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











Base de dados
Intervalo de ano de publicação
1.
Elife ; 132024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38441552

RESUMO

The mammary gland is a unique organ that undergoes dynamic alterations throughout a female's reproductive life, making it an ideal model for developmental, stem cell and cancer biology research. Mammary gland development begins in utero and proceeds via a quiescent bud stage before the initial outgrowth and subsequent branching morphogenesis. How mammary epithelial cells transit from quiescence to an actively proliferating and branching tissue during embryogenesis and, importantly, how the branch pattern is determined remain largely unknown. Here, we provide evidence indicating that epithelial cell proliferation and onset of branching are independent processes, yet partially coordinated by the Eda signaling pathway. Through heterotypic and heterochronic epithelial-mesenchymal recombination experiments between mouse mammary and salivary gland tissues and ex vivo live imaging, we demonstrate that unlike previously concluded, the mode of branching is an intrinsic property of the mammary epithelium whereas the pace of growth and the density of ductal tree are determined by the mesenchyme. Transcriptomic profiling and ex vivo and in vivo functional studies in mice disclose that mesenchymal Wnt/ß-catenin signaling, and in particular IGF-1 downstream of it critically regulate mammary gland growth. These results underscore the general need to carefully deconstruct the different developmental processes producing branched organs.


Assuntos
Células Epiteliais , Via de Sinalização Wnt , Camundongos , Animais , Epitélio/metabolismo , Células Epiteliais/fisiologia , Proliferação de Células , Morfogênese , Mesoderma , Glândulas Mamárias Animais/metabolismo
2.
Cell Rep ; 39(6): 110793, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35545054

RESUMO

Ribosomopathies constitute a range of disorders associated with defective protein synthesis mainly affecting hematopoietic stem cells (HSCs) and erythroid development. Here, we demonstrate that deletion of poly-pyrimidine-tract-binding protein 1 (PTBP1) in the hematopoietic compartment leads to the development of a ribosomopathy-like condition. Specifically, loss of PTBP1 is associated with decreases in HSC self-renewal, erythroid differentiation, and protein synthesis. Consistent with its function as a splicing regulator, PTBP1 deficiency results in splicing defects in hundreds of genes, and we demonstrate that the up-regulation of a specific isoform of CDC42 partly mimics the protein-synthesis defect associated with loss of PTBP1. Furthermore, PTBP1 deficiency is associated with a marked defect in ribosome biogenesis and a selective reduction in the translation of mRNAs encoding ribosomal proteins. Collectively, this work identifies PTBP1 as a key integrator of ribosomal functions and highlights the broad functional repertoire of RNA-binding proteins.


Assuntos
Células-Tronco Hematopoéticas , Ribossomos , Eritrócitos/metabolismo , Eritropoese , Células-Tronco Hematopoéticas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo
3.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30070361

RESUMO

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.


Assuntos
Receptores ErbB/metabolismo , Receptor IGF Tipo 1/deficiência , Receptor de Insulina/deficiência , Câncer Papilífero da Tireoide/metabolismo , Células Epiteliais da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Câncer Papilífero da Tireoide/patologia , Células Epiteliais da Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Tireotropina/biossíntese , Tireotropina/metabolismo
4.
Endocrinology ; 158(7): 2269-2283, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28881863

RESUMO

Growth hormone (GH) and insulinlike growth factor (IGF) promote aging and age-related pathologies. Inhibiting this pathway by targeting IGF receptor (IGF-1R) is a promising strategy to extend life span, alleviate age-related diseases, and reduce tumor growth. Although anti-IGF-1R agents are being developed, long-term effects of IGF-1R blockade remain unknown. In this study, we used ubiquitous inducible IGF-1R knockout (UBIKOR) to suppress signaling in all adult tissues and screened health extensively. Surprisingly, UBIKOR mice showed no overt defects and presented with rather inconspicuous health, including normal cognition. Endocrine GH and IGF-1 were strongly upregulated without causing acromegaly. UBIKOR mice were strikingly lean with coordinate changes in body composition and organ size. They were insulin resistant but preserved physiological energy expenditure and displayed enhanced fasting metabolic flexibility. Thus, long-term IGF-1R blockade generated beneficial effects on aging-relevant metabolism, but exposed to high GH. This needs to be considered when targeting IGF-1R to protect from neurodegeneration, retard aging, or fight cancer.


Assuntos
Metabolismo Energético/genética , Hormônio do Crescimento/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Receptor IGF Tipo 1/genética , Magreza/genética , Animais , Composição Corporal/efeitos dos fármacos , Composição Corporal/genética , Metabolismo Energético/efeitos dos fármacos , Feminino , Deleção de Genes , Hormônio do Crescimento Humano/análogos & derivados , Hormônio do Crescimento Humano/farmacologia , Resistência à Insulina/genética , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor IGF Tipo 1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Magreza/metabolismo
5.
Brain ; 140(7): 2012-2027, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28595357

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Região CA1 Hipocampal/metabolismo , Fármacos Neuroprotetores/metabolismo , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/genética , Transcriptoma , Envelhecimento/metabolismo , Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/administração & dosagem , Animais , Ansiedade/induzido quimicamente , Ansiedade/complicações , Ansiedade/prevenção & controle , Encefalite/induzido quimicamente , Encefalite/complicações , Encefalite/prevenção & controle , Feminino , Infusões Intraventriculares , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/complicações , Transtornos da Memória/prevenção & controle , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo
6.
PLoS One ; 10(3): e0119392, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793522

RESUMO

BACKGROUND: In the testis, thyroid hormone (T3) regulates the number of gametes produced through its action on Sertoli cell proliferation. However, the role of T3 in the regulation of steroidogenesis is still controversial. METHODS: The TRαAMI knock-in allele allows the generation of transgenic mice expressing a dominant-negative TRα1 (thyroid receptor α1) isoform restricted to specific target cells after Cre-loxP recombination. Here, we introduced this mutant allele in both Sertoli and Leydig cells using a novel aromatase-iCre (ARO-iCre) line that expresses Cre recombinase under control of the human Cyp19(IIa)/aromatase promoter. FINDINGS: We showed that loxP recombination induced by this ARO-iCre is restricted to male and female gonads, and is effective in Sertoli and Leydig cells, but not in germ cells. We compared this model with the previous introduction of TRαAMI specifically in Sertoli cells in order to investigate T3 regulation of steroidogenesis. We demonstrated that TRαAMI-ARO males exhibited increased testis weight, increased sperm reserve in adulthood correlated to an increased proliferative index at P3 in vivo, and a loss of T3-response in vitro. Nevertheless, TRαAMI-ARO males showed normal fertility. This phenotype is similar to TRαAMI-SC males. Importantly, plasma testosterone and luteinizing hormone levels, as well as mRNA levels of steroidogenesis enzymes StAR, Cyp11a1 and Cyp17a1 were not affected in TRαAMI-ARO. CONCLUSIONS/SIGNIFICANCE: We concluded that the presence of a mutant TRαAMI allele in both Leydig and Sertoli cells does not accentuate the phenotype in comparison with its presence in Sertoli cells only. This suggests that direct T3 regulation of steroidogenesis through TRα1 is moderate in Leydig cells, and that Sertoli cells are the main target of T3 action in the testis.


Assuntos
Expressão Gênica , Células Intersticiais do Testículo/metabolismo , Células de Sertoli/metabolismo , Receptores alfa dos Hormônios Tireóideos/genética , Animais , Proliferação de Células , Fertilidade , Marcação de Genes , Humanos , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Testosterona/sangue
7.
J Hematol Oncol ; 8: 3, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25648584

RESUMO

BACKGROUND: The tyrosine kinase receptor insulin-like growth factor 1 receptor (IGF-IR) contributes to the initiation and progression of many types of malignancies. We previously showed that IGF-2, which binds IGF-IR, is an extrinsic factor that supports the ex vivo expansion of hematopoietic stem cells (HSCs). We also demonstrated that IGF-IR is not required for HSC activity in vivo. METHODS AND RESULTS: Here we investigated the role of IGF-IR in chronic myeloid leukemia (CML) using the retroviral BCR/ABL transplantation mouse model. Existing antibodies against IGF-IR are not suitable for flow cytometry; therefore, we generated a fusion of the human IgG Fc fragment with mutant IGF-2 that can bind to IGF-IR. We used this fusion protein to evaluate mouse primary hematopoietic populations. Through transplantation assays with IGF-IR(+) and IGF-IR(-) cells, we demonstrated that IGF-IR is expressed on all mouse HSCs. The expression of IGF-IR is much higher on CML cells than on acute lymphoblastic leukemia (ALL) cells. The depletion of IGF-IR expression in BCR/ABL(+) cells led to the development of ALL (mostly T cell ALL) but not CML. Lack of IGF-IR resulted in decreased self-renewal of the BCR/ABL(+) CML cells in the serial replating assay. CONCLUSION: IGF-IR regulates the cell fate determination of BCR/ABL(+) leukemia cells and supports the self-renewal of CML cells.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Receptor IGF Tipo 1/metabolismo , Animais , Western Blotting , Citometria de Fluxo , Proteínas de Fusão bcr-abl/genética , Humanos , Imuno-Histoquímica , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
8.
PLoS One ; 7(11): e48071, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23139760

RESUMO

BACKGROUND: Insulin-like growth factors (IGF-I and -II) are pleiotropic regulators of somatic growth and development in vertebrate species. Endocrine and paracrine effects of both hormones are mediated by a common IGF type 1 receptor (IGF-1R). Lethal respiratory failure in neonatal IGF-1R knockout mice suggested a particular role for this receptor in pulmonary development, and we therefore investigated the consequences of IGF-1R inactivation in lung tissue. METHODS AND FINDINGS: We first generated compound heterozygous mutant mice harboring a hypomorphic (Igf1r(neo)) and a null (Igf1r(-)) allele. These IGF-1R(neo/-) mice express only 22% of normal IGF-1R levels and are viable. In adult IGF-1R(neo/-) mice, we assessed lung morphology and respiratory physiology and found normal histomorphometric characteristics and normal breathing response to hypercapnia. We then generated homozygous IGF-1R knockout mutants (IGF-1R(-/-)) and analyzed their lung development during late gestation using histomorphometric and immunohistochemical methods. IGF-1R(-/-) embryos displayed severe lung hypoplasia and markedly underdeveloped diaphragms, leading to lethal neonatal respiratory distress. Importantly, IGF-1R(-/-) lungs from late gestation embryos were four times smaller than control lungs and showed markedly thickened intersaccular mesenchyme, indicating strongly delayed lung maturation. Cell proliferation and apoptosis were significantly increased in IGF-1R(-/-) lung tissue as compared with IGF-1R(+/+) controls. Immunohistochemistry using pro-SP-C, NKX2-1, CD31 and vWF as markers revealed a delay in cell differentiation and arrest in the canalicular stage of prenatal respiratory organ development in IGF-1R(-/-) mutant mice. CONCLUSIONS/SIGNIFICANCE: We found that low levels of IGF-1R were sufficient to ensure normal lung development in mice. In contrast, complete absence of IGF-1R significantly delayed end-gestational lung maturation. Results indicate that IGF-1R plays essential roles in cell proliferation and timing of cell differentiation during fetal lung development.


Assuntos
Pulmão/crescimento & desenvolvimento , Morfogênese , Receptor IGF Tipo 1/deficiência , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Imuno-Histoquímica , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos Knockout , Morfogênese/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Progesterona/farmacologia , Ventilação Pulmonar/efeitos dos fármacos , Receptor IGF Tipo 1/metabolismo
9.
Exp Hematol ; 40(9): 715-723.e6, 2012 09.
Artigo em Inglês | MEDLINE | ID: mdl-22613471

RESUMO

Malignant transformation of normal hematopoietic progenitors is a multistep process that likely requires interaction between collaborating oncogenic signals at critical junctures. For instance, the MLL-AF9 fusion oncogene is thought to contribute to myeloid leukemogenesis by driving a hematopoietic stem cell-like "self-renewal" gene expression signature in committed myeloid progenitors. In addition, insulin-like growth factor (IGF) signaling has been implicated in self-renewal/pluripotency in hematopoietic and embryonic stem cell contexts and supports cell growth/survival by activation of downstream pathways, including phosphatidylinositol 3-kinase/Akt and Ras/Raf/extracellular signal-regulated kinase. We hypothesized that IGF signaling could be an important contributor in the process of cellular transformation and/or clonal propagation. Utilizing an MLL-AF9 mouse bone marrow transplantation model of acute myelogenous leukemia, we discovered that committed myeloid progenitor cells with genetically reduced levels of IGF1R were less susceptible to leukemogenic transformation due, at least in part, to a cell-autonomous defect in clonogenic activity. Rather unexpectedly, genetic deletion of IGF1R by inducible Cre recombinase had no effect on growth/survival of established leukemia cells. These findings suggest that IGF1R signaling contributes to transformation of normal myeloid progenitor cells, but is not required for propagation of the leukemic clone once it has become established. We also show that treatment of mouse MLL-AF9 acute myelogenous leukemia cells with BMS-536924, an IGF1R/insulin receptor-selective tyrosine kinase inhibitor, blocked cell growth, suggesting its efficacy in this model may be due to inhibition of insulin receptor and/or related tyrosine kinases, and raising the possibility that similar IGF1R inhibitors in clinical development may be acting through alternate/related pathways.


Assuntos
Transformação Celular Neoplásica/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Fusão Oncogênica/genética , Receptor IGF Tipo 1/genética , Animais , Benzimidazóis/farmacologia , Western Blotting , Transplante de Medula Óssea , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Fusão Oncogênica/metabolismo , Pirazóis/farmacologia , Piridonas/farmacologia , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/genética , Análise de Sobrevida , Triazinas/farmacologia
10.
J Exp Med ; 208(9): 1809-22, 2011 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-21807868

RESUMO

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer of immature T cells that often shows aberrant activation of Notch1 and PI3K-Akt pathways. Although mutations that activate PI3K-Akt signaling have previously been identified, the relative contribution of growth factor-dependent activation is unclear. We show here that pharmacologic inhibition or genetic deletion of insulin-like growth factor 1 receptor (IGF1R) blocks the growth and viability of T-ALL cells, whereas moderate diminution of IGF1R signaling compromises leukemia-initiating cell (LIC) activity as defined by transplantability in syngeneic/congenic secondary recipients. Furthermore, IGF1R is a Notch1 target, and Notch1 signaling is required to maintain IGF1R expression at high levels in T-ALL cells. These findings suggest effects of Notch on LIC activity may be mediated in part by enhancing the responsiveness of T-ALL cells to ambient growth factors, and provide strong rationale for use of IGF1R inhibitors to improve initial response to therapy and to achieve long-term cure of patients with T-ALL.


Assuntos
Regulação Leucêmica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptor IGF Tipo 1/biossíntese , Receptor Notch1/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Receptor IGF Tipo 1/genética , Receptor Notch1/genética
11.
Blood ; 118(12): 3236-43, 2011 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-21821709

RESUMO

The role of IGF binding protein 2 (IGFBP2) in cell growth is intriguing and largely undefined. Previously we identified IGFBP2 as an extrinsic factor that supports ex vivo expansion of hematopoietic stem cells (HSCs). Here we showed that IGFBP2-null mice have fewer HSCs than wild-type mice. While IGFBP2 has little cell-autonomous effect on HSC function, we found decreased in vivo repopulation of HSCs in primary and secondary transplanted IGFBP2-null recipients. Importantly, bone marrow stromal cells that are deficient for IGFBP2 have significantly decreased ability to support the expansion of repopulating HSCs. To investigate the mechanism by which IGFBP2 supports HSC activity, we demonstrated that HSCs in IGFBP2-null mice had decreased survival and cycling, down-regulated expression of antiapoptotic factor Bcl-2, and up-regulated expression of cell cycle inhibitors p21, p16, p19, p57, and PTEN. Moreover, we found that the C-terminus, but not the RGD domain, of extrinsic IGFBP2 was essential for support of HSC activity. Defective signaling of the IGF type I receptor did not rescue the decreased repopulation of HSCs in IGFBP2-null recipients, suggesting that the environmental effect of IGFBP2 on HSCs is independent of IGF-IR mediated signaling. Therefore, as an environmental factor, IGFBP2 supports the survival and cycling of HSCs.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Contagem de Células , Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Regulação para Baixo/efeitos dos fármacos , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/genética , Receptor IGF Tipo 1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
12.
PLoS One ; 6(3): e18054, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21464968

RESUMO

Solid tumors are composed of cancerous cells and non-cancerous stroma. A better understanding of the tumor stroma could lead to new therapeutic applications. However, the exact compositions and functions of the tumor stroma are still largely unknown. Here, using a Lewis lung carcinoma implantation mouse model, we examined the hematopoietic compartments in tumor stroma and tumor-bearing mice. Different lineages of differentiated hematopoietic cells existed in tumor stroma with the percentage of myeloid cells increasing and the percentage of lymphoid and erythroid cells decreasing over time. Using bone marrow reconstitution analysis, we showed that the tumor stroma also contained functional hematopoietic stem cells. All hematopoietic cells in the tumor stroma originated from bone marrow. In the bone marrow and peripheral blood of tumor-bearing mice, myeloid populations increased and lymphoid and erythroid populations decreased and numbers of hematopoietic stem cells markedly increased with time. To investigate the function of hematopoietic cells in tumor stroma, we co-implanted various types of hematopoietic cells with cancer cells. We found that total hematopoietic cells in the tumor stroma promoted tumor development. Furthermore, the growth of the primary implanted Lewis lung carcinomas and their metastasis were significantly decreased in mice reconstituted with IGF type I receptor-deficient hematopoietic stem cells, indicating that IGF signaling in the hematopoietic tumor stroma supports tumor outgrowth. These results reveal that hematopoietic cells in the tumor stroma regulate tumor development and that tumor progression significantly alters the host hematopoietic compartment.


Assuntos
Compartimento Celular , Células-Tronco Hematopoéticas/patologia , Neoplasias/patologia , Animais , Contagem de Células , Diferenciação Celular , Proliferação de Células , Transplante de Células-Tronco Hematopoéticas , Camundongos , Lesões Pré-Cancerosas/patologia , Células Estromais/patologia
13.
Mol Endocrinol ; 22(11): 2531-43, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18801929

RESUMO

The receptors for IGF-I (IGF-IR) and insulin (IR) have been implicated in physiological cardiac growth, but it is unknown whether IGF-IR or IR signaling are critically required. We generated mice with cardiomyocyte-specific knockout of IGF-IR (CIGF1RKO) and compared them with cardiomyocyte-specific insulin receptor knockout (CIRKO) mice in response to 5 wk exercise swim training. Cardiac development was normal in CIGF1RKO mice, but the hypertrophic response to exercise was prevented. In contrast, despite reduced baseline heart size, the hypertrophic response of CIRKO hearts to exercise was preserved. Exercise increased IGF-IR content in control and CIRKO hearts. Akt phosphorylation increased in exercise-trained control and CIRKO hearts and, surprisingly, in CIGF1RKO hearts as well. In exercise-trained control and CIRKO mice, expression of peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) and glycogen content were both increased but were unchanged in trained CIGF1RKO mice. Activation of AMP-activated protein kinase (AMPK) and its downstream target eukaryotic elongation factor-2 was increased in exercise-trained CIGF1RKO but not in CIRKO or control hearts. In cultured neonatal rat cardiomyocytes, activation of AMPK with 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) prevented IGF-I/insulin-induced cardiomyocyte hypertrophy. These studies identify an essential role for IGF-IR in mediating physiological cardiomyocyte hypertrophy. IGF-IR deficiency promotes energetic stress in response to exercise, thereby activating AMPK, which leads to phosphorylation of eukaryotic elongation factor-2. These signaling events antagonize Akt signaling, which although necessary for mediating physiological cardiac hypertrophy, is insufficient to promote cardiac hypertrophy in the absence of myocardial IGF-I signaling.


Assuntos
Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Receptor IGF Tipo 1/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Cardiomegalia/patologia , Crescimento Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Camundongos , Camundongos Knockout , Modelos Cardiovasculares , Contração Miocárdica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Esforço Físico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/genética , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Ribonucleotídeos/farmacologia , Transdução de Sinais , Natação
14.
Cancer Res ; 68(9): 3495-504, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18451178

RESUMO

Insulin-like growth factor-I (IGF-I) is a polypeptide hormone that can influence growth, differentiation, and survival of cells expressing the cognate type 1 receptor (IGF-IR). To better understand cell autonomous IGF-IR signaling in the epithelial compartment of the prostate gland, we generated a conditional (Cre/loxP) prostate-specific IGF-IR knockout mouse model. In contrast to epidemiologic studies that established a correlation between elevated serum IGF-I and the risk of developing prostate cancer, we show that abrogation of IGF-IR expression in the dorsal and lateral prostate could activate extracellular signal-regulated kinase 1/2 signaling and cause cell autonomous proliferation and hyperplasia. Moreover, persistent loss of IGF-IR expression in dorsal and ventral lobes induced p53-regulated apoptosis and cellular senescence rescue programs, predicting that titration of IGF-IR signaling might facilitate growth of tumors with compromised p53 activity. Therefore, we crossed the mice carrying the prostate-specific IGF-IR knockout alleles into the transgenic adenocarcinoma of the mouse prostate model that is driven, in part, by T antigen-mediated functional inactivation of p53. Consistent with our prediction, prostate epithelial-specific deletion of IGF-IR accelerated the emergence of aggressive prostate cancer when p53 activity was compromised. Collectively, these data support a critical role for IGF-IR signaling in prostate tumorigenesis and identify an important IGF-IR-dependent growth control mechanism.


Assuntos
Adenocarcinoma/genética , Epitélio/metabolismo , Deleção de Genes , Próstata/metabolismo , Neoplasias da Próstata/genética , Receptor IGF Tipo 1/genética , Adenocarcinoma/metabolismo , Fatores Etários , Animais , Apoptose/genética , Proliferação de Células , Senescência Celular/genética , Regulação Neoplásica da Expressão Gênica , Marcação de Genes , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos/genética , Hiperplasia Prostática/genética , Hiperplasia Prostática/patologia , Neoplasias da Próstata/metabolismo , Receptor IGF Tipo 1/metabolismo
15.
Proc Natl Acad Sci U S A ; 104(21): 8977-82, 2007 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-17416680

RESUMO

Insulin and insulin-like growth factor 1 (IGF1) are ubiquitous growth factors that regulate proliferation in most mammalian tissues including pancreatic islets. To explore the specificity of insulin receptors in compensatory beta-cell growth, we examined two models of insulin resistance. In the first model, we used liver-specific insulin receptor knockout (LIRKO) mice, which exhibit hyperinsulinemia without developing diabetes due to a compensatory increase in beta-cell mass. LIRKO mice, also lacking functional insulin receptors in beta-cells (beta IRKO/LIRKO), exhibited severe glucose intolerance but failed to develop compensatory islet hyperplasia, together leading to early death. In the second model, we examined the relative significance of insulin versus IGF1 receptors in islet growth by feeding high-fat diets to beta IRKO and beta-cell-specific IGF1 receptor knockout (beta IGFRKO) mice. Although both groups on the high-fat diet developed insulin resistance, beta IRKO, but not beta IGFRKO, mice exhibited poor islet growth consistent with insulin-stimulated phosphorylation, nuclear exclusion of FoxO1, and reduced expression of Pdx-1. Together these data provide direct genetic evidence that insulin/FoxO1/Pdx-1 signaling is one pathway that is crucial for islet compensatory growth response to insulin resistance.


Assuntos
Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Receptor de Insulina/metabolismo , Transporte Ativo do Núcleo Celular , Ração Animal , Animais , Proliferação de Células , Gorduras/farmacologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Homeostase , Hiperplasia/induzido quimicamente , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patologia , Fígado/metabolismo , Camundongos , Camundongos Knockout , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Transdução de Sinais
16.
Mol Cell Biol ; 26(7): 2675-87, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16537911

RESUMO

The insulin-like growth factor 1 receptor (IGF-1R) is a multifunctional receptor that mediates signals for cell proliferation, differentiation, and survival. Genetic experiments showed that IGF-1R inactivation in skin results in a disrupted epidermis. However, because IGF-1R-null mice die at birth, it is difficult to study the effects of IGF-1R on skin. By using a combined approach of conditional gene ablation and a three-dimensional organotypic model, we demonstrate that IGF-1R-deficient skin cocultures show abnormal maturation and differentiation patterns. Furthermore, IGF-1R-null keratinocytes exhibit accelerated differentiation and decreased proliferation. Investigating the signaling pathway downstream of IGF-1R reveals that insulin receptor substrate 2 (IRS-2) overexpression compensates for the lack of IGF-1R, whereas IRS-1 overexpression does not. We also demonstrate that phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1 and 2 are involved in the regulation of skin keratinocyte differentiation and take some part in mediating the inhibitory signal of IGF-1R on differentiation. In addition, we show that mammalian target of rapamycin plays a specific role in mediating IGF-1R impedance of action on keratinocyte differentiation. In conclusion, these results reveal that IGF-1R plays an inhibitory role in the regulation of skin development and differentiation.


Assuntos
Diferenciação Celular , Queratinócitos/citologia , Queratinócitos/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Pele/citologia , Pele/crescimento & desenvolvimento , Animais , Apoptose , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Técnicas de Cocultura , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Queratinócitos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinases/farmacologia , Receptor IGF Tipo 1/deficiência , Serina-Treonina Quinases TOR
17.
Int J Cancer ; 114(4): 668-72, 2005 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15609331

RESUMO

Numerous tumours, including hepatocarcinomas, produce IGFs, and some depend on these growth factors in a paracrine or autocrine fashion. We have shown that c-myc-induced experimental hepatocarcinogenesis is associated with enhanced production of IGF-II. To assess the role of the IGF-I receptor (IGF-IR) in hepatocarcinogenesis, we generated conditional mutant mice that overexpressed c-myc and were knocked out for IGF-IR specifically in the liver. We compared these mice with littermate controls that also overexpressed c-myc but had wild-type IGF-IR alleles. We found that the pretumoral phase, induced by early c-myc expression and characterised by increased cell proliferation, was largely unaffected by the lack of IGF-IR. To our further surprise, hepatocellular carcinomas (HCCs) lacking IGF-IR readily developed and progressed at the same rate as control HCCs. At 9 months, all c-myc transgenic mice displayed well-differentiated multifocal tumours, regardless of whether their livers-and their tumours-were able to produce IGF-IR. Levels of IRS-1 and IRS-2 were elevated in all tumours in the presence or absence of IGF-IR, suggesting that the signalling pathway downstream of IGF-IR is activated via IGF-IR-independent mechanisms in HCC. In conclusion, the deregulation of IGF signalling pathways, which often occurs during liver tumorigenesis, does not necessarily require IGF-IRs, and hepatic IGF-IR alone may not play a determinant role in c-myc-induced hepatocarcinogenesis.


Assuntos
Neoplasias Hepáticas/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/fisiologia , Alelos , Animais , Western Blotting , Proliferação de Células , Genótipo , Hepatócitos/citologia , Hepatócitos/metabolismo , Imuno-Histoquímica , Fígado/metabolismo , Neoplasias Hepáticas/etiologia , Camundongos , Camundongos Transgênicos , Mutação , Regiões Promotoras Genéticas , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo
19.
J Clin Invest ; 111(12): 1835-42, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12813019

RESUMO

Both insulin and IGF-1 have been implicated in control of retinal endothelial cell growth, neovascularization, and diabetic retinopathy. To precisely define the role of insulin and IGF-1 signaling in endothelium in these processes, we have used the oxygen-induced retinopathy model to study mice with a vascular endothelial cell-specific knockout of the insulin receptor (VENIRKO) or IGF-1 receptor (VENIFARKO). Following relative hypoxia, VENIRKO mice show a 57% decrease in retinal neovascularization as compared with controls. This is associated with a blunted rise in VEGF, eNOS, and endothelin-1. By contrast, VENIFARKO mice show only a 34% reduction in neovascularization and a very modest reduction in mediator generation. These data indicate that both insulin and IGF-1 signaling in endothelium play a role in retinal neovascularization through the expression of vascular mediators, with the effect of insulin being most important in this process.


Assuntos
Retinopatia Diabética/prevenção & controle , Neovascularização Patológica , Receptor IGF Tipo 1/deficiência , Receptor de Insulina/deficiência , Vasos Retinianos/patologia , Animais , Retinopatia Diabética/etiologia , Retinopatia Diabética/patologia , Retinopatia Diabética/fisiopatologia , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/fisiologia , Endotelina-1/fisiologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Hipóxia/patologia , Hipóxia/fisiopatologia , Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Linfocinas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/fisiologia , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Bull Acad Natl Med ; 187(7): 1225-43; discussion 1244-7, 2003.
Artigo em Francês | MEDLINE | ID: mdl-15146601

RESUMO

Research into the somatotropic axis (growth hormone, insulin-like growth factors) has received renewed attention in the last few years, owing to techniques for genetic modification in the mouse, essentially transgenesis, and gene inactivation. The results, compared with phenotypic and molecular data for various pathological conditions, e.g. short stature, have increased our understanding of the function of the various parameters of the somatotropic axis, especially those of the insulin-like growth factor (IGF) system. During fetal development, IGF-II (which is subject to imprinting) and IGF-I are essential for increases in height and weight and for development of the various organs, adipose tissue in particular. The action of these two IGF is mediated by the IGF type 1 receptor. Total inactivation of the IGF-I gene, the paternal allele of the IGF-II gene or the IGF type 1 receptor gene leads to growth retardation resulting in mouse fetuses weighing only 60%, 60% and 45% of their wild-type controls. Combinations of these inactivations have shown that IGF-II also exerts its effects by interacting with another, as yet uncharacterized receptor. Moreover, inactivation of the IGF-I gene specifically in the liver, organ which normally maintains the high IGF-I concentrations in the blood (endocrine action), has no effect on growth, demonstrating that local IGF-I plays an auto/paracrine role in the periphery, under the direct influence of growth hormone (GH). However, endocrine IGF-I plays an important role in metabolism and cellular proliferation, and its function in mitosis may favor the development of cancer in the breast, colon and prostate. Mice with homozygous inactivation of the IGF type 1 receptor gene are not viable after birth. Partial and conditional inactivation have made it possible to study the physiology of these factors in adult animals. In contrast to IGF-II, the IGF type 1 receptor is, like IGF-I, involved in postnatal growth regulation. Lack of IGF type 1 receptor or of IGF-I affects both height and weight, and shows that this receptor is particularly involved in promoting pubertal growth. The activated receptor acts differently and specifically in each tissue. Finally, the IGF type 1 receptor has recently been implicated in the control of lifespan: a 50% decrease in the number of receptors increased the lifespan of these mice by 26%. This increase displayed sexual dimorphism, being greater for females (33%) than for males (16%). Invalidations combining different properties--specificities according to tissue, development stage or inducibility by an exogenous product--will facilitate fine dissection of the roles of the various ubiquitous actors in the IGF system.


Assuntos
Hormônio do Crescimento/genética , Hormônio do Crescimento/fisiologia , Receptor IGF Tipo 1/fisiologia , Somatomedinas/genética , Somatomedinas/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Expectativa de Vida , Camundongos , Camundongos Transgênicos , Neoplasias/fisiopatologia , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA