Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(41): 20679-20688, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31548432

RESUMO

The presence of progenitor or stem cells in the adult pancreas and their potential involvement in homeostasis and cancer development remain unresolved issues. Here, we show that mouse centroacinar cells can be identified and isolated by virtue of the mitochondrial enzyme Aldh1b1 that they uniquely express. These cells are necessary and sufficient for the formation of self-renewing adult pancreatic organoids in an Aldh1b1-dependent manner. Aldh1b1-expressing centroacinar cells are largely quiescent, self-renew, and, as shown by genetic lineage tracing, contribute to all 3 pancreatic lineages in the adult organ under homeostatic conditions. Single-cell RNA sequencing analysis of these cells identified a progenitor cell population, established its molecular signature, and determined distinct differentiation pathways to early progenitors. A distinct feature of these progenitor cells is the preferential expression of small GTPases, including Kras, suggesting that they might be susceptible to Kras-driven oncogenic transformation. This finding and the overexpression of Aldh1b1 in human and mouse pancreatic cancers, driven by activated Kras, prompted us to examine the involvement of Aldh1b1 in oncogenesis. We demonstrated genetically that ablation of Aldh1b1 completely abrogates tumor development in a mouse model of KrasG12D-induced pancreatic cancer.


Assuntos
Família Aldeído Desidrogenase 1/metabolismo , Aldeído-Desidrogenase Mitocondrial/metabolismo , Carcinoma Ductal Pancreático/patologia , Transformação Celular Neoplásica/patologia , Mutação , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Células-Tronco/patologia , Família Aldeído Desidrogenase 1/genética , Aldeído-Desidrogenase Mitocondrial/genética , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Diferenciação Celular , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Análise de Célula Única , Células-Tronco/metabolismo
2.
Nature ; 473(7346): 230-3, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21562564

RESUMO

Notch signalling is a central regulator of differentiation in a variety of organisms and tissue types. Its activity is controlled by the multi-subunit γ-secretase (γSE) complex. Although Notch signalling can play both oncogenic and tumour-suppressor roles in solid tumours, in the haematopoietic system it is exclusively oncogenic, notably in T-cell acute lymphoblastic leukaemia, a disease characterized by Notch1-activating mutations. Here we identify novel somatic-inactivating Notch pathway mutations in a fraction of patients with chronic myelomonocytic leukaemia (CMML). Inactivation of Notch signalling in mouse haematopoietic stem cells (HSCs) results in an aberrant accumulation of granulocyte/monocyte progenitors (GMPs), extramedullary haematopoieisis and the induction of CMML-like disease. Transcriptome analysis revealed that Notch signalling regulates an extensive myelomonocytic-specific gene signature, through the direct suppression of gene transcription by the Notch target Hes1. Our studies identify a novel role for Notch signalling during early haematopoietic stem cell differentiation and suggest that the Notch pathway can play both tumour-promoting and -suppressive roles within the same tissue.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor/fisiologia , Leucemia Mielomonocítica Crônica/genética , Leucemia Mielomonocítica Crônica/patologia , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células Cultivadas , Perfilação da Expressão Gênica , Inativação Gênica , Células Progenitoras de Granulócitos e Macrófagos/citologia , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Receptores Notch/deficiência , Fatores de Transcrição HES-1 , Células Tumorais Cultivadas
3.
Nature ; 459(7249): 1000-4, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19536265

RESUMO

T-cell acute lymphoblastic leukaemia (T-ALL) is a blood malignancy afflicting mainly children and adolescents. T-ALL patients present at diagnosis with increased white cell counts and hepatosplenomegaly, and are at an increased risk of central nervous system (CNS) relapse. For that reason, T-ALL patients usually receive cranial irradiation in addition to intensified intrathecal chemotherapy. The marked increase in survival is thought to be worth the considerable side-effects associated with this therapy. Such complications include secondary tumours, neurocognitive deficits, endocrine disorders and growth impairment. Little is known about the mechanism of leukaemic cell infiltration of the CNS, despite its clinical importance. Here we show, using T-ALL animal modelling and gene-expression profiling, that the chemokine receptor CCR7 (ref. 5) is the essential adhesion signal required for the targeting of leukaemic T-cells into the CNS. Ccr7 gene expression is controlled by the activity of the T-ALL oncogene Notch1 and is expressed in human tumours carrying Notch1-activating mutations. Silencing of either CCR7 or its chemokine ligand CCL19 (ref. 6) in an animal model of T-ALL specifically inhibits CNS infiltration. Furthermore, murine CNS-targeting by human T-ALL cells depends on their ability to express CCR7. These studies identify a single chemokine-receptor interaction as a CNS 'entry' signal, and open the way for future pharmacological targeting. Targeted inhibition of CNS involvement in T-ALL could potentially decrease the intensity of CNS-targeted therapy, thus reducing its associated short- and long-term complications.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Leucemia de Células T/metabolismo , Leucemia de Células T/patologia , Receptores CCR7/metabolismo , Transdução de Sinais , Animais , Adesão Celular , Linhagem Celular Tumoral , Quimiocina CCL19/deficiência , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores CCR7/deficiência
4.
Am J Physiol Renal Physiol ; 302(11): F1362-73, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22461304

RESUMO

The adult kidney contains a population of low-cycling cells that resides in the papilla. These cells retain for long periods S-phase markers given as a short pulse early in life; i.e., they are label-retaining cells (LRC). In previous studies in adult rat and mice, we found that shortly after acute kidney injury many of the quiescent papillary LRC started proliferating (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009; Oliver JA, Maarouf O, Cheema FH, Martens TP, Al-Awqati Q. J Clin Invest 114: 795-804, 2004) and, with cell-tracking experiments, we found upward migration of some papillary cells including LRC (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009). To identify molecular cues involved in the activation (i.e., proliferation and/or migration) of the papillary LRC that follows injury, we isolated these cells from the H2B-GFP mice and found that they migrated and proliferated in response to the cytokine stromal cell-derived factor-1 (SDF-1). Moreover, in a papillary organ culture assay, the cell growth out of the upper papilla was dependent on the interaction of SDF-1 with its receptor Cxcr4. Interestingly, location of these two proteins in the kidney revealed a complementary location, with SDF-1 being preferentially expressed in the medulla and Cxcr4 more abundant in the papilla. Blockade of Cxcr4 in vivo prevented mobilization of papillary LRC after transient kidney ischemic injury and worsened its functional consequences. The data indicate that the SDF-1/Cxcr4 axis is a critical regulator of papillary LRC activation following transient kidney injury and during organ repair.


Assuntos
Injúria Renal Aguda/patologia , Quimiocina CXCL12/farmacologia , Nefropatias/patologia , Medula Renal/crescimento & desenvolvimento , Injúria Renal Aguda/fisiopatologia , Animais , Western Blotting , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Indicadores e Reagentes , Nefropatias/fisiopatologia , Medula Renal/patologia , Medula Renal/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo
5.
Proc Natl Acad Sci U S A ; 106(7): 2359-64, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19174523

RESUMO

Considering the strong association between dysregulated insulin-like growth factor (IGF) signaling and various human cancers, we have used an expedient combination of genetic analysis and pharmacological treatment to evaluate the potential of the type 1 IGF receptor (Igf1r) for targeted anticancer therapy in a mouse model of mammary tumorigenesis. In this particular strain of genetically modified animals, histopathologically heterogeneous invasive carcinomas exhibiting up-regulation of the Igf1r gene developed extremely rapidly by mammary gland-specific overexpression of constitutively active oncogenic Kras* (mutant Kras(G12D)). Immunophenotyping data and expression profiling analyses showed that, except for a minor luminal component, these mouse tumors resembled basal-like human breast cancers. This is a group of aggressive tumors of poor prognosis for which there is no targeted therapy currently available, and it includes a subtype correlating with KRAS locus amplification. Conditional ablation of Igf1r in the mouse mammary epithelium increased the latency of Kras*-induced tumors very significantly (approximately 11-fold in comparison with the intact model), whereas treatment of tumor-bearing animals by administration of picropodophyllin (PPP), a specific Igf1r inhibitor, resulted in a dramatic decrease in tumor mass of the main forms of basal-like carcinomas. PPP also was effective against xenografts of the human basal-like cancer cell line MDA-MB-231, which carries a KRAS(G13D) mutation.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Regulação Neoplásica da Expressão Gênica , Receptor IGF Tipo 1/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Podofilotoxina/análogos & derivados , Podofilotoxina/farmacologia , Transdução de Sinais , Proteínas ras/metabolismo
6.
J Biol Chem ; 285(52): 41044-50, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20947509

RESUMO

Signaling by receptor tyrosine kinases regulates pancreatic ß cell function. Inactivation of insulin receptor (InsR), IGF1 receptor (Igf1r), or Irs1 in ß cells impairs insulin secretion. Conversely, Irs2 ablation impairs ß cell replication. In this study, we examined aspects of the Igf1r regulatory signaling cascade in ß cells. To examine genetically the involvement of Irs1 and Irs2 in Igf1r signaling, we generated double mutant mice lacking Igf1r specifically in pancreatic ß cells in an Irs1- or Irs2-null background. We show that Igf1r/Irs1 double mutants do not differ phenotypically from Irs1 single mutants and exhibit hyperinsulinemia, while maintaining normal ß cell mass and glucose tolerance. In contrast, lack of Igf1r function in ß cells aggravates the consequences of Irs2 ablation in double mutants and results in lethal diabetes by 6 weeks of age. This additivity of phenotypic manifestations indicates that Irs2 serves a pathway that is largely independent of Igf1r signaling. Consistent with the view that the latter is the InsR pathway, we show that combined ß cell-specific knock-out of both Insr and Igf1r results in a phenocopy of double mutants lacking Igf1r and Irs2. We conclude that Igf1r signals primarily through Irs1 and affects insulin secretion, whereas ß cell proliferation is mainly regulated by InsR using Irs2 as a downstream signaling effector. The insulin and IGF pathways appear to control ß cell functions independently and selectively.


Assuntos
Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Animais , Proliferação de Células , Diabetes Mellitus/genética , Diabetes Mellitus/mortalidade , Glucose/genética , Glucose/metabolismo , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Insulina/genética , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Camundongos , Camundongos Knockout , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
7.
Proc Natl Acad Sci U S A ; 105(49): 19378-83, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-19033454

RESUMO

The mammalian insulin-like growth factor 1 (IGF1), which is a member of a major growth-promoting signaling system, is produced by many tissues and functions throughout embryonic and postnatal development in an autocrine/paracrine fashion. In addition to this local action, IGF1 secreted by the liver and circulating in the plasma presumably acts systemically as a classical hormone. However, an endocrine role of IGF1 in growth control was disputed on the basis of the results of a conditional, liver-specific Igf1 gene knockout in mice, which reduced significantly the level of serum IGF1, but did not affect average body weight. Because alternate interpretations of these negative data were tenable, we addressed genetically the question of hormonal IGF1 action by using a positive experimental strategy based on the features of the cre/loxP recombination system. Thus, we generated bitransgenic mice carrying in an Igf1 null background a dormant Igf1 cDNA placed downstream of a transcriptional "stop" DNA sequence flanked by loxP sites (floxed) and also a cre transgene driven by a liver-specific promoter. The Igf1 cDNA, which was inserted by knock-in into the mutated and inactive Igf1 locus itself to ensure proper transcriptional regulation, was conditionally expressed from cognate promoters exclusively in the liver after Cre-mediated excision of the floxed block. Our genetic study demonstrated that the endocrine IGF1 plays a very significant role in mouse growth, as its action contributes approximately30% of the adult body size and sustains postnatal development, including the reproductive functions of both mouse sexes.


Assuntos
Sistema Endócrino/crescimento & desenvolvimento , Sistema Endócrino/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Animais , Sequência de Bases , Tamanho Corporal/fisiologia , Feminino , Fertilidade/fisiologia , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fenótipo
8.
Am J Pathol ; 174(1): 276-86, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095950

RESUMO

Mutations in the phosphatase and tensin homologue (PTEN)/phosphatidylinositol-3 kinase-alpha (PI3K) signaling pathway are frequently found in human cancer. In addition, Pten(+/-) mice develop tumors in multiple organs because of the activation of the PI3K signaling cascade. Because activation of PI3K signaling leads to feedback inhibition of insulin receptor substrate-2 (IRS2) expression, an upstream activator of PI3K, we therefore anticipated that IRS2 expression would be low in tumors that lack PTEN. Surprisingly, however, an elevation of IRS2 was often detected in tumor samples in which PTEN levels were compromised. To determine the potential contribution of Irs2 to tumor progression, Pten(+/-) mice were crossed with Irs2(+/-) mice. Deletion of Irs2 did not affect the initiation of neoplasia found in Pten(+/-) mice but suppressed cancer cell growth, proliferation, and invasion through the basement membrane. Deletion of Irs2 also attenuated the expression of Myc in prostatic intraepithelial neoplasia in Pten(+/-) mice. In addition, the expression levels of IRS2 and MYC were highly correlated in human prostate cancer, and IRS2 could stimulate MYC expression in cultured cells. Our findings provide evidence that the PI3K-activating adaptor Irs2 contributes to tumor progression in Pten(+/-) mice by stimulating both Myc and DNA synthesis.


Assuntos
Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Progressão da Doença , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Mutantes , Neoplasias/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transfecção
9.
Nature ; 426(6964): 291-5, 2003 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-14628051

RESUMO

In mice, gonads are formed shortly before embryonic day 10.5 by the thickening of the mesonephros and consist of somatic cells and migratory primordial germ cells. The male sex-determining process is set in motion by the sex-determining region of the Y chromosome (Sry), which triggers differentiation of the Sertoli cell lineage. In turn, Sertoli cells function as organizing centres and direct differentiation of the testis. In the absence of Sry expression, neither XX nor XY gonads develop testes, and alterations in Sry expression are often associated with abnormal sexual differentiation. The molecular signalling mechanisms by which Sry specifies the male pathway and models the undifferentiated gonad are unknown. Here we show that the insulin receptor tyrosine kinase family, comprising Ir, Igf1r and Irr, is required for the appearance of male gonads and thus for male sexual differentiation. XY mice that are mutant for all three receptors develop ovaries and show a completely female phenotype. Reduced expression of both Sry and the early testis-specific marker Sox9 indicates that the insulin signalling pathway is required for male sex determination.


Assuntos
Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Diferenciação Sexual , Transdução de Sinais , Testículo/embriologia , Testículo/metabolismo , Animais , Biomarcadores/análise , Diferenciação Celular , Divisão Celular , Transtornos do Desenvolvimento Sexual , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Masculino , Camundongos , Camundongos Knockout , Mutação/genética , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo , Fenótipo , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Cromossomos Sexuais/genética , Diferenciação Sexual/genética , Testículo/citologia
10.
J Am Soc Nephrol ; 20(11): 2315-27, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19762493

RESUMO

The kidney papilla contains a population of cells with several characteristics of adult stem cells, including the retention of proliferation markers during long chase periods (i.e., they are label-retaining cells [LRCs]). To determine whether the papillary LRCs generate new cells in the normal adult kidney, we examined cell proliferation throughout the kidney and found that the upper papilla is a site of enhanced cell cycling. Using genetically modified mice that conditionally expressed green fluorescence protein fused to histone 2B, we observed that the LRCs of the papilla proliferated only in its upper part, where they associate with "chains" of cycling cells. The papillary LRCs decreased in number with age, suggesting that the cells migrated to the upper papilla before entering the cell cycle. To test this directly, we marked papillary cells with vital dyes in vivo and found that some cells in the kidney papilla, including LRCs, migrated toward other parts of the kidney. Acute kidney injury enhanced both cell migration and proliferation. These results suggest that during normal homeostasis, LRCs of the kidney papilla (or their immediate progeny) migrate to the upper papilla and form a compartment of rapidly proliferating cells, which may play a role in repair after ischemic injury.


Assuntos
Movimento Celular , Proliferação de Células , Rim/citologia , Fatores Etários , Animais , Rim/crescimento & desenvolvimento , Ratos , Coloração e Rotulagem
11.
Sci Rep ; 10(1): 207, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937792

RESUMO

Using an efficient gene targeting approach, we developed a novel mouse line that expresses the tetracycline-controlled transactivator (tTA) from the constitutively active Eef1a1 locus in a Cre recombinase-inducible manner. The temporally and spatially controlled expression of the EF1-LSL-tTA knockin and activation of tTA-driven responder transgenes was tested using four transgenic lines that express Cre under tissue-specific promoters of the pancreas, mammary gland and other secretory tissues, as well as an interferon-inducible promoter. In all models, the endogenous Eef1a1 promoter facilitated a cell-type-specific activation of target genes at high levels without exogenous enhancer elements. The applicability of the EF1-LSL-tTA strain for biological experiments was tested in two studies related to mammary gland development and tumorigenesis. First, we validated the crucial role of active STAT5 as a survival factor for functionally differentiated epithelial cells by expressing a hyperactive STAT5 mutant in the mammary gland during postlactational remodeling. In a second experiment, we assessed the ability of the EF1-tTA to initiate tumor formation through upregulation of mutant KRAS. The collective results show that the EF1-LSL-tTA knockin line is a versatile genetic tool that can be applied to constitutively express transgenes in specific cell types to examine their biological functions at defined developmental stages.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fator 1 de Elongação de Peptídeos/metabolismo , Tetraciclina/farmacologia , Transgenes/fisiologia , Animais , Antibacterianos/farmacologia , Feminino , Genes Reporter , Integrases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Distribuição Tecidual , Transativadores
12.
Mol Cancer Res ; 18(6): 822-834, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32152233

RESUMO

The PI3K signaling pathway is frequently mutated in head and neck squamous cell carcinoma (HNSCC), often via gain-of-function (GOF) mutations in the PIK3CA gene. Here, we present novel genetically engineered mouse models (GEMM) carrying a GOF allele Loxp-STOP-Loxp(LSL)-PIK3CAH1047R (E20) alone or in combination with heterozygous LSL-p53+/R172H (p53) mutation with tissue-specific expression to interrogate the role of oncogenic PIK3CA in transformation of upper aerodigestive track epithelium. We demonstrated that the GOF PIK3CA mutation promoted progression of 4-nitroquinoline 1-oxide-induced oral squamous cell carcinoma (OSCC) in both E20 single mutant and E20/p53 double mutant mice, with frequent distal metastasis detected only in E20/p53 GEMM. Similar to in human OSCC, loss of p16 was associated with progression of OSCC in these mice. RNA-seq analyses revealed that among the common genes differentially expressed in primary OSCC cell lines derived from E20, p53, and E20/p53 GEMMs compared with those from the wild-type mice, genes associated with proliferation and cell cycle were predominantly represented, which is consistent with the progressive loss of p16 detected in these GEMMs. Importantly, all of these OSCC primary cell lines exhibited enhanced sensitivity to BYL719 and cisplatin combination treatment in comparison with cisplatin alone in vitro and in vivo, regardless of p53 and/or p16 status. Given the prevalence of mutations in p53 and the PI3K pathways in HNSCC in conjunction with loss of p16 genetically or epigenetically, this universal increased sensitivity to cisplatin and BYL719 combination therapy in cancer cells with PIK3CA mutation represents an opportunity to a subset of patients with HNSCC. IMPLICATIONS: Our results suggest that combination therapy of cisplatin and PI3K inhibitor may be worthy of consideration in patients with HNSCC with PIK3CA mutation.


Assuntos
4-Nitroquinolina-1-Óxido/toxicidade , Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias de Cabeça e Pescoço/patologia , Mutação , Carcinoma de Células Escamosas de Cabeça e Pescoço/secundário , Proteína Supressora de Tumor p53/genética , Animais , Carcinógenos/toxicidade , Progressão da Doença , Neoplasias de Cabeça e Pescoço/induzido quimicamente , Neoplasias de Cabeça e Pescoço/genética , Camundongos , Camundongos Nus , Carcinoma de Células Escamosas de Cabeça e Pescoço/induzido quimicamente , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
13.
Mol Cell Biol ; 25(5): 1869-78, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15713641

RESUMO

Studies of Drosophila and mammals have revealed the importance of insulin signaling through phosphatidylinositol 3-kinase and the serine/threonine kinase Akt/protein kinase B for the regulation of cell, organ, and organismal growth. In mammals, three highly conserved proteins, Akt1, Akt2, and Akt3, comprise the Akt family, of which the first two are required for normal growth and metabolism, respectively. Here we address the function of Akt3. Like Akt1, Akt3 is not required for the maintenance of normal carbohydrate metabolism but is essential for the attainment of normal organ size. However, in contrast to Akt1-/- mice, which display a proportional decrease in the sizes of all organs, Akt3-/- mice present a selective 20% decrease in brain size. Moreover, although Akt1- and Akt3-deficient brains are reduced in size to approximately the same degree, the absence of Akt1 leads to a reduction in cell number, whereas the lack of Akt3 results in smaller and fewer cells. Finally, mammalian target of rapamycin signaling is attenuated in the brains of Akt3-/- but not Akt1-/- mice, suggesting that differential regulation of this pathway contributes to an isoform-specific regulation of cell growth.


Assuntos
Encéfalo/enzimologia , Encéfalo/crescimento & desenvolvimento , Proteínas Oncogênicas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Glicemia/análise , Peso Corporal/genética , Peso Corporal/fisiologia , Encéfalo/citologia , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Insulina/sangue , Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miocárdio/citologia , Proteínas Oncogênicas/genética , Tamanho do Órgão/genética , Tamanho do Órgão/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases S6 Ribossômicas/metabolismo
14.
J Clin Invest ; 113(2): 209-19, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14722613

RESUMO

Insulin promotes both metabolism and growth. However, it is unclear whether insulin-dependent growth is merely a result of its metabolic actions. Targeted ablation of insulin receptor (Insr) has not clarified this issue, because of early postnatal lethality. To examine this question, we generated mice with variable cellular mosaicism for null Insr alleles. Insr ablation in approximately 80% of cells caused extreme growth retardation, lipoatrophy, and hypoglycemia, a clinical constellation that resembles the human syndrome of leprechaunism. Insr ablation in 98% of cells, while resulting in similar growth retardation and lipoatrophy, caused diabetes without beta-cell hyperplasia. The growth retardation was associated with a greater than 60-fold increase in the expression of hepatic insulin-like growth factor binding protein-1. These findings indicate that insulin regulates growth independently of metabolism and that the number of insulin receptors is an important determinant of the specificity of insulin action.


Assuntos
Receptor de Insulina/genética , Tecido Adiposo/metabolismo , Alelos , Animais , Northern Blotting , Western Blotting , Divisão Celular , Genótipo , Hiperplasia , Hipoglicemia/genética , Imuno-Histoquímica , Insulina/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Ligantes , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Mosaicismo , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
15.
J Clin Invest ; 110(7): 1011-9, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12370279

RESUMO

Defective insulin secretion is a feature of type 2 diabetes that results from inadequate compensatory increase of beta cell mass and impaired glucose-dependent insulin release. beta cell proliferation and secretion are thought to be regulated by signaling through receptor tyrosine kinases. In this regard, we sought to examine the potential proliferative and/or antiapoptotic role of IGFs in beta cells by tissue-specific conditional mutagenesis ablating type 1 IGF receptor (IGF1R) signaling. Unexpectedly, lack of functional IGF1R did not affect beta cell mass, but resulted in age-dependent impairment of glucose tolerance, associated with a decrease of glucose- and arginine-dependent insulin release. These observations reveal a requirement of IGF1R-mediated signaling for insulin secretion.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptor IGF Tipo 1/fisiologia , Animais , Exocitose , Transportador de Glucose Tipo 2 , Secreção de Insulina , Camundongos , Proteínas de Transporte de Monossacarídeos/análise , Fenótipo , RNA Mensageiro/análise , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/genética
16.
Oncogene ; 23(8): 1558-65, 2004 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-14661057

RESUMO

We have developed and tested successfully a general method based on Cre-mediated recombination that can be used for ubiquitous or tissue-specific expression of protein products, including tumor-inducing oncoproteins. Depending on the specificity of a chosen promoter driving cre expression, tumors develop by design in bitransgenic mouse progeny derived by crossing Cre-producing mice with partners carrying a dormant oncogenic transgene (targeted into the 3' noncoding region of the cytoplasmic beta-actin locus) that becomes functional after excision of a 'floxed' DNA segment. To provide proof-of-principle, we have used as models transgenes encoding the polyomavirus middle T antigen (PVMT) and the T antigens of the SV40 early region (SVER). Cre-dependent activation of widespread SVER expression resulted in hyperplasias or invasive tumors affecting particular visceral smooth muscles, whereas Cre-dependent, mammary gland-specific expression of PVMT-induced adenocarcinomas, according to plan. Unexpectedly, we also encountered spontaneous (Cre-independent) oncogene expression occurring as a rare event, which simulates the initiation of sporadic tumors and leads to PVMT-induced hemangiomas and mammary carcinomas or SVER-induced disseminated sarcomas, thus, revealing particular tissue susceptibilities to the actions of these oncoproteins.


Assuntos
Modelos Animais de Doenças , Mutagênese Insercional/métodos , Neoplasias/genética , Proteínas Oncogênicas/metabolismo , Actinas/genética , Animais , Antígenos Transformantes de Poliomavirus/genética , Camundongos , Camundongos Transgênicos , Músculo Liso/patologia , Neoplasias/patologia , Proteínas Oncogênicas/genética , Oncogenes , Regiões Promotoras Genéticas , Transgenes
17.
J Neurosci ; 23(20): 7710-8, 2003 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-12930811

RESUMO

We examined the role of IGF signaling in the remyelination process by disrupting the gene encoding the type 1 IGF receptor (IGF1R) specifically in the mouse brain by Cre-mediated recombination and then exposing these mutants and normal siblings to cuprizone. This neurotoxicant induces a demyelinating lesion in the corpus callosum that is reversible on termination of the insult. Acute demyelination and oligodendrocyte depletion were the same in mutants and controls, but the mutants did not remyelinate adequately. We observed that oligodendrocyte progenitors did not accumulate, proliferate, or survive within the mutant mice, compared with wild type, indicating that signaling through the IGF1R plays a critical role in remyelination via effects on oligodendrocyte progenitors.


Assuntos
Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Sistema Nervoso Central/fisiologia , Corpo Caloso/citologia , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/fisiologia , Cuprizona/toxicidade , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , Microglia/citologia , Mutação , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Receptor IGF Tipo 1/genética , Somatomedinas/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Fator de Necrose Tumoral alfa/análise
18.
J Natl Cancer Inst ; 106(12)2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25306215

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDA) is frequently driven by oncogenic KRAS(KRAS*) mutations. We developed a mouse model of KRAS*-induced PDA and, based on genetic results demonstrating that KRAS* tumorigenicity depends on Myc activity, we evaluated the therapeutic potential of an orally administered anti-Myc drug. METHODS: We tested the efficacy of Mycro3, a small-molecule inhibitor of Myc-Max dimerization, in the treatment of mouse PDA (n = 9) and also of xenografts of human pancreatic cancer cell lines (NOD/SCID mice, n = 3-12). Tumor responses to the drug were evaluated by PET/CT imaging, and histological, immunohistochemical, molecular and microarray analyses. The Student's t test was used for differences between groups. All statistical tests were two-sided. RESULTS: Transgenic overexpression of KRAS* in the pancreas resulted in pancreatic intraepithelial neoplasia in two-week old mice, which developed invasive PDA a week later and became moribund at one month. However, this aggressive form of pancreatic tumorigenesis was effectively prevented by genetic ablation of Myc specifically in the pancreas. We then treated moribund, PDA-bearing mice daily with the Mycro3 Myc-inhibitor. The mice survived until killed at two months. PET/CT image analysis (n = 5) demonstrated marked shrinkage of PDA, while immunohistochemical analyses showed an increase in cancer cell apoptosis and reduction in cell proliferation (treated/untreated proliferation index ratio: 0.29, P < .001, n = 3, each group). Tumor growth was also drastically attenuated in Mycro3-treated NOD/SCID mice (n = 12) carrying orthotopic or heterotopic xenografts of human pancreatic cancer cells (eg, mean tumor weight ± SD of treated heterotopic xenografts vs vehicle-treated controls: 15.2±5.8 mg vs 230.2±43.9 mg, P < .001). CONCLUSION: These results provide strong justification for eventual clinical evaluation of anti-Myc drugs as potential chemotherapeutic agents for the treatment of PDA.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Tiazóis/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Técnicas de Introdução de Genes , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Tomografia por Emissão de Pósitrons , Proteínas Proto-Oncogênicas c-myc/genética , Tomografia Computadorizada por Raios X , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Mol Cancer Ther ; 10(4): 697-707, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21447712

RESUMO

Inhibition of the insulin-like growth factor 1 receptor (Igf1r) is an approach being taken in clinical trials to overcome the dismal outcome for metastatic alveolar rhabdomyosarcoma (ARMS), an aggressive muscle cancer of children and young adults. In our study, we address the potential mechanism(s) of Igf1r inhibitor resistance that might be anticipated for patients. Using a genetically engineered mouse model of ARMS, validated for active Igf1r signaling, we show that the prototypic Igf1r inhibitor NVP-AEW541 can inhibit cell growth and induce apoptosis in vitro in association with decreased Akt and Mapk phosphorylation. However, drug resistance in vivo is more common and is accompanied by Igf1r overexpression, Mapk reactivation, and Her2 overexpression. Her2 is found to form heterodimers with Igf1r in resistant primary tumor cell cultures, and stimulation with Igf2 leads to Her2 phosphorylation. The Her2 inhibitor lapatinib cooperates with NVP-AEW541 to reduce Igf1r phosphorylation and to inhibit cell growth even though lapatinib alone has little effect on growth. These results point to the potential therapeutic importance of simultaneous targeting of Igf1r and Her2 to abrogate resistance.


Assuntos
Pirimidinas/farmacologia , Pirróis/farmacologia , Receptor IGF Tipo 1/antagonistas & inibidores , Rabdomiossarcoma/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Criança , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/patologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Lapatinib , Camundongos , Fosforilação/efeitos dos fármacos , Codorniz , Quinazolinas/farmacologia , Interferência de RNA , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rabdomiossarcoma/genética , Rabdomiossarcoma/patologia , Carga Tumoral/efeitos dos fármacos , Células Tumorais Cultivadas , Adulto Jovem
20.
Neuron ; 57(6): 847-57, 2008 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-18367086

RESUMO

Olfactory neurons project their axons to spatially invariant glomeruli in the olfactory bulb, forming an ordered pattern of innervation comprising the olfactory sensory map. A mirror symmetry exists within this map, such that neurons expressing a given receptor typically project to one glomerulus on the medial face and one glomerulus on the lateral face of the bulb. The mechanisms underlying an olfactory neuron's choice to project medially versus laterally remain largely unknown, however. Here we demonstrate that insulin-like growth factor (IGF) signaling is required for sensory innervation of the lateral olfactory bulb. Mutations that eliminate IGF signaling cause axons destined for targets in the lateral bulb to shift to ectopic sites on the ventral-medial surface. Using primary cultures of olfactory and cerebellar neurons, we further show that IGF is a chemoattractant for axon growth cones. Together these observations reveal a role of IGF signaling in sensory map formation and axon guidance.


Assuntos
Axônios/fisiologia , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Condutos Olfatórios/fisiologia , Neurônios Receptores Olfatórios/citologia , Transdução de Sinais/fisiologia , Animais , Axônios/efeitos dos fármacos , Cerebelo/citologia , Fatores Quimiotáticos/farmacologia , Cromonas/farmacologia , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Técnicas In Vitro , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Camundongos Transgênicos , Morfolinas/farmacologia , Mutação/fisiologia , Bulbo Olfatório/citologia , Bulbo Olfatório/embriologia , Bulbo Olfatório/crescimento & desenvolvimento , Proteína de Marcador Olfatório/genética , Proteína de Marcador Olfatório/metabolismo , Condutos Olfatórios/embriologia , Neurônios Receptores Olfatórios/efeitos dos fármacos , Neurônios Receptores Olfatórios/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA