Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34681917

RESUMO

Malignant melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. We recently showed that the extracellular signal-regulated kinase 5 (ERK5), encoded by the MAPK7 gene, plays a pivotal role in melanoma by regulating cell functions necessary for tumour development, such as proliferation. Hedgehog-GLI signalling is constitutively active in melanoma and is required for proliferation. However, no data are available in literature about a possible interplay between Hedgehog-GLI and ERK5 pathways. Here, we show that hyperactivation of the Hedgehog-GLI pathway by genetic inhibition of the negative regulator Patched 1 increases the amount of ERK5 mRNA and protein. Chromatin immunoprecipitation showed that GLI1, the major downstream effector of Hedgehog-GLI signalling, binds to a functional non-canonical GLI consensus sequence at the MAPK7 promoter. Furthermore, we found that ERK5 is required for Hedgehog-GLI-dependent melanoma cell proliferation, and that the combination of GLI and ERK5 inhibitors is more effective than single treatments in reducing cell viability and colony formation ability in melanoma cells. Together, these findings led to the identification of a novel Hedgehog-GLI-ERK5 axis that regulates melanoma cell growth, and shed light on new functions of ERK5, paving the way for new therapeutic options in melanoma and other neoplasms with active Hedgehog-GLI and ERK5 pathways.


Assuntos
MAP Quinase Quinase 5/genética , Melanoma/genética , Proteína Quinase 7 Ativada por Mitógeno/genética , Neoplasias Cutâneas/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Imunoprecipitação da Cromatina , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , MAP Quinase Quinase 5/metabolismo , Melanoma/metabolismo , Camundongos , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Células NIH 3T3 , Receptor Patched-1/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Melanoma Maligno Cutâneo
2.
Cancer Res ; 81(16): 4218-4229, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34215617

RESUMO

Meningiomas are the most common benign brain tumors. Mutations of the E3 ubiquitin ligase TRAF7 occur in 25% of meningiomas and commonly cooccur with mutations in KLF4, yet the functional link between TRAF7 and KLF4 mutations remains unclear. By generating an in vitro meningioma model derived from primary meningeal cells, we elucidated the cooperative interactions that promote meningioma development. By integrating TRAF7-driven ubiquitinome and proteome alterations in meningeal cells and the TRAF7 interactome, we identified TRAF7 as a proteostatic regulator of RAS-related small GTPases. Meningioma-associated TRAF7 mutations disrupted either its catalytic activity or its interaction with RAS GTPases. TRAF7 loss in meningeal cells altered actin dynamics and promoted anchorage-independent growth by inducing CDC42 and RAS signaling. TRAF deficiency-driven activation of the RAS/MAPK pathway promoted KLF4-dependent transcription that led to upregulation of the tumor-suppressive Semaphorin pathway, a negative regulator of small GTPases. KLF4 loss of function disrupted this negative feedback loop and enhanced mutant TRAF7-mediated cell transformation. Overall, this study provides new mechanistic insights into meningioma development, which could lead to novel treatment strategies. SIGNIFICANCE: The intricate molecular cross-talk between the ubiquitin ligase TRAF7 and the transcription factor KLF4 provides a first step toward the identification of new therapies for patients with meningioma.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Meningioma/genética , Mutação , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Proteínas ras/genética , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Biologia Computacional , Células HEK293 , Humanos , Fator 4 Semelhante a Kruppel/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteoma , Semaforinas/metabolismo , Análise de Sequência de DNA , Transdução de Sinais , Ativação Transcricional , Ubiquitina/química , Proteína cdc42 de Ligação ao GTP/genética , Proteínas ras/metabolismo
3.
Circ Res ; 126(10): 1379-1393, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32175818

RESUMO

RATIONALE: Noonan syndrome (NS) is one of the most frequent genetic disorders. Bleeding problems are among the most common, yet poorly defined complications associated with NS. A lack of consensus on the management of bleeding complications in patients with NS indicates an urgent need for new therapeutic approaches. OBJECTIVE: Bleeding disorders have recently been described in patients with NS harboring mutations of LZTR1 (leucine zipper-like transcription regulator 1), an adaptor for CUL3 (CULLIN3) ubiquitin ligase complex. Here, we assessed the pathobiology of LZTR1-mediated bleeding disorders. METHODS AND RESULTS: Whole-body and vascular specific knockout of Lztr1 results in perinatal lethality due to cardiovascular dysfunction. Lztr1 deletion in blood vessels of adult mice leads to abnormal vascular leakage. We found that defective adherent and tight junctions in Lztr1-depleted endothelial cells are caused by dysregulation of vesicular trafficking. LZTR1 affects the dynamics of fusion and fission of recycling endosomes by controlling ubiquitination of the ESCRT-III (endosomal sorting complex required for transport III) component CHMP1B (charged multivesicular protein 1B), whereas NS-associated LZTR1 mutations diminish CHMP1B ubiquitination. LZTR1-mediated dysregulation of CHMP1B ubiquitination triggers endosomal accumulation and subsequent activation of VEGFR2 (vascular endothelial growth factor receptor 2) and decreases blood levels of soluble VEGFR2 in Lztr1 haploinsufficient mice. Inhibition of VEGFR2 activity by cediranib rescues vascular abnormalities observed in Lztr1 knockout mice Conclusions: Lztr1 deletion phenotypically overlaps with bleeding diathesis observed in patients with NS. ELISA screening of soluble VEGFR2 in the blood of LZTR1-mutated patients with NS may predict both the severity of NS phenotypes and potential responders to anti-VEGF therapy. VEGFR inhibitors could be beneficial for the treatment of bleeding disorders in patients with NS.


Assuntos
Vasos Sanguíneos/metabolismo , Endossomos/metabolismo , Células Endoteliais/metabolismo , Hemorragia/metabolismo , Síndrome de Noonan/metabolismo , Fatores de Transcrição/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Malformações Vasculares/metabolismo , Animais , Vasos Sanguíneos/anormalidades , Vasos Sanguíneos/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Modelos Animais de Doenças , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/genética , Endossomos/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Haploinsuficiência , Células HeLa , Hemorragia/genética , Hemorragia/patologia , Hemorragia/prevenção & controle , Humanos , Linfocinas/genética , Linfocinas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica , Síndrome de Noonan/tratamento farmacológico , Síndrome de Noonan/genética , Síndrome de Noonan/patologia , Fosforilação , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Quinazolinas/farmacologia , Transdução de Sinais , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Ubiquitinação , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Malformações Vasculares/tratamento farmacológico , Malformações Vasculares/genética , Malformações Vasculares/patologia
4.
Oncogene ; 37(19): 2601-2614, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29483645

RESUMO

Malignant melanoma is among the most aggressive cancers and its incidence is increasing worldwide. Targeted therapies and immunotherapy have improved the survival of patients with metastatic melanoma in the last few years; however, available treatments are still unsatisfactory. While the role of the BRAF-MEK1/2-ERK1/2 pathway in melanoma is well established, the involvement of mitogen-activated protein kinases MEK5-ERK5 remains poorly explored. Here we investigated the function of ERK5 signaling in melanoma. We show that ERK5 is consistently expressed in human melanoma tissues and is active in melanoma cells. Genetic silencing and pharmacological inhibition of ERK5 pathway drastically reduce the growth of melanoma cells and xenografts harboring wild-type (wt) or mutated BRAF (V600E). We also found that oncogenic BRAF positively regulates expression, phosphorylation, and nuclear localization of ERK5. Importantly, ERK5 kinase and transcriptional transactivator activities are enhanced by BRAF. Nevertheless, combined pharmacological inhibition of BRAFV600E and MEK5 is required to decrease nuclear ERK5, that is critical for the regulation of cell proliferation. Accordingly, combination of MEK5 or ERK5 inhibitors with BRAFV600E inhibitor vemurafenib is more effective than single treatments in reducing colony formation and growth of BRAFV600E melanoma cells and xenografts. Overall, these data support a key role of the ERK5 pathway for melanoma growth in vitro and in vivo and suggest that targeting ERK5, alone or in combination with BRAF-MEK1/2 inhibitors, might represent a novel approach for melanoma treatment.


Assuntos
Melanoma/patologia , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos , Transplante de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Vemurafenib/farmacologia
5.
Methods Mol Biol ; 1322: 71-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26179040

RESUMO

The measurement of the transcriptional activity of the HH signaling pathway is widely used as an indication of pathway activation. Luciferase reporter assays are powerful tools to measure the specific ability of a transcription factor to bind to its consensus sequence and to activate transcription of target genes. Here, we describe a protocol to measure the transcriptional activity of the HH pathway in normal and cancer cells. This technique allows studying the activity of GLI transcription factors and their modulation by drugs and/or other factors.


Assuntos
Expressão Gênica , Genes Reporter , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Transcrição Gênica , Técnicas de Cultura de Células , Linhagem Celular , Humanos , Luciferases/genética , Luciferases/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Transfecção
6.
Expert Rev Mol Med ; 17: e5, 2015 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-25660620

RESUMO

The HEDGEHOG-GLI (HH-GLI) signalling is a key pathway critical in embryonic development, stem cell biology and tissue homeostasis. In recent years, aberrant activation of HH-GLI signalling has been linked to several types of cancer, including those of the skin, brain, lungs, prostate, gastrointestinal tract and blood. HH-GLI signalling is initiated by binding of HH ligands to the transmembrane receptor PATCHED and is mediated by transcriptional effectors that belong to the GLI family, whose activity is finely tuned by a number of molecular interactions and post-translation modifications. Several reports suggest that the activity of the GLI proteins is regulated by several proliferative and oncogenic inputs, in addition or independent of upstream HH signalling. The identification of this complex crosstalk and the understanding of how the major oncogenic signalling pathways interact in cancer is a crucial step towards the establishment of efficient targeted combinatorial treatments. Here we review recent findings on the cooperative integration of HH-GLI signalling with the major oncogenic inputs and we discuss how these cues modulate the activity of the GLI proteins in cancer. We then summarise the latest advances on SMO and GLI inhibitors and alternative approaches to attenuate HH signalling through rational combinatorial therapies.


Assuntos
Transformação Celular Neoplásica , Proteínas Hedgehog/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/genética , Humanos , Ligantes , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco
7.
Ann Saudi Med ; 34(1): 1-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24658547

RESUMO

Pancreatic neuroendocrine neoplasms are relatively rare entities, representing approximately 1% to 2% of all pancreatic tumors. Owing to their rarity as well as their relatively indolent natural history, treatment approaches are not yet standardized. A formal pancreatic resection is usually mandatory for large and localized sporadic pancreatic tumors or in the presence of symptoms. However, in small and asymptomatic lesions, a conservative approach consisting in a careful wait-and-see policy is going to appear as more appropriate, particularly when, to remove the lesion, an aggressive surgical procedure is required, such as pancreaticoduodenectomy or distal splenopancreatectomy, depending on the localization of the tumor. Surgery has also a significant role in locally advanced and metastatic forms. In the setting of MEN 1 syndrome or Von-Hippel Lindau disease, the tumor size and the possible symptoms should be considered in the evaluation of a proper treatment.


Assuntos
Tumores Neuroendócrinos/cirurgia , Neoplasias Pancreáticas/cirurgia , Humanos , Neoplasia Endócrina Múltipla Tipo 1/complicações , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Pancreaticoduodenectomia , Carga Tumoral , Doença de von Hippel-Lindau/complicações
8.
Curr Protoc Stem Cell Biol ; Chapter 3: Unit 3.6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23404674

RESUMO

Melanoma is the most aggressive skin cancer. This unit illustrates protocols for culture and isolation of human melanoma cancer stem cells/tumor-initiating cells (CSC/TIC). We describe two complementary methods to enrich for melanoma CSC/TIC. The first approach exploits the ability of CSC/TIC to grow as tumor spheres in low-adherent culture conditions, as previously shown for neural stem cells and human embryonic stem cells. As a second approach, melanoma CSC/TIC are enriched by fluorescence-activated cell sorting for the aldehyde dehydrogenase (ALDH) enzyme activity. We previously showed that melanoma cells with high ALDH activity (ALDH(high)) are endowed with higher self-renewal and tumorigenic abilities than the population with low activity (ALDH(low)), suggesting that ALDH might be a good marker to select for melanoma CSC/TIC. This unit will also describe how to functionally test melanoma CSC/TIC by determining self-renewal in vitro and tumor-forming abilities in vivo using orthotopic xenograft assay.


Assuntos
Separação Celular/métodos , Melanoma/patologia , Células-Tronco Neoplásicas/patologia , Animais , Proliferação de Células , Humanos , Camundongos , Camundongos Nus , Esferoides Celulares/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cell Cycle ; 12(2): 353-64, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23287475

RESUMO

One undisputed milestone of traditional oncology is neoplastic progression, which consists of a progressive selection of dedifferentiated cells driven by a chance sequence of genetic mutations. Recently it has been demonstrated that the overexpression of well-defined transcription factors reprograms somatic cells to the pluripotent stem status. The demonstration raises crucial questions as to whether and to what extent this reprogramming contributes to tumorigenesis, and whether the epigenetic changes involved in it are reversible. Here, we show for the first time that a tumor produced in vivo by a chemical carcinogen is the product of the interaction between neoplastic progression and reprogramming. The experimental model employed the prototype of ascites tumors, the Yoshida AH130 hepatoma and other neoplasias, including human melanoma. AH130 hepatoma was started in the liver by the carcinogen o-aminoazotoluene. This compound binds to and abolishes the p53 protein, producing a genomic instability that promotes both the neoplastic progression and the hepatoma reprogramming. Eventually this tumor contained 100% CD133(+) elements and pO(2)-dependent percentages of the three embryonic transcription factors Nanog, Klf4 and c-Myc. Once transferred into aerobic cultures, the minor cellular fraction expressing this triad generates various types of adherent cells, which are progressively substituted by non-tumorigenic elements committed to fibromuscular, neuronal and glial differentiation. This reprogramming appears to be accomplished stepwise, with the assembly of the triad into a sophisticated transcriptional, oxygen-dependent circuitry, in which Nanog and Klf4 antagonistically regulate c-Myc, and hence, cell hypoxia survival and cell cycle activation.


Assuntos
Desdiferenciação Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/metabolismo , Animais , Desdiferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Primers do DNA/genética , Citometria de Fluxo , Instabilidade Genômica/efeitos dos fármacos , Humanos , Fator 4 Semelhante a Kruppel , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Microscopia Eletrônica , Proteína Homeobox Nanog , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Proteína Supressora de Tumor p53/metabolismo , o-Aminoazotolueno/metabolismo , o-Aminoazotolueno/toxicidade
10.
Stem Cells ; 30(9): 1808-18, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22730244

RESUMO

The question of whether cancer stem/tumor-initiating cells (CSC/TIC) exist in human melanomas has arisen in the last few years. Here, we have used nonadherent spheres and the aldehyde dehydrogenase (ALDH) enzymatic activity to enrich for CSC/TIC in a collection of human melanomas obtained from a broad spectrum of sites and stages. We find that melanomaspheres display extensive in vitro self-renewal ability and sustain tumor growth in vivo, generating human melanoma xenografts that recapitulate the phenotypic composition of the parental tumor. Melanomaspheres express high levels of Hedgehog (HH) pathway components and of embryonic pluripotent stem cell factors SOX2, NANOG, OCT4, and KLF4. We show that human melanomas contain a subset of cells expressing high ALDH activity (ALDH(high)), which is endowed with higher self-renewal and tumorigenic abilities than the ALDH(low) population. A good correlation between the number of ALDH(high) cells and sphere formation efficiency was observed. Notably, both pharmacological inhibition of HH signaling by the SMOOTHENED (SMO) antagonist cyclopamine and GLI antagonist GANT61 and stable expression of shRNA targeting either SMO or GLI1 result in a significant decrease in melanoma stem cell self-renewal in vitro and a reduction in the number of ALDH(high) melanoma stem cells. Finally, we show that interference with the HH-GLI pathway through lentiviral-mediated silencing of SMO and GLI1 drastically diminishes tumor initiation of ALDH(high) melanoma stem cells. In conclusion, our data indicate an essential role of the HH-GLI1 signaling in controlling self-renewal and tumor initiation of melanoma CSC/TIC. Targeting HH-GLI1 is thus predicted to reduce the melanoma stem cell compartment.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteínas Hedgehog/metabolismo , Melanoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fatores de Transcrição/metabolismo , Aldeído Desidrogenase/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Células HEK293 , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/genética , Humanos , Fator 4 Semelhante a Kruppel , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Transplante Heterólogo , Proteína GLI1 em Dedos de Zinco
11.
Cancer Res ; 69(2): 616-24, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19147576

RESUMO

Activation of hypoxia-inducible factors (HIF), responsible for tumor angiogenesis and glycolytic switch, is regulated by reduced oxygen availability. Normally, HIF-alpha proteins are maintained at low levels, controlled by site-specific hydroxylation carried out by HIF prolyl hydroxylases (PHD) and subsequent proteasomal degradation via the von Hippel-Lindau ubiquitin ligase. Using a yeast two-hybrid screen, we identified an interaction between melanoma antigen-11 (MAGE-11) cancer-testis antigen and the major HIF-alpha hydroxylating enzyme PHD2. The interaction was confirmed by a pull-down assay, coimmunoprecipitation, and colocalization in both normoxic and hypoxic conditions. Furthermore, MAGE-9, the closest homologue of MAGE-11, was also found to interact with PHD2. MAGE-11 inhibited PHD activity without affecting protein levels. This inhibition was accompanied by stabilization of ectopic or endogenous HIF-1alpha protein. Knockdown of MAGE-11 by small interfering RNA results in decreased hypoxic induction of HIF-1alpha and its target genes. Inhibition of PHD by MAGE-11, and following activation of HIFs, is a novel tumor-associated HIF regulatory mechanism. This finding provides new insights into the significance of MAGE expression in tumors and may provide valuable tools for therapeutic intervention because of the restricted expression of the MAGE gene family in cancers, but not in normal tissues.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Sequência de Aminoácidos , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Regulação para Baixo , Células HeLa , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Prolina Dioxigenases do Fator Induzível por Hipóxia , Dados de Sequência Molecular , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Interferência de RNA , Transcrição Gênica , Transfecção , Ubiquitina/metabolismo
12.
Stem Cells ; 25(3): 807-15, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17158237

RESUMO

The comprehension of the basic biology of stem cells is expected to provide a useful insight into the pathogenesis of cancer. In particular, there is evidence that hypoxia promotes stem cell renewal in vitro as well as in vivo. It therefore seems reasonable that stem cell survival and hypoxia response are strictly connected at molecular level. We here report that the 66-kDa isoform of the SHC gene (p66Shc) is induced in a breast cancer cell line by the exposure to hypoxic environment and that it controls the expression of the stem cell regulatory gene Notch-3. Then, we show that p66Shc/Notch-3 interplay modulates self-renewal (by inducing the Notch-ligand Jagged-1) and hypoxia survival (by inducing the hypoxia-survival gene carbonic anhydrase IX) in mammary gland stem/progenitor cells, expanded in vitro as multicellular spheroids (mammospheres). We conclude that mechanisms that regulate stem cell renewal and hypoxia survival are integrated at the level of the p66Shc/Notch3 interplay. Because Notch-3, Jagged-1, and carbonic anhydrase IX are dysregulated in breast cancer, and because p66Shc is an aging-regulating gene, we envision that these data may help in understanding the relationship among aging, cancer, and stem cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Mama/química , Mama/fisiologia , Divisão Celular/fisiologia , Hipóxia Celular/fisiologia , Receptores Notch/fisiologia , Células-Tronco/fisiologia , Neoplasias da Mama , Carcinoma Ductal/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Luciferases/genética , Receptor Notch3 , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA