) maintain muscle mass via suppression of forkhead box O (FoxO) transcription factors, but whether FoxO activation coordinates atrophy in concert with mitochondrial dysfunction is unknown. We show that mitochondrial respiration and complex I activity were decreased in streptozotocin (STZ) diabetic muscle, but these defects were reversed in muscle-specific FoxO1, -3, and -4 triple-KO (M-FoxO TKO) mice rendered diabetic with STZ. In the absence of systemic glucose or lipid abnormalities, muscle-specific IR KO (M-IR-/-) or combined IR/IGF1R KO (MIGIRKO) impaired mitochondrial respiration, decreased ATP production, and increased ROS. These mitochondrial abnormalities were not present in muscle-specific IR, IGF1R, and FoxO1, -3, and -4 quintuple-KO mice (M-QKO). Acute tamoxifen-inducible deletion of IR and IGF1R also decreased muscle pyruvate respiration, complex I activity, and supercomplex assembly. Although autophagy was increased when IR and IGF1R were deleted in muscle, mitophagy was not increased. Mechanistically, RNA-Seq revealed that complex I core subunits were decreased in STZ-diabetic and MIGIRKO muscle, and these changes were not present with FoxO KO in STZ-FoxO TKO and M-QKO mice. Thus, insulin-deficient diabetes or loss of insulin/IGF-1 action in muscle decreases complex I-driven mitochondrial respiration and supercomplex assembly in part by FoxO-mediated repression of complex I subunit expression.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Factores de Transcripción Forkhead/metabolismo , Músculo Esquelético/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Metabolismo Energético , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Masculino , Ratones , Ratones Noqueados , Mitocondrias Musculares/metabolismo , Modelos Biológicos , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Receptor de Insulina/deficiencia , Receptor de Insulina/genética
2.
Int J Radiat Oncol Biol Phys ; 110(2): 526-538, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33385497

RESUMEN

PURPOSE: Type II pneumocyte (alveolar epithelial cells type II [AECII]) senescence has been implicated in the progression of lung fibrosis. The capacity of senescent cells to modulate pulmonary macrophages to drive fibrosis is unexplored. Insulin-like growth factor-1 receptor (IGF-1R) signaling has been implicated as a regulator of senescence and aging. METHODS AND MATERIALS: Mice with an AECII-specific deletion of IGF-1R received thoracic irradiation (n ≥ 5 per condition), and the effect of IGF-1R deficiency on radiation-induced AECII senescence and macrophage polarization to an alternatively activated phenotype (M2) was investigated. IGF-1R signaling, macrophage polarization, and senescence were evaluated in surgically resected human lung (n = 63). RESULTS: IGF-1R deficient mice demonstrated reduced AECII senescence (senescent AECII/field; intact: 7.25% ± 3.5% [mean ± SD], deficient: 2.75% ± 2.8%, P = .0001), reduced accumulation of M2 macrophages (intact: 24.7 ± 2.2 cells/field, deficient: 15.5 ± 1.2 cells/field, P = .0086), and fibrosis (hydroxyproline content; intact: 71.9 ± 21.7 µg/lung, deficient: 31.7 ± 7.9, P = .0485) after irradiation. Senescent AECII enhanced M2 polarization in a paracrine fashion (relative Arg1 mRNA, 0 Gy: 1.0 ± 0.4, 17.5 Gy: 7.34 ± 0.5, P < .0001). Evaluation of surgical samples from patients treated with chemoradiation demonstrated increased expression of IGF-1 (unirradiated: 10.2% ± 4.9% area, irradiated: 15.1% ± 11.5%, P = .0377), p21 (unirradiated: 0.013 ± 0.02 histoscore, irradiated: 0.084 ± 0.09 histoscore, P = .0002), IL-13 (unirradiated: 13.7% ± 2.8% area, irradiated: 21.7% ± 3.8%, P < .0001), and M2 macrophages in fibrotic regions relative to nonfibrotic regions (unirradiated: 11.4 ± 12.2 CD163 + cells/core, irradiated: 43.1 ± 40.9 cells/core, P = .0011), consistent with findings from animal models of lung fibrosis. CONCLUSIONS: This study demonstrates that senescent AECII are necessary for the progression of pulmonary fibrosis and serve as a targetable, chronic stimuli for macrophage activation in fibrotic lung.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Polaridad Celular , Senescencia Celular/fisiología , Macrófagos Alveolares/fisiología , Fibrosis Pulmonar/etiología , Receptor IGF Tipo 1/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Senescencia Celular/efectos de la radiación , Quimioradioterapia , Eliminación de Gen , Humanos , Hidroxiprolina/análisis , Pulmón/metabolismo , Pulmón/efectos de la radiación , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Activación de Macrófagos , Macrófagos Alveolares/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/fisiopatología , Traumatismos Experimentales por Radiación/prevención & control , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética
3.
Am J Physiol Heart Circ Physiol ; 319(4): H730-H743, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32795184

RESUMEN

Insulin-like growth factor-1 (IGF-1) decreases atherosclerosis in apolipoprotein E (Apoe)-deficient mice when administered systemically. However, mechanisms for its atheroprotective effect are not fully understood. We generated endothelium-specific IGF-1 receptor (IGF1R)-deficient mice on an Apoe-deficient background to assess effects of IGF-1 on the endothelium in the context of hyperlipidemia-induced atherosclerosis. Endothelial deficiency of IGF1R promoted atherosclerotic burden, when animals were fed on a high-fat diet for 12 wk or normal chow for 12 mo. Under the normal chow feeding condition, the vascular relaxation response to acetylcholine was increased in the endothelial IGF1R-deficient aorta; however, feeding of a high-fat diet substantially attenuated the relaxation response, and there was no difference between endothelial IGF1R-deficient and control mice. The endothelium and its intercellular junctions provide a barrier function to the vasculature. In human aortic endothelial cells, IGF-1 upregulated occludin, claudin 5, VE-cadherin, JAM-A, and CD31 expression levels, and vice versa, specific IGF1R inhibitor, picropodophyllin, an IGF1R-neutralizing antibody (αIR3), or siRNA to IGF1R abolished the IGF-1 effects on junction and adherens proteins, suggesting that IGF-1 promoted endothelial barrier function. Moreover, endothelial transwell permeability assays indicated that inhibition of IGF-1 signaling elevated solute permeability through the monolayer of human aortic endothelial cells. In summary, endothelial IGF1R deficiency increases atherosclerosis, and IGF-1 positively regulates tight junction protein and adherens junction protein levels and endothelial barrier function. Our findings suggest that the elevation of the endothelial junction protein level is, at least in part, the mechanism for antiatherogenic effects of IGF-1.NEW & NOTEWORTHY Endothelial insulin-like growth factor-1 (IGF-1) receptor deficiency significantly elevated atherosclerotic burden in apolipoprotein E-deficient mice, mediated at least in part by downregulation of intercellular junction proteins and, thus, elevated endothelial permeability. This study revealed a novel role for IGF-1 in supporting endothelial barrier function. These findings suggest that IGF-1's ability to promote endothelial barrier function may offer a novel therapeutic strategy for vascular diseases such as atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Receptor IGF Tipo 1/deficiencia , Animales , Antígenos CD/metabolismo , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/patología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Placa Aterosclerótica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Células THP-1 , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
4.
Amino Acids ; 52(4): 543-553, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32236698

RESUMEN

The aim of the current study was to investigate whether doublecortin (DCX), insulin-like growth factor receptor 1 (IGF-1R) and metabotropic glutamate receptor 5 (mGluR5) levels are indeed modified in the aging rat hippocampal individual subareas (rather than total hippocampal tissue as in previous reports) at the protein and mRNA level and whether the methylation status contributes to these changes. Since the aging population is not homogeneous in terms of spatial memory performance, we examined whether changes in DCX, IGF-1R and mGluR5 are linked to cognitive aging. Aged (22 months) male Sprague Dawley rats were trained in the hole-board, a spatial memory task, and were subdivided according to performance to aged impaired and aged unimpaired groups. Age- and memory performance-dependent changes in mRNA steady-state levels, protein levels and DNA methylation status of DCX, IGF-1R and mGluR5 were evaluated by RT-PCR, immunoblotting and bisulfite pyrosequencing. Extending previous findings, we detected decreased DCX protein and mRNA levels in dentate gyrus (DG) of aged animals. IGF-1 signaling is a key event and herein we show that mRNA levels for IGF-1R were unchanged although reduced at the protein level. This finding may simply reflect that these protein levels are regulated at the level of protein synthesis as well as protein degradation. We provide evidence that promoter methylation is not involved in regulation of mRNA and protein levels of DCX, IGF-1R and mGluR5 during aging. Moreover, there was no significant difference between aged rats with impaired and aged rats with unimpaired memory at the protein and mRNA level. Findings propose that changes in the abovementioned protein levels may not be relevant for performance in the spatial memory task used in aged rats.


Asunto(s)
Hipocampo/metabolismo , Proteínas Asociadas a Microtúbulos/deficiencia , Neuropéptidos/deficiencia , Receptor IGF Tipo 1/deficiencia , Envejecimiento/metabolismo , Animales , Cognición , Metilación de ADN , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Masculino , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/genética , Neuropéptidos/análisis , Neuropéptidos/genética , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/análisis , Receptor IGF Tipo 1/genética , Receptor del Glutamato Metabotropico 5/análisis , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Memoria Espacial
5.
Arterioscler Thromb Vasc Biol ; 38(10): 2306-2317, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354209

RESUMEN

Objective- IGF-1 (insulin-like growth factor 1) is a major autocrine/paracrine growth factor, which promotes cell proliferation, migration, and survival. We have shown previously that IGF-1 reduced atherosclerosis and promoted features of stable atherosclerotic plaque in Apoe-/- mice-an animal model of atherosclerosis. The aim of this study was to assess effects of smooth muscle cell (SMC) IGF-1 signaling on the atherosclerotic plaque. Approach and Results- We generated Apoe-/- mice with IGF1R (IGF-1 receptor) deficiency in SMC and fibroblasts (SM22α [smooth muscle protein 22 α]-CreKI/IGF1R-flox mice). IGF1R was decreased in the aorta and adventitia of SM22α-CreKI/IGF1R-flox mice and also in aortic SMC, embryonic, skin, and lung fibroblasts isolated from SM22α-CreKI/IGF1R-flox mice. IGF1R deficiency downregulated collagen mRNA-binding protein LARP6 (La ribonucleoprotein domain family, member 6) and vascular collagen, and mice exhibited growth retardation. The high-fat diet-fed SM22α-CreKI/IGF1R-flox mice had increased atherosclerotic burden and inflammatory responses. α-SMA (α-smooth muscle actin)-positive plaque cells had reduced proliferation and elevated apoptosis. SMC/fibroblast-targeted decline in IGF-1 signaling decreased atherosclerotic plaque SMC, markedly depleted collagen, reduced plaque fibrous cap, and increased plaque necrotic cores. Aortic SMC isolated from SM22α-CreKI/IGF1R-flox mice had decreased cell proliferation, migration, increased sensitivity to apoptosis, and these effects were associated with disruption of IGF-1-induced Akt signaling. Conclusions- IGF-1 signaling in SMC and in fibroblast is a critical determinant of normal vascular wall development and atheroprotection.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica , Regiones Promotoras Genéticas , Receptor IGF Tipo 1/deficiencia , Actinas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Autoantígenos/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibrosis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/genética , Ribonucleoproteínas/metabolismo , Transducción de Señal , Antígeno SS-B
6.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30070361

RESUMEN

Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.


Asunto(s)
Receptores ErbB/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor de Insulina/deficiencia , Cáncer Papilar Tiroideo/metabolismo , Células Epiteliales Tiroideas/metabolismo , Neoplasias de la Tiroides/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal , Cáncer Papilar Tiroideo/patología , Células Epiteliales Tiroideas/patología , Neoplasias de la Tiroides/patología , Tirotropina/biosíntesis , Tirotropina/metabolismo
7.
Artif Cells Nanomed Biotechnol ; 46(2): 293-302, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28362176

RESUMEN

Here, we investigated the effects of dual delivery of IGF-1R siRNA and doxorubicin by chitosan nanoparticles on viability of A549 lung cancer cells line by utilization of MTT and qRT-PCR. Furthermore apoptosis and migration of treated cells were assessed by Annexin-PI and wound healing assays, respectively. The chitosan nanoparticles had about 176 nm size with zeta potential and polydispersive index about 11 mV and 0.3, respectively. The IGF-1R siRNA had synergistic effect on DOX-induced cytotoxicity and apoptosis in tumour cells. In addition, siRNA/DOX-loaded chitosan nanoparticles could significantly decrease migration and expressions of mmp9, VEGF and STAT3 in A549 cells.


Asunto(s)
Quitosano/química , Doxorrubicina/química , Neoplasias Pulmonares/patología , Nanopartículas/química , ARN Interferente Pequeño/química , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Células A549 , Apoptosis/efectos de los fármacos , Apoptosis/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Doxorrubicina/farmacología , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Metaloproteinasa 9 de la Matriz/genética , Invasividad Neoplásica , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética , Factor A de Crecimiento Endotelial Vascular/genética
8.
Brain ; 140(7): 2012-2027, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28595357

RESUMEN

Seminal studies using post-mortem brains of patients with Alzheimer's disease evidenced aberrant insulin-like growth factor 1 receptor (IGF1R) signalling. Addressing causality, work in animal models recently demonstrated that long-term suppression of IGF1R signalling alleviates Alzheimer's disease progression and promotes neuroprotection. However, the underlying mechanisms remain largely elusive. Here, we showed that genetically ablating IGF1R in neurons of the ageing brain efficiently protects from neuroinflammation, anxiety and memory impairments induced by intracerebroventricular injection of amyloid-ß oligomers. In our mutant mice, the suppression of IGF1R signalling also invariably led to small neuronal soma size, indicative of profound changes in cellular homeodynamics. To gain insight into transcriptional signatures leading to Alzheimer's disease-relevant neuronal defence, we performed genome-wide microarray analysis on laser-dissected hippocampal CA1 after neuronal IGF1R knockout, in the presence or absence of APP/PS1 transgenes. Functional analysis comparing neurons in early-stage Alzheimer's disease with IGF1R knockout neurons revealed strongly convergent transcriptomic signatures, notably involving neurite growth, cytoskeleton organization, cellular stress response and neurotransmission. Moreover, in Alzheimer's disease neurons, a high proportion of genes responding to Alzheimer's disease showed a reversed differential expression when IGF1R was deleted. One of the genes consistently highlighted in genome-wide comparison was the neurofilament medium polypeptide Nefm. We found that NEFM accumulated in hippocampus in the presence of amyloid pathology, and decreased to control levels under IGF1R deletion, suggesting that reorganized cytoskeleton likely plays a role in neuroprotection. These findings demonstrated that significant resistance of the brain to amyloid-ß can be achieved lifelong by suppressing neuronal IGF1R and identified IGF-dependent molecular pathways that coordinate an intrinsic program for neuroprotection against proteotoxicity. Our data also indicate that neuronal defences against Alzheimer's disease rely on an endogenous gene expression profile similar to the neuroprotective response activated by genetic disruption of IGF1R signalling. This study highlights neuronal IGF1R signalling as a relevant target for developing Alzheimer's disease prevention strategies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Región CA1 Hipocampal/metabolismo , Fármacos Neuroprotectores/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Transcriptoma , Envejecimiento/metabolismo , Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/administración & dosificación , Animales , Ansiedad/inducido químicamente , Ansiedad/complicaciones , Ansiedad/prevención & control , Encefalitis/inducido químicamente , Encefalitis/complicaciones , Encefalitis/prevención & control , Femenino , Infusiones Intraventriculares , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/prevención & control , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/metabolismo
9.
Sci Rep ; 7(1): 4290, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28655914

RESUMEN

IGF1R (Insulin-like Growth Factor 1 Receptor) is a tyrosine kinase with pleiotropic cellular functions. IGF activity maintains human lung homeostasis and is implicated in pulmonary diseases such as cancer, ARDS, COPD, asthma and fibrosis. Here we report that lung transcriptome analysis in mice with a postnatally-induced Igf1r gene deletion showed differentially expressed genes with potentially protective roles related to epigenetics, redox and oxidative stress. After bleomycin-induced lung injury, IGF1R-deficient mice demonstrated improved survival within a week. Three days post injury, IGF1R-deficient lungs displayed changes in expression of IGF system-related genes and reduced vascular fragility and permeability. Mutant lungs presented reduced inflamed area, down-regulation of pro-inflammatory markers and up-regulation of resolution indicators. Decreased inflammatory cell presence in BALF was reflected in diminished lung infiltration mainly affecting neutrophils, also corroborated by reduced neutrophil numbers in bone marrow, as well as reduced lymphocyte and alveolar macrophage counts. Additionally, increased SFTPC expression together with hindered HIF1A expression and augmented levels of Gpx8 indicate that IGF1R deficiency protects against alveolar damage. These findings identify IGF1R as an important player in murine acute lung inflammation, suggesting that targeting IGF1R may counteract the inflammatory component of many lung diseases.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/metabolismo , Bleomicina/efectos adversos , Inflamación/etiología , Inflamación/metabolismo , Receptor IGF Tipo 1/deficiencia , Lesión Pulmonar Aguda/patología , Animales , Biomarcadores , Permeabilidad Capilar , Proliferación Celular , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación
10.
J Cell Physiol ; 232(10): 2722-2730, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28112398

RESUMEN

Increasing number of studies have shown nuclear localization of the insulin-like growth factor 1 receptor (nIGF-1R) in tumor cells and its links to adverse clinical outcome in various cancers. Any obvious cell physiological roles of nIGF-1R have, however, still not been disclosed. Previously, we reported that IGF-1R translocates to cell nucleus and modulates gene expression by binding to enhancers, provided that the receptor is SUMOylated. In this study, we constructed stable transfectants of wild type IGF1R (WT) and triple-SUMO-site-mutated IGF1R (TSM) using igf1r knockout mouse fibroblasts (R-). Cell clones (R-WT and R-TSM) expressing equal amounts of IGF-1R were selected for experiments. Phosphorylation of IGF-1R, Akt, and Erk upon IGF-1 stimulation was equal in R-WT and R-TSM. WT was confirmed to enter nuclei. TSM did also undergo nuclear translocation, although to a lesser extent. This may be explained by that TSM heterodimerizes with insulin receptor, which is known to translocate to cell nuclei. R-WT proliferated substantially faster than R-TSM, which did not differ significantly from the empty vector control. Upon IGF-1 stimulation G1-S-phase progression of R-WT increased from 12 to 38%, compared to 13 to 20% of R-TSM. The G1-S progression of R-WT correlated with increased expression of cyclin D1, A, and CDK2, as well as downregulation of p27. This suggests that SUMO-IGF-1R affects upstream mechanisms that control and coordinate expression of cell cycle regulators. Further studies to identify such SUMO-IGF-1R dependent mechanisms seem important.


Asunto(s)
Proliferación Celular , Fibroblastos/metabolismo , Fase G1 , Receptor IGF Tipo 1/metabolismo , Receptores de Somatomedina/metabolismo , Fase S , Sumoilación , Animales , Células Cultivadas , Ciclina A/metabolismo , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genotipo , Ratones Noqueados , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Receptores de Somatomedina/deficiencia , Receptores de Somatomedina/genética , Transducción de Señal , Factores de Tiempo , Transfección
11.
Physiol Rep ; 4(23)2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27923977

RESUMEN

The major site of fructose metabolism in the kidney is the proximal tubule (PT). To test whether insulin and/or IGF1 signaling in the PT is involved in renal structural/functional responses to dietary fructose, we bred mice with dual knockout (KO) of the insulin receptor (IR) and the IGF1 receptor (IGF1R) in PT by Cre-lox recombination, using a γ-glutamyl transferase promoter. KO mice had slightly (~10%) reduced body and kidney weights, as well as, a reduction in mean protein-to-DNA ratio in kidney cortex suggesting smaller cell size. Under control diet, IR and IGF1R protein band densities were 30-50% (P < 0.05) lower than WT, and the relative difference was greater in male animals. Male, but not female KO, also had significantly reduced band densities for Akt (protein kinase B), phosphorylated AktT308 and IRY1162/1163 A high-fructose diet (1-month) led to a significant increase in kidney weight in WT males (12%), but not in KO males or in either genotype of female mice. Kidney enlargement in the WT males was accompanied by a small, insignificant fall in protein-to-DNA ratio, supporting hyperplasia rather than hypertrophy. Fructose feeding of male WT mice led to significantly higher sodium bicarbonate exchanger (NBCe1), sodium hydrogen exchanger (NHE3), sodium phosphate co-transporter (NaPi-2), and transforming growth factor-ß (TGF-ß) abundances, as compared to male KO, suggesting elevated transport capacity and an early feature of fibrosis may have accompanied the renal enlargement. Overall, IR and/or IGF1R appear to have a role in PT cell size and enlargement in response to high-fructose diet.


Asunto(s)
Fructosa/farmacología , Túbulos Renales Proximales/metabolismo , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Animales , Dieta , Femenino , Fructosa/administración & dosificación , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/deficiencia , Receptor de Insulina/metabolismo , Factores Sexuales , Simportadores de Sodio-Bicarbonato/genética , Simportadores de Sodio-Bicarbonato/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/genética , Proteínas Cotransportadoras de Sodio-Fosfato/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
12.
Nature ; 539(7630): 570-574, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27820945

RESUMEN

Professional phagocytes (such as macrophages) and non-professional phagocytes (such as epithelial cells) clear billions of apoptotic cells and particles on a daily basis. Although professional and non-professional macrophages reside in proximity in most tissues, whether they communicate with each other during cell clearance, and how this might affect inflammation, is not known. Here we show that macrophages, through the release of a soluble growth factor and microvesicles, alter the type of particles engulfed by non-professional phagocytes and influence their inflammatory response. During phagocytosis of apoptotic cells or in response to inflammation-associated cytokines, macrophages released insulin-like growth factor 1 (IGF-1). The binding of IGF-1 to its receptor on non-professional phagocytes redirected their phagocytosis, such that uptake of larger apoptotic cells was reduced whereas engulfment of microvesicles was increased. IGF-1 did not alter engulfment by macrophages. Macrophages also released microvesicles, whose uptake by epithelial cells was enhanced by IGF-1 and led to decreased inflammatory responses by epithelial cells. Consistent with these observations, deletion of IGF-1 receptor in airway epithelial cells led to exacerbated lung inflammation after allergen exposure. These genetic and functional studies reveal that IGF-1- and microvesicle-dependent communication between macrophages and epithelial cells can critically influence the magnitude of tissue inflammation in vivo.


Asunto(s)
Células Epiteliales/citología , Macrófagos/citología , Fagocitos/citología , Fagocitosis , Neumonía , Alérgenos/inmunología , Animales , Apoptosis , Comunicación Celular , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Fibroblastos/citología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Fagocitos/inmunología , Fagocitos/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/metabolismo , Sistema Respiratorio/citología , Somatomedinas/metabolismo
13.
Int J Biochem Cell Biol ; 78: 130-140, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417237

RESUMEN

As an important type of somatic cell, granulosa cells play a major role in deciding the fate of follicles. Therefore, analyses of granulosa cell apoptosis and follicular atresia have become hotspots of animal research. Autophagy is a cellular catabolic mechanism that protects cells from stress conditions, including starvation, hypoxia, and accumulation of misfolded proteins. However, the relationship between autophagy and apoptosis in granulosa cells is not well known. Here, we demonstrate that let-7g regulates the mouse granulosa cell autophagy signaling pathway by inhibiting insulin-like growth factor 1 receptor expression and affecting the phosphorylation of protein kinase B/mammalian target of rapamycin. Small interference-mediated knockdown of insulin-like growth factor 1 receptor significantly promoted autophagy signaling of mouse granulosa cells. In contrast, overexpression of insulin-like growth factor 1 receptor in mouse granulosa cells attenuated autophagy activity in the presence of let-7g. In addition, overexpression of let-7g increased the apoptosis rate, as indicated by an increased number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cells. Finally, 3-methyladenine as well as the lysosomal enzyme inhibitor chloroquine partially blocked apoptosis. In summary, this study demonstrates that let-7g regulates autophagy in mouse granulosa cells by targeting insulin-like growth factor 1 receptor and downregulating protein kinase B/mammalian target of rapamycin signaling, and that mouse granulosa cell autophagy induced by let-7g participates in apoptosis.


Asunto(s)
Autofagia/genética , Células de la Granulosa/citología , MicroARNs/genética , Receptor IGF Tipo 1/genética , Animales , Secuencia de Bases , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Células de la Granulosa/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/deficiencia , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
14.
Circulation ; 133(23): 2263-78, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27154724

RESUMEN

BACKGROUND: We have previously shown that systemic infusion of insulin-like growth factor-1 (IGF-1) exerts anti-inflammatory and antioxidant effects and reduces atherosclerotic burden in apolipoprotein E (Apoe)-deficient mice. Monocytes/macrophages express high levels of IGF-1 receptor (IGF1R) and play a pivotal role in atherogenesis, but the potential effects of IGF-1 on their function are unknown. METHODS AND RESULTS: To determine mechanisms whereby IGF-1 reduces atherosclerosis and to explore the potential involvement of monocytes/macrophages, we created monocyte/macrophage-specific IGF1R knockout (MΦ-IGF1R-KO) mice on an Apoe(-/-) background. We assessed atherosclerotic burden, plaque features of stability, and monocyte recruitment to atherosclerotic lesions. Phenotypic changes of IGF1R-deficient macrophages were investigated in culture. MΦ-IGF1R-KO significantly increased atherosclerotic lesion formation, as assessed by Oil Red O staining of en face aortas and aortic root cross-sections, and changed plaque composition to a less stable phenotype, characterized by increased macrophage and decreased α-smooth muscle actin-positive cell population, fibrous cap thinning, and decreased collagen content. Brachiocephalic artery lesions of MΦ-IGF1R-KO mice had histological features implying plaque vulnerability. Macrophages isolated from MΦ-IGF1R-KO mice showed enhanced proinflammatory responses on stimulation by interferon-γ and oxidized low-density lipoprotein and elevated antioxidant gene expression levels. Moreover, IGF1R-deficient macrophages had decreased expression of ABCA1 and ABCG1 and reduced lipid efflux. CONCLUSIONS: Our data indicate that macrophage IGF1R signaling suppresses macrophage and foam cell accumulation in lesions and reduces plaque vulnerability, providing a novel mechanism whereby IGF-1 exerts antiatherogenic effects.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica , Receptor IGF Tipo 1/deficiencia , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Plasticidad de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/farmacología , Interferón gamma/farmacología , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Noqueados , Fenotipo , Receptor IGF Tipo 1/genética , Rotura Espontánea
15.
Cardiovasc Res ; 111(3): 184-93, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27013635

RESUMEN

AIMS: Pulmonary hypertension is a progressive disease with poor prognosis, characterized by pathological inward remodelling and loss of patency of the lung vasculature. The right ventricle is co-affected by pulmonary hypertension, which triggers events such as hypoxia and/or increased mechanical load. Initially the right ventricle responds with 'adaptive' hypertrophy, which is often rapidly followed by 'maladaptive' changes leading to right heart decompensation and failure, which is the ultimate cause of death. METHODS AND RESULTS: We report here that miR-223 is expressed in the murine lung and right ventricle at higher levels than in the left ventricle. Moreover, lung and right-ventricular miR-223 levels were markedly down-regulated by hypoxia. Correspondingly, increasing right-ventricular load by pulmonary artery banding, induced right-ventricular ischaemia, and the down-regulation of miR-223. Lung and right ventricle miR-223 down-regulation were linked with increased expression of the miR-223 target; insulin-like growth factor-I receptor (IGF-IR) and IGF-I downstream signalling. Similarly, miR-223 was decreased and IGF-IR increased in human pulmonary hypertension. Notably in young mice, miR-223 overexpression, the genetic inactivation or pharmacological inhibition of IGF-IR, all attenuated right-ventricular hypertrophy and improved right heart function under conditions of hypoxia or increased afterload. CONCLUSION: These findings highlight the early role of pulmonary and right-ventricular miR-223 and the IGF-IR in the right heart failure programme initiated by pulmonary hypoxia and increased mechanical load and may lead to the development of novel therapeutic strategies that target the development of PH and right heart failure.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Pulmón/metabolismo , MicroARNs/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Somatomedina/metabolismo , Disfunción Ventricular Derecha/metabolismo , Función Ventricular Derecha , Animales , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Ventrículos Cardíacos/fisiopatología , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Hipertensión Pulmonar/prevención & control , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/complicaciones , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Imidazoles/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Fenotipo , Piridinas/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Transducción de Señal , Disfunción Ventricular Derecha/genética , Disfunción Ventricular Derecha/fisiopatología , Disfunción Ventricular Derecha/prevención & control
16.
Cell Rep ; 14(7): 1571-1580, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26876170

RESUMEN

The hallmarks of age-related immune senescence are chronic inflammation, aberrant expansion of effector memory, and loss of naive T lymphocytes due in part to systemic activation of innate immune sensor NLRP3 inflammasome in myeloid lineage cells. The endogenous mechanisms that regulate inflammasome activation during aging are unknown. Here, we present evidence that growth hormone receptor (GH-R)-dependent downregulation of NLRP3 inflammasome in macrophages is linked to pro-longevity effects that maintain immune system homeostasis in aging. Deletion of GH-R prevented the macrophage-driven age-related activation of inflammasome in response to NLRP3 ligands and also increased the preservation of naive T cells, even in advanced age and with higher IFNγ secretion from effector cells. The mechanism of inflammasome inhibition is linked to autocrine somatotropic axis as ablation of IGF1R in macrophages lowered the NLRP3 inflammasome activation. Together, our findings show that functional somatotropic axis in macrophages controls inflammation, thus linking NLRP3-mediated innate immune signaling to health span and longevity.


Asunto(s)
Envejecimiento/genética , Proteínas Portadoras/genética , Inflamasomas/genética , Macrófagos/inmunología , Receptor IGF Tipo 1/genética , Receptores de Somatotropina/genética , Envejecimiento/inmunología , Animales , Comunicación Autocrina , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Proteínas Portadoras/inmunología , Regulación de la Expresión Génica , Homeostasis/inmunología , Inmunidad Innata , Memoria Inmunológica , Inflamasomas/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Longevidad/genética , Longevidad/inmunología , Macrófagos/citología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/inmunología , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/inmunología , Transducción de Señal , Bazo/citología , Bazo/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
17.
Neuron ; 89(3): 583-97, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26804996

RESUMEN

The insulin-like growth factor-1 receptor (IGF-1R) signaling is a key regulator of lifespan, growth, and development. While reduced IGF-1R signaling delays aging and Alzheimer's disease progression, whether and how it regulates information processing at central synapses remains elusive. Here, we show that presynaptic IGF-1Rs are basally active, regulating synaptic vesicle release and short-term plasticity in excitatory hippocampal neurons. Acute IGF-1R blockade or transient knockdown suppresses spike-evoked synaptic transmission and presynaptic cytosolic Ca(2+) transients, while promoting spontaneous transmission and resting Ca(2+) level. This dual effect on transmitter release is mediated by mitochondria that attenuate Ca(2+) buffering in the absence of spikes and decrease ATP production during spiking activity. We conclude that the mitochondria, activated by IGF-1R signaling, constitute a critical regulator of information processing in hippocampal neurons by maintaining evoked-to-spontaneous transmission ratio, while constraining synaptic facilitation at high frequencies. Excessive IGF-1R tone may contribute to hippocampal hyperactivity associated with Alzheimer's disease.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/citología , Mitocondrias/metabolismo , Neuronas/metabolismo , Receptor IGF Tipo 1/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Hipocampo/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Ratones , Potenciales Postsinápticos Miniatura/fisiología , Plasticidad Neuronal/fisiología , Terminales Presinápticos/metabolismo , Cultivo Primario de Células , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/deficiencia , Transducción de Señal/fisiología , Vesículas Sinápticas/metabolismo
19.
Aging Cell ; 14(5): 847-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26219530

RESUMEN

Downregulation of insulin-like growth factor (IGF) pathways prolongs lifespan in various species, including mammals. Still, the cellular mechanisms by which IGF signaling controls the aging trajectory of individual organs are largely unknown. Here, we asked whether suppression of IGF-I receptor (IGF-1R) in adult stem cells preserves long-term cell replacement, and whether this may prevent age-related functional decline in a regenerating tissue. Using neurogenesis as a paradigm, we showed that conditional knockout of IGF-1R specifically in adult neural stem cells (NSC) maintained youthful characteristics of olfactory bulb neurogenesis within an aging brain. We found that blocking IGF-I signaling in neural precursors increased cumulative neuroblast production and enhanced neuronal integration into the olfactory bulb. This in turn resulted in neuro-anatomical changes that improved olfactory function. Interestingly, mutants also displayed long-term alterations in energy metabolism, possibly related to IGF-1R deletion in NSCs throughout lifespan. We explored Akt and ERK signaling cascades and revealed differential regulation downstream of IGF-1R, with Akt phosphorylation preferentially decreased in IGF-1R(-/-) NSCs within the niche, and ERK pathway downregulated in differentiated neurons of the OB. These challenging experimental results were sustained by data from mathematical modeling, predicting that diminished stimulation of growth is indeed optimal for tissue aging. Thus, inhibiting growth and longevity gene IGF-1R in adult NSCs induced a gain-of-function phenotype during aging, marked by optimized management of cell renewal, and enhanced olfactory sensory function.


Asunto(s)
Envejecimiento/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Bulbo Olfatorio/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Animales , Ratones , Ratones Noqueados , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética
20.
Nucl Med Commun ; 36(6): 636-45, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25759946

RESUMEN

PURPOSE: Serum and intracellular instability limits the therapeutic applications of short interfering RNA (siRNA) as a radiopharmaceutical. Chemical modifications like phosphorothioate (PS) substitution and 2'-O-methoxy (2'-O-Me) modifications could eliminate such limitations. In this study, the effects of PS and 2'-O-Me modifications at the backbone of siRNA on serum stability and RNA interference activity were investigated. MATERIALS AND METHODS: Fully PS and 2'-O-Me-modified type 1 insulin-like growth factor receptor (IGF-1R) siRNA was radiolabeled with lutetium-177 ((177)Lu) through p-SCN-Bn-DTPA as a chelator. After purification with Vivaspin and PD-10 columns, the radiolabs were examined for stability in serum by instant thin-layer chromatography and polyacrylamide gel electrophoresis. The level of IGF-1R in response to the modified and labeled IGF-1R siRNA was examined using RT-PCR and ELISA assay in colon cancer cells. The effects of such siRNA on the prevention of proliferation of colon cancer cells and its apoptosis were investigated using MTT assay and Annexin-V/propidium iodide double staining, respectively. Cellular accumulation quantities of the labeled and modified IGF-1R siRNA were determined using a γ-counter by taking advantage of (177)Lu as a γ-emitter. RESULTS: Both the modified (177)Lu-siRNA complex and the modified nonlabeled siRNA showed significant stability in serum. The levels of IGF-1R mRNA and protein significantly decreased with both the labeled and nonlabeled IGF-1R siRNAs, but no such reduction in IGF-1R was observed with luciferase siRNA (P<0.01). Proliferation decreased significantly and apoptosis increased in the cells treated with modified (177)Lu-IGF-1R siRNA in comparison with either (177)Lu or labeled luciferase siRNA (P<0.001). CONCLUSION: Uniform chemically modified siRNAs can form stable complexes with Lu that pronounce its cytotoxic effect through apoptosis in colon cancer cells.


Asunto(s)
Lutecio/química , Interferencia de ARN , Estabilidad del ARN , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Radioisótopos/química , Apoptosis/genética , Transporte Biológico , Línea Celular Tumoral , Proliferación Celular/genética , Quelantes/química , Técnicas de Química Sintética , Técnicas de Silenciamiento del Gen , Humanos , Cinética , Fosfatos/química , ARN Interferente Pequeño/metabolismo , Tolerancia a Radiación , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...