Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 309
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Horm Res Paediatr ; 95(6): 619-630, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36446332

RESUMO

The growth hormone (GH)-insulin-like growth factor (IGF) cascade is central to the regulation of growth and metabolism. This article focuses on the history of the components of the IGF system, with an emphasis on the peptide hormones, IGF-I and -II, their cell surface receptors, and the IGF binding proteins (IGFBPs) and IGFBP proteases that regulate the availability of the peptide hormones for interaction with their receptors in relevant target tissues. We describe landmark events in the evolution of the somatomedin hypothesis, including evidence that has become available from experiments at the molecular and cellular levels, whole animal and tissue-specific gene knockouts, studies of cancer epidemiology, identification of prismatic human cases, and short- and long-term clinical trials of IGF-I therapy in humans. In addition, this new evidence has expanded our clinical definition of GH insensitivity (GHI) beyond growth hormone receptor mutations (classic Laron syndrome) to include conditions that cause primary IGF deficiency by impacting post-receptor signal transduction, IGF production, IGF availability to interact with the IGF-I receptor (IGF-1R), and defects in the IGF-1R, itself. We also discuss the clinical aspects of IGFs, from their description as insulin-like activity, to the use of IGF-I in the diagnosis and treatment of GH deficiency, and to the use of recombinant human IGF-I for therapy of children with GHI.


Assuntos
Fator de Crescimento Insulin-Like II , Fator de Crescimento Insulin-Like I , Síndrome de Laron , Animais , Humanos , Fator de Crescimento Insulin-Like I/deficiência , Fator de Crescimento Insulin-Like I/história , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like I/uso terapêutico , Síndrome de Laron/tratamento farmacológico , Síndrome de Laron/genética , Síndrome de Laron/história , Síndrome de Laron/fisiopatologia , Hormônios Peptídicos , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Somatomedinas/deficiência , Somatomedinas/história , Somatomedinas/fisiologia , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/história , Fator de Crescimento Insulin-Like II/fisiologia , Fator de Crescimento Insulin-Like II/uso terapêutico
2.
Dev Cell ; 57(1): 63-79.e8, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34963058

RESUMO

In all eutherian mammals, growth of the fetus is dependent upon a functional placenta, but whether and how the latter adapts to putative fetal signals is currently unknown. Here, we demonstrate, through fetal, endothelial, hematopoietic, and trophoblast-specific genetic manipulations in the mouse, that endothelial and fetus-derived IGF2 is required for the continuous expansion of the feto-placental microvasculature in late pregnancy. The angiocrine effects of IGF2 on placental microvasculature expansion are mediated, in part, through IGF2R and angiopoietin-Tie2/TEK signaling. Additionally, IGF2 exerts IGF2R-ERK1/2-dependent pro-proliferative and angiogenic effects on primary feto-placental endothelial cells ex vivo. Endothelial and fetus-derived IGF2 also plays an important role in trophoblast morphogenesis, acting through Gcm1 and Synb. Thus, our study reveals a direct role for the imprinted Igf2-Igf2r axis on matching placental development to fetal growth and establishes the principle that hormone-like signals from the fetus play important roles in controlling placental microvasculature and trophoblast morphogenesis.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Placenta/irrigação sanguínea , Receptor IGF Tipo 2/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Células Endoteliais/metabolismo , Feminino , Desenvolvimento Fetal , Feto/metabolismo , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/metabolismo , Neovascularização Fisiológica/fisiologia , Placenta/metabolismo , Placenta/fisiologia , Placentação , Gravidez , Receptor IGF Tipo 2/fisiologia , Fatores de Transcrição/genética , Trofoblastos/metabolismo
3.
Endocr Relat Cancer ; 27(3): 175-186, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31951591

RESUMO

By the strictest of definitions, a genetic driver of tumorigenesis should fulfill two criteria: it should be altered in a high percentage of patient tumors, and it should also be able to cause the same type of tumor to form in mice. No gene that fits either of these criteria has ever been found for ileal neuroendocrine tumors (I-NETs), which in humans are known for an unusual lack of recurrently mutated genes, and which have never been detected in mice. In the following report, we show that I-NETs can be generated by transgenic RT2 mice, which is a classic model for a genetically unrelated disease, pancreatic neuroendocrine tumors (PNETs). The ability of RT2 mice to generate I-NETs depended upon genetic background. I-NETs appeared in a B6AF1 genetic background, but not in a B6 background nor even in an AB6F1 background. AB6F1 and B6AF1 have identical nuclear DNA but can potentially express different allelic forms of imprinted genes. This led us to test human I-NETs for loss of imprinting, and we discovered that the IGF2 gene showed loss of imprinting and increased expression in the I-NETs of 57% of patients. By increasing IGF2 activity genetically, I-NETs could be produced by RT2 mice in a B6 genetic background, which otherwise never developed I-NETs. The facts that IGF2 is altered in a high percentage of patients with I-NETs and that I-NETs can form in mice that have elevated IGF2 activity, define IGF2 as the first genetic driver of ileal neuroendocrine tumorigenesis.


Assuntos
Neoplasias do Íleo/etiologia , Fator de Crescimento Insulin-Like II/fisiologia , Tumores Neuroendócrinos/etiologia , Animais , Modelos Animais de Doenças , Feminino , Impressão Genômica , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos
4.
Cells ; 8(10)2019 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-31590432

RESUMO

When originally discovered, one of the initial observations was that, when all of the insulin peptide was depleted from serum, the vast majority of the insulin activity remained and this was due to a single additional peptide, IGF-II. The IGF-II gene is adjacent to the insulin gene, which is a result of gene duplication, but has evolved to be considerably more complicated. It was one of the first genes recognised to be imprinted and expressed in a parent-of-origin specific manner. The gene codes for IGF-II mRNA, but, in addition, also codes for antisense RNA, long non-coding RNA, and several micro RNA. Recent evidence suggests that each of these have important independent roles in metabolic regulation. It has also become clear that an alternatively spliced form of the insulin receptor may be the principle IGF-II receptor. These recent discoveries have important implications for metabolic disorders and also for cancer, for which there is renewed acknowledgement of the importance of metabolic reprogramming.


Assuntos
Diabetes Mellitus/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Neoplasias/metabolismo , Obesidade/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like II/genética , Camundongos , Ratos , Receptor IGF Tipo 2/fisiologia
5.
Med Sci Monit ; 25: 4655-4664, 2019 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-31230061

RESUMO

BACKGROUND The aim of this study was to explore the effect of dexmedetomidine (DEX)-mediated insulin-like growth factor 2 (IGF2) signal pathway on immune function and cancer cell invasion and migration in rats with ovarian cancer. MATERIAL AND METHODS Forty rats with ovarian cancer were divided into 4 groups: model group, and low dose (0.2 µg/kg/hour DEX), medium dose (1.0 µg/kg/hour DEX), and high dose (5.0 µg/kg/hour DEX) DEX groups. In addition, 10 Fischer344 rats were selected as a normal group. Human NUTU-19 poorly differentiated epithelial ovarian cancer cell line cells were divided into 4 groups: a blank group and low dose, medium dose, and high dose DEX NUTU-19 groups. RESULTS Compared with the normal group, in the other groups the serum interleukin (IL)-2 and interferon gamma (INF-γ) levels, CD4⁺ and CD8⁺ percentages, CD4⁺/CD8⁺ ratio, and transformation rate of splenic lymphocytes were decreased, and the serum tumor necrosis factor alpha (TNF-alpha) level, IGF2, insulin-like growth factor 1 receptor (IGF1R), insulin receptor substrate 1 (IRS1) mRNA, and protein expressions in ovarian tissue were increased (all P<0.05). Results in the DEX groups compared with model group were the opposite of those in the other groups compared with normal group (all P<0.05). Compared with the blank group, in the other groups the proliferation, invasion, and migration of ovarian cancer cells were reduced significantly (all P<0.05). Compared with the low dose DEX NUTU-19 group, in the high dose DEX NUTU-19 group the invasion and migration of ovarian cancer cells weakened significantly (both P<0.05). CONCLUSIONS A certain dose of DEX can effectively inhibit IGF2 signal pathway activation to improve the immune function of rats with ovarian cancer, inhibiting the invasion and migration of ovarian cancer cells.


Assuntos
Dexmedetomidina/farmacologia , Fator de Crescimento Insulin-Like II/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Movimento Celular/fisiologia , Feminino , Fator de Crescimento Insulin-Like II/fisiologia , Interleucina-2/metabolismo , NF-kappa B/metabolismo , Invasividade Neoplásica/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Ratos , Ratos Endogâmicos F344 , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
6.
Cell Metab ; 29(6): 1363-1375.e8, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-30745181

RESUMO

Recent investigations revealed that macrophages could be trained with an altered responsiveness, raising the possibility of combating autoimmune diseases by imparting anti-inflammatory capabilities to these cells. While investigating the effect of mesenchymal stem cells on experimental autoimmune encephalomyelitis (EAE), we found a critical role of insulin-like growth factor 2 (IGF-2) in training macrophages to become anti-inflammatory during their maturation. IGF-2 exerts its effects by preprogramming maturing macrophages to commit oxidative phosphorylation (OXPHOS). IGF-2-preprogrammed macrophages maintained the mitochondrial complex V activities even upon pro-inflammation stimulation, thus enabling an elevated programmed death-ligand 1 (PD-L1) expression. PD-L1 neutralization abolished the beneficial effect of IGF-2 on EAE. Furthermore, adoptive transfer of IGF-2-preprogrammed macrophages to EAE mice increased Tregs and alleviated the diseases. Our results demonstrate that shaping macrophage responsiveness by IGF-2 is effective in managing inflammatory diseases, and the OXPHOS commitment can be preset to determine the anti-inflammatory fate of macrophages.


Assuntos
Anti-Inflamatórios/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Macrófagos/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Transferência Adotiva , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/terapia , Feminino , Humanos , Fator de Crescimento Insulin-Like II/antagonistas & inibidores , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/farmacologia , Células THP-1
7.
Oncol Res ; 27(2): 269-279, 2019 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-29562959

RESUMO

MicroRNAs are essential regulators of cancer-associated genes at the posttranscriptional level, and their expression is altered in cancer tissues. Herein we sought to identify the regulation of miR-615-3p in NSCLC progression and its mechanism. miR-615-3p expression was significantly downregulated in NSCLC tissue compared to control normal tissue. Exogenous overexpression of miR-615-3p inhibited the growth and metastasis of NSCLC cells. In addition, the in vivo mouse xenograft model showed that overexpression of miR-615-3p inhibited NSCLC growth and lung metastasis, whereas decreased expression of miR-615-3p caused an opposite outcome. Furthermore, we revealed that insulin-like growth factor 2 (IGF2) expression was negatively correlated with the miR-615-3p level in NSCLC specimens, and IGF2 knockdown mimicked the effect of miR-615-3p inhibition on NSCLC cell proliferation, migration, and invasion. In addition, overexpression of IGF2 rescued the inhibition of miR-615-3p in NSCLC cells. Together, our results indicated that miR-615-3p played important roles in the regulation of NSCLC growth and metastasis by targeting IGF2.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Fator de Crescimento Insulin-Like II/antagonistas & inibidores , Neoplasias Pulmonares/patologia , MicroRNAs/fisiologia , Regiões 3' não Traduzidas , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Movimento Celular , Humanos , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Neoplasias Pulmonares/genética , Camundongos , Invasividade Neoplásica , Metástase Neoplásica
8.
Yi Chuan ; 39(12): 1150-1157, 2017 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-29258985

RESUMO

As one of the first identified long non-coding RNAs (lncRNAs), H19 plays a wide range of roles in vivo, including not only as a tumor suppressor and oncogene involved in disease process, but also as a regulator of growth and development of multiple tissues in mammalian embryos. The function of H19 in muscles (both skeletal and cardiac muscle) draws widespread attention due to the following two reasons. On one hand, H19 promotes myogenic differentiation and myogenesis of skeletal muscle satellite cells (SMSCs) via regulating Igf2 in cis. On the other hand, H19 also modulates the target genes in trans, including sponging let-7, miR-106 or miR-29 to mediate myocyte glucose uptake, cardiomyocyte proliferation and tendon repair, as well as promote embryonic development and muscle regeneration through binding to MBD1 as a chromatin modifier. In this review, we summarize the role of H19 in mammalian muscles, which will provide a reference for further research to unveil the molecular mechanism of muscle growth and development.


Assuntos
Desenvolvimento Muscular , Músculos/fisiologia , RNA Longo não Codificante/fisiologia , Animais , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Glucose/metabolismo , Humanos , Fator de Crescimento Insulin-Like II/fisiologia , Regeneração , Fatores de Transcrição/metabolismo
9.
J Craniomaxillofac Surg ; 45(2): 198-202, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28089088

RESUMO

Synovial chondromatosis (SC) is a benign disease of the joints without a known cause. It sometimes affects the temporomandibular joint (TMJ) and is accompanied by pain, swelling, malocclusion, and crepitation. It has been divided into three stages by Milgram and is supposed to originate from the synovia and cartilage of a joint (Milgram, 1977b). The aim of this study was to examine an involvement of the insulin-like growth factors (IGF-I/-II) and their binding proteins (IGFBP-1 to -6) in the etiology of this disease. Therefore 23 specimen of SC from 16 patients were immunohistochemically stained and microscopically examined. Staining was assessed semiquantitatively: negative (-), weakly positive ((+)), moderately positive (+), strongly positive (++) and very strongly positive (+++). It could be seen that especially the chondro- and fibrocytes and the synovia showed positive staining for almost all IGFs and IGFBPs. The underlying tissue, consisting of connective tissue or chondroid matrix, was stained as well but more weakly so. We conclude that the IGF/IGFBP system seems to contribute to the pathogenesis of SC, especially IGF-I and -II, and their effects enhancing binding protein 5.


Assuntos
Condromatose Sinovial/etiologia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Somatomedinas/metabolismo , Transtornos da Articulação Temporomandibular/etiologia , Adulto , Idoso , Condrócitos/metabolismo , Condrócitos/patologia , Condromatose Sinovial/patologia , Corantes , Feminino , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Masculino , Pessoa de Meia-Idade , Somatomedinas/fisiologia , Líquido Sinovial/metabolismo , Transtornos da Articulação Temporomandibular/patologia
10.
Tumour Biol ; 37(9): 12485-12495, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27337954

RESUMO

Increased expression of insulin-like growth factor 2 (IGF2) is found in tumors of colorectal cancer (CRC) patients exhibiting a gained region on chromosome 11q15 and is implicated in poor patient survival. This study analyzes in vitro phenotypic- and gene expression changes associated with IGF2 shRNA-mediated knockdown. Initially, doxycycline inducible IGF2 knockdown cell lines were generated in the CRC cell lines SW480 and LS174T. The cells were analyzed for changes in proliferation, cell cycle, apoptosis, adhesion, and invasion. Expression profiling analysis was performed, and, for a subset of the identified genes, expression was validated by qRT-PCR and Western blot. IGF2 knockdown inhibited cell proliferation in both cell lines induced G1 cell cycle blockade and decreased adhesion to several extracellular matrix proteins. Knockdown of IGF2 did not alter invasiveness in SW480 cells, while a slight increase in apoptosis was seen only in the LS174T cell line. Knockdown of IGF2 in SW480 deregulated 58 genes, several of which were associated with proliferation and cell-cell/cell-ECM contacts. A subset of these genes, including CDK2, YAP1, and BIRC5 (Survivin), are members of a common network. This study supports the concept of direct autocrine/paracrine tumor cell activation through IGF2 and a shows role of IGF2 in CRC proliferation, adhesion and, to a limited extent, apoptosis.


Assuntos
Neoplasias Colorretais/patologia , Fator de Crescimento Insulin-Like II/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Antígenos CD , Caderinas/genética , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Quinase 2 Dependente de Ciclina/fisiologia , Desmoplaquinas/genética , Humanos , Fator de Crescimento Insulin-Like II/genética , Fosfoproteínas/genética , RNA Interferente Pequeno/genética , Fatores de Transcrição , Proteínas de Sinalização YAP , gama Catenina
11.
In Vitro Cell Dev Biol Anim ; 52(5): 607-15, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26956358

RESUMO

Insulin-like growth factors (IGFs) are involved in growth and tissue development, including diseases such as type-2 diabetes and cancers. However, their roles in lineage specification, especially in early mammalian neural development, are poorly understood. Here, we analyzed the protein expression of IGF-2 in early mouse embryo, and it was preferentially detected in anterior mesodermal tissue, adjacent to the neural plate. We utilized a self-organizing neural tissue culture system and analyzed the direct effect of IGF-2 on the general neural marker Sox1. Interestingly, using recombinant IGF-2 and a chemical inhibitor of its receptor (IGF-1R), we found that the IGF-2/IGF-1R pathway positively regulated Sox1 expression in embryonic stem (ES) cell-derived neural tissue. Furthermore, to visualize the expression patterns of other neural markers, we used reporter ES cell lines and we found that the IGF-2/IGF-1R signaling upregulated the expression of the posterior neural marker Irx3. In contrast, the anterior neural marker Six3 was downregulated by IGF-2/IGF-1R signaling. Together, our results demonstrate that IGF-2/IGF-1R signaling has different effects on neural marker expression, which may influence the early regional identity of ES cell-derived neural tissues.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Placa Neural/crescimento & desenvolvimento , Receptor IGF Tipo 1/fisiologia , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células-Tronco Embrionárias/metabolismo , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Placa Neural/metabolismo , Receptor IGF Tipo 1/metabolismo , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Fatores de Transcrição/genética , Proteína Homeobox SIX3
12.
Gynecol Oncol ; 141(1): 121-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26751131

RESUMO

OBJECTIVES: Aberrant expressions of LIN-28B, let-7a and IGF-II occur in epithelial ovarian cancer, and the LIN-28B/let-7a/IGF-II axis is associated with human disease. The purpose of this study was to investigate the associations between LIN-28B/let-7a/IGF-II axis molecular subtypes and epithelial ovarian cancer prognosis. METHODS: Using quantitative reverse transcription PCR, we analyzed LIN-28B, let-7a and IGF-II mRNA in 211 primary epithelial ovarian cancer tissues, and also performed Classification and Regression Tree (CART) and survival analyses. RESULTS: Four terminal subtypes were identified in the CART analysis in combination with survival analysis. Kaplan-Meier survival curves showed that subtypes LIN-28B(low)let-7a(low) and LIN-28B(low)let-7a(high)IGF-II(low) had significantly better survival than subtypes LIN-28B(high) or LIN-28B(low)let-7a(high)IGF-II(high) (p<0.0001 for overall, p=0.017 for progression-free survival, respectively). Multivariate Cox regression models showed that compared to subtype LIN-28B(high), subtypes LIN-28B(low)let-7a(low) and LIN-28B(low)let-7a(high)IGF-II(low) had significantly reduced mortality and reduced relapse risks. Moreover, subtype LIN-28B(low)let-7a(low) had better response to chemotherapy than subtype LIN-28B(high). CONCLUSIONS: These results suggest that molecular subtypes of the LIN-28B/let-7a/IGF-II axis associate with heterogeneous progression and may have clinical implications in predicting epithelial ovarian cancer prognosis.


Assuntos
Fator de Crescimento Insulin-Like II/fisiologia , MicroRNAs/fisiologia , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Ovarianas/mortalidade , Proteínas de Ligação a RNA/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Epitelial do Ovário , Feminino , Humanos , Fator de Crescimento Insulin-Like II/genética , MicroRNAs/genética , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Prognóstico , Modelos de Riscos Proporcionais , Proteínas de Ligação a RNA/genética
13.
Cardiovasc Res ; 106(1): 131-42, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25678587

RESUMO

AIMS: Activation of an osteogenic transcriptional program contributes to the initiation of aortic calcification in atherosclerosis. The role of microRNAs in regulating aortic calcification is understudied. We tested the hypothesis that miR-30e regulates an osteogenic program in bone marrow-derived mesenchymal stem cells (MSCs), aortic smooth muscle cells (SMCs), and ApoE(-/-) mice. METHODS AND RESULTS: In aortas of wild-type mice, we found that miR-30e is highly expressed in medial SMCs. In aortas of old ApoE(-/-) mice, we found that miR-30e transcripts are down-regulated in an inverse relation to the osteogenic markers Runx2, Opn, and Igf2. In vitro, miR-30e over-expression reduced the proliferation of MSCs and SMCs while increasing adipogenic differentiation of MSCs and smooth muscle differentiation of SMCs. In MSCs and SMCs over-expressing miR-30e, microarrays and qPCR showed repression of an osteogenic gene panel including Igf2. Inhibiting miR-30e in MSCs increased Igf2 transcripts. In SMCs, IGF2 recombinant protein rescued miR-30e-repressed osteogenic differentiation. Luciferase and mutagenesis assays showed binding of miR-30e to a novel and essential site at the 3'UTR of Igf2. In ApoE(-/-) mice, injections of antimiR-30e oligos increased Igf2 expression in the aortas and livers and significantly enhanced OPN protein expression and calcium deposition in aortic valves. CONCLUSION: miR-30e represses the osteogenic program in MSCs and SMCs by targeting IGF2 and drives their differentiation into adipogenic or smooth muscle lineage, respectively. Our data suggest that down-regulation of miR-30e in aortas with age and atherosclerosis triggers vascular calcification. The miR-30e pathway plays an important regulatory role in vascular diseases.


Assuntos
Apolipoproteínas E/deficiência , Fator de Crescimento Insulin-Like II/fisiologia , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/fisiologia , Músculo Liso Vascular/fisiologia , Osteogênese/fisiologia , Envelhecimento/fisiologia , Animais , Aorta/citologia , Apolipoproteínas E/genética , Apolipoproteínas E/fisiologia , Células da Medula Óssea/citologia , Calcinose/fisiopatologia , Diferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/citologia
14.
J Mol Endocrinol ; 54(1): R1-13, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25351818

RESUMO

Endothelial cells line blood vessels and modulate vascular tone, thrombosis, inflammatory responses and new vessel formation. They are implicated in many disease processes including atherosclerosis and cancer. IGFs play a significant role in the physiology of endothelial cells by promoting migration, tube formation and production of the vasodilator nitric oxide. These actions are mediated by the IGF1 and IGF2/mannose 6-phosphate receptors and are modulated by a family of high-affinity IGF binding proteins. IGFs also increase the number and function of endothelial progenitor cells, which may contribute to protection from atherosclerosis. IGFs promote angiogenesis, and dysregulation of the IGF system may contribute to this process in cancer and eye diseases including retinopathy of prematurity and diabetic retinopathy. In some situations, IGF deficiency appears to contribute to endothelial dysfunction, whereas IGF may be deleterious in others. These differences may be due to tissue-specific endothelial cell phenotypes or IGFs having distinct roles in different phases of vascular disease. Further studies are therefore required to delineate the therapeutic potential of IGF system modulation in pathogenic processes.


Assuntos
Células Endoteliais/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Animais , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Comunicação Celular , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Doenças Retinianas/metabolismo , Doenças Retinianas/fisiopatologia
15.
Oncogene ; 34(35): 4558-69, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25435370

RESUMO

Interstitial fluid flow in and around the tumor tissue is a physiologically relevant mechanical signal that regulates intracellular signaling pathways throughout the tumor. Yet, the effects of interstitial flow and associated fluid shear stress on the tumor cell function have been largely overlooked. Using in vitro bioengineering models in conjunction with molecular cell biology tools, we found that fluid shear (2 dyn/cm(2)) markedly upregulates matrix metalloproteinase 12 (MMP-12) expression and its activity in human chondrosarcoma cells. MMP-12 expression is induced in human chondrocytes during malignant transformation. However, the signaling pathway regulating MMP-12 expression and its potential role in human chondrosarcoma cell invasion and metastasis have yet to be delineated. We discovered that fluid shear stress induces the synthesis of insulin growth factor-2 (IGF-2) and vascular endothelial growth factor (VEGF) B and D, which in turn transactivate MMP-12 via PI3-K, p38 and JNK signaling pathways. IGF-2-, VEGF-B- or VEGF-D-stimulated chondrosarcoma cells display markedly higher migratory and invasive potentials in vitro, which are blocked by inhibiting MMP-12, PI3-K, p38 or JNK activity. Moreover, recombinant human MMP-12 or MMP-12 overexpression can potentiate chondrosarcoma cell invasion in vitro and the lung colonization in vivo. By reconstructing and delineating the signaling pathway regulating MMP-12 activation, potential therapeutic strategies that interfere with chondrosarcoma cell invasion may be identified.


Assuntos
Neoplasias Ósseas/enzimologia , Condrossarcoma/enzimologia , Fator de Crescimento Insulin-Like II/fisiologia , Neoplasias Pulmonares/enzimologia , Metaloproteinase 12 da Matriz/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Animais , Fenômenos Biomecânicos , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular , Condrossarcoma/secundário , Ativação Enzimática , Indução Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/secundário , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Invasividade Neoplásica , Transplante de Neoplasias
16.
Reproduction ; 147(5): 703-17, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24481956

RESUMO

Although there is no acute luteolytic mechanism in the absence of pregnancy in the bitch, a precise and well-timed embryo-maternal interaction seems to be required for the initiation and maintenance of gestation. As only limited information is available about these processes in dogs, in this study, the uterine expression of possible decidualization markers was investigated during the pre-implantation stage (days 10-12) of pregnancy and in the corresponding nonpregnant controls. In addition, the expression of selected genes associated with blastocyst development and/or implantation was investigated in embryos flushed from the uteri of bitches used for this study (unhatched and hatched blastocysts). There was an upregulated expression of prolactin receptor (PRLR) and IGF2 observed pre-implantation. The expression of PRL and of IGF1 was unaffected, and neither was the expression of progesterone- or estrogen receptor ß (ESR2). In contrast, (ESR1) levels were elevated during early pregnancy. Prostaglandin (PG)-system revealed upregulated expression of PGE2-synthase and its receptors, PTGER2 and PTGER4, and of the PG-transporter. Elevated levels of AKR1C3 mRNA, but not the protein itself, were noted. Expression of prostaglandin-endoperoxide synthase 2 (PTGS2) remained unaffected. Most of the transcripts were predominantly localized to the uterine epithelial cells, myometrium and, to a lesser extent, to the uterine stroma. PGES (PTGES) mRNA was abundantly expressed in both groups of embryos and appeared higher in the hatched ones. The expression level of IGF2 mRNA appeared higher than that of IGF1 mRNA in hatched embryos. In unhatched embryos IGF1, IGF2, and PTGS2 mRNA levels were below the detection limit.


Assuntos
Cães/fisiologia , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Prenhez/fisiologia , Útero/fisiologia , Animais , Cães/genética , Desenvolvimento Embrionário/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Prenhez/genética , RNA Mensageiro/genética , RNA Mensageiro/fisiologia , Receptores da Prolactina/genética , Receptores da Prolactina/fisiologia , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/fisiologia
18.
Stem Cells ; 31(10): 2095-103, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23836547

RESUMO

Human embryonic stem cells (hESCs) can be progressively differentiated into definitive endoderm (DE), hepatic progenitors, and hepatocytes, and thus provide an excellent model system for the mechanistic study of hepatocyte differentiation, which is currently poorly understood. Here, we found that insulin enhanced hepatocyte differentiation from hESC-derived DE. Insulin activated the PI3K/AKT pathway, but not the mitogen-activated protein kinase pathway in the DE cells, and inhibition of the PI3K/AKT pathways by inhibitors markedly inhibited hepatocyte differentiation. In addition, insulin-like growth factor 1 (IGF1) and IGF2 also activated the PI3K/AKT pathway in DE cells and their expression was robustly upregulated during hepatocyte differentiation from DE. Furthermore, inhibition of IGF receptor 1 (IGF1R) by a small molecule inhibitor PPP or knockdown of the IGF1R by shRNA attenuated hepatocyte differentiation. Moreover, simultaneous knockdown of the IGF1R and the insulin receptor with shRNAs markedly reduced the activation of AKT and substantially impaired hepatocyte differentiation. The PI3K pathway specifically enhanced the expression of HNF1 and HNF4 to regulate hepatocyte differentiation from DE. Although inhibition of the PI3K pathway was previously shown to be required for the induction of DE from hESCs, our study revealed a positive role of the PI3K pathway in hepatocyte differentiation after the DE stage, and has advanced our understanding of hepatocyte cell fate determination.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Hepatócitos/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Insulina/fisiologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Linhagem Celular , Sobrevivência Celular , Humanos , Fator de Crescimento Insulin-Like II/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/fisiologia , Receptor de Insulina/fisiologia , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
19.
Endocr Rev ; 34(6): 798-826, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23671155

RESUMO

Tumors of mesenchymal and epithelial origin produce IGF-2, which activates pathways in the tumors. In a minority of patients, the tumors (hepatomas, fibromas, and fibrosarcomas are the most common among many) release into the circulation enough IGF-2-related peptides to mimic the fasting hypoglycemia characteristic of patients with insulin-producing islet-cell tumors. Rarely, markedly elevated IGF-2 levels produce somatic changes suggestive of acromegaly. Typically, the elevated IGF-2 levels are associated with suppressed plasma levels of insulin, IGF-1, and GH. Complicating the pathophysiology are the IGF binding proteins (IGFBPs) that can bind IGF-2 and IGF-1, modifying hormone metabolism and action. IGFBP concentrations are often altered in the presence of these tumors. At the cellular level, the 3 hormone-related ligands, IGF-2, IGF-1, and insulin, all bind to 4 (or more) types of IGF-1 receptor (IGF-1R) and insulin receptor (IR). Each receptor has its own characteristic affinity for each ligand, a tyrosine kinase, and overlapping profiles of action in the target cells. The IGF-2R, in addition to binding mannose-6-phosphate-containing proteins, provides an IGF-2 degradation pathway. Recent evidence suggests IGF-2R involvement also in signal transduction. Surgery, the treatment of choice, can produce a cure. For patients not cured by surgery, multiple therapies exist, for the tumor and for hypoglycemia. Potential future therapeutic approaches are sketched. From 1910 to 1930, hypoglycemia, insulin, insulinomas, and non-islet-cell tumors were recognized. The latter third of the century witnessed the emergence of the immunoassay for insulin; the IGFs, their binding proteins, and assays to measure them; and receptors for the insulin-related peptides as well as the intracellular pathways beyond the receptor. In closing, we replace non-islet-cell tumor hypoglycemia, an outdated and misleading label, with IGF-2-oma, self-explanatory and consistent with names of other hormone-secreting tumors.


Assuntos
Hipoglicemia/etiologia , Fator de Crescimento Insulin-Like II/efeitos adversos , Fator de Crescimento Insulin-Like II/metabolismo , Neoplasias/complicações , Neoplasias/metabolismo , Acromegalia/diagnóstico , Acromegalia/metabolismo , Animais , Autoimunidade , Humanos , Hipoglicemia/diagnóstico , Hipoglicemia/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Neoplasias/epidemiologia , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo
20.
Cancer Res ; 73(13): 3974-86, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23633480

RESUMO

Insulin receptor (IR) exists as two isoforms resulting from the alternative splicing of IR pre-mRNA. IR-B promotes the metabolic effects of insulin, whereas IR-A rather signals proliferative effects. IR-B is predominantly expressed in the adult liver. Here, we show that the alternative splicing of IR pre-mRNA is dysregulated in a panel of 85 human hepatocellular carcinoma (HCC) while being normal in adjacent nontumor liver tissue. An IR-B to IR-A switch is frequently observed in HCC tumors regardless of tumor etiology. Using pharmacologic and siRNA approaches, we show that the autocrine or paracrine activation of the EGF receptor (EGFR)/mitogen-activated protein/extracellular signal-regulated kinase pathway increases the IR-A:IR-B ratio in HCC cell lines, but not in normal hepatocytes, by upregulating the expression of the splicing factors CUGBP1, hnRNPH, hnRNPA1, hnRNPA2B1, and SF2/ASF. In HCC tumors, there is a significant correlation between the expression of IR-A and that of splicing factors. Dysregulation of IR pre-mRNA splicing was confirmed in a chemically induced model of HCC in rat but not in regenerating livers after partial hepatectomy. This study identifies a mechanism responsible for the generation of mitogenic IR-A and provides a novel interplay between IR and EGFR pathways in HCC. Increased expression of IR-A during neoplastic transformation of hepatocytes could mediate some of the adverse effects of hyperinsulinemia on HCC.


Assuntos
Antígenos CD/genética , Carcinoma Hepatocelular/metabolismo , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas Experimentais/metabolismo , Receptor de Insulina/genética , Animais , Antígenos CD/metabolismo , Proteínas CELF1 , Transformação Celular Neoplásica/metabolismo , Expressão Gênica , Células Hep G2 , Hepatócitos/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Insulina/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Regeneração Hepática , Sistema de Sinalização das MAP Quinases , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ratos , Receptor de Insulina/metabolismo , Fatores de Processamento de Serina-Arginina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA