Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Commun Biol ; 5(1): 1068, 2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36207615

RESUMO

TGF-ß signaling is involved in pancreatic ductal adenocarcinoma (PDAC) tumorigenesis, representing one of the four major pathways genetically altered in 100% of PDAC cases. TGF-ß exerts complex and pleiotropic effects in cancers, notably via the activation of SMAD pathways, predominantly SMAD2/3/4. Though SMAD2 and 3 are rarely mutated in cancers, SMAD4 is lost in about 50% of PDAC, and the role of SMAD2/3 in a SMAD4-null context remains understudied. We herein provide evidence of a SMAD2/3 oncogenic effect in response to TGF-ß1 in SMAD4-null human PDAC cancer cells. We report that inactivation of SMAD2/3 in SMAD4-negative PDAC cells compromises TGF-ß-driven collective migration mediated by FAK and Rho/Rac signaling. Moreover, RNA-sequencing analyses highlight a TGF-ß gene signature related to aggressiveness mediated by SMAD2/3 in the absence of SMAD4. Using a PDAC patient cohort, we reveal that SMAD4-negative tumors with high levels of phospho-SMAD2 are more aggressive and have a poorer prognosis. Thus, loss of SMAD4 tumor suppressive activity in PDAC leads to an oncogenic gain-of-function of SMAD2/3, and to the onset of associated deleterious effects.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Proteína Smad3/metabolismo , Carcinogênese/genética , Carcinoma Ductal Pancreático/metabolismo , Humanos , Neoplasias Pancreáticas/metabolismo , RNA , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Neoplasias Pancreáticas
2.
Sci Rep ; 10(1): 14491, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32879407

RESUMO

GLI1 expression is broadly accepted as a marker of Hedgehog pathway activation in tumors. Efficacy of Hedgehog inhibitors is essentially limited to tumors bearing activating mutations of the pathway. GLI2, a critical Hedgehog effector, is necessary for GLI1 expression and is a direct transcriptional target of TGF-ß/SMAD signaling. We examined the expression correlations of GLI1/2 with TGFB and HH genes in 152 distinct transcriptome datasets totaling over 23,500 patients and representing 37 types of neoplasms. Their prognostic value was measured in over 15,000 clinically annotated tumor samples from 26 tumor types. In most tumor types, GLI1 and GLI2 follow a similar pattern of expression and are equally correlated with HH and TGFB genes. However, GLI1/2 broadly share prognostic value with TGFB genes and a mesenchymal/EMT signature, not with HH genes. Our results provide a likely explanation for the frequent failure of anti-Hedgehog therapies in tumors, as they suggest a key role for TGF-ß, not Hedgehog, ligands, in tumors with elevated GLI1/2-expression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/genética , Neoplasias/diagnóstico , Proteínas Nucleares/genética , Fator de Crescimento Transformador beta/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/genética , Biologia Computacional , Perfilação da Expressão Gênica , Humanos , Ligantes , Análise Multivariada , Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Modelos de Riscos Proporcionais , Fatores de Risco , Transdução de Sinais/genética , Transcriptoma
3.
Biochem J ; 477(17): 3131-3145, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32766732

RESUMO

The Hedgehog-regulated transcription factors GLI1 and GLI2 play overlapping roles in development and disease; however, the mechanisms underlying their interplay remain elusive. We report for the first time that GLI1 and GLI2 physically and functionally interact in cancer cells. GLI1 and GLI2 were shown to co-immunoprecipitate in PANC1 pancreatic cancer cells and RMS13 rhabdomyosarcoma cells. Mapping analysis demonstrated that the zinc finger domains of both proteins are required for their heteromerization. RNAi knockdown of either GLI1 or GLI2 inhibited expression of many well-characterized GLI target genes (BCL2, MYCN, PTCH2, IL7 and CCND1) in PANC1 cells, whereas PTCH1 expression was only inhibited by GLI1 depletion. qPCR screening of a large set of putative canonical and non-canonical Hedgehog/GLI targets identified further genes (e.g. E2F1, BMP1, CDK2) strongly down-regulated by GLI1 and/or GLI2 depletion in PANC1 cells, and demonstrated that ANO1, AQP1 and SOCS1 are up-regulated by knockdown of either GLI1 or GLI2. Chromatin immunoprecipitation showed that GLI1 and GLI2 occupied the same regions at the BCL2, MYCN and CCND1 promoters. Furthermore, depletion of GLI1 inhibited GLI2 occupancy at these promoters, suggesting that GLI1/GLI2 interaction is required for the recruitment of GLI2 to these sites. Together, these findings indicate that GLI1 and GLI2 co-ordinately regulate the transcription of some genes, and provide mechanistic insight into the roles of GLI proteins in carcinogenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Rabdomiossarcoma/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo , Linhagem Celular Tumoral , Proteínas Hedgehog/genética , Humanos , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Multimerização Proteica , Rabdomiossarcoma/genética , Rabdomiossarcoma/patologia , Proteína GLI1 em Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/genética
4.
J Dermatol Sci ; 94(3): 321-329, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31208857

RESUMO

BACKGROUND: Tyrosinase-Related Protein 2 (TRP2) is an enzyme involved in melanogenesis, that also exerts proliferative, anti-apoptotic and immunogenic functions in melanoma cells. TRP2 transcription is regulated by the melanocytic master transcription factor MITF. GLI2, a transcription factor that acts downstream of Hedgehog signaling, is also a direct transcriptional target of the TGF-ß/SMAD pathway that contributes to melanoma progression and exerts transcriptional antagonistic activities against MITF. OBJECTIVES: To characterize the molecular events responsible for TGF-ß and GLI2 repression of TRP2 expression. METHODS: In silico promoter analysis, transient cell transfection experiments with 5'-end TRP2 promoter deletion constructs, chromatin immuno-precipitation, and site-directed promoter mutagenesis were used to dissect the molecular mechanisms of TRP2 gene regulation by TGF-ß and GLI2. RESULTS: We demonstrate that TGF-ß and GLI2-specific TRP2 repression involves direct mechanisms that occur in addition to MITF downregulation by TGF-ß and GLI2. We identify two functional GLI2 binding sites within the TRP2 promoter that are critical for TGF-ß and GLI2 responsiveness, one of them overlapping a CREB binding site. GLI2 and CREB competing for the same cis-element is associated with opposite transcriptional outcome. CONCLUSION: Our results further refine the understanding of how TGF-ß and GLI2 control the phenotypic plasticity of melanoma cells. In particular, we identify critical GLI2-binding cis-elements within the TRP2 promoter region that allow for its transcriptional repression independently from MITF concomitant downregulation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Oxirredutases Intramoleculares/genética , Melanoma/genética , Proteínas Nucleares/metabolismo , Neoplasias Cutâneas/genética , Fator de Crescimento Transformador beta/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo , Linhagem Celular Tumoral , Biologia Computacional , Humanos , Melanoma/patologia , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas/genética , Neoplasias Cutâneas/patologia , Transcrição Gênica
5.
J Investig Dermatol Symp Proc ; 19(2): S87-S88, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30471761

RESUMO

Hedgehog (HH) signaling plays an important role both during embryonic development and adult life. It is involved in the regulation of cell differentiation, cell proliferation and tissue polarity, as well as in the maintenance of stem cells, tissue repair, and regeneration (Briscoe and Therond, 2013; Jiang and Hui, 2008). Three ligands, Indian, Sonic, and Desert HH, can activate this pathway. Binding of HH ligands to their receptor, PTCH1 (Figure 1) lift its inhibition on SMO, resulting in activation and nuclear translocation of GLI transcription factors (Javelaud et al., 2012). The vertebrate GLI gene family is composed of three distinct genes GLI1, GLI2, and GLI3, encoding Krüppel-like transcription factors. GLI proteins exhibit distinct regulations, biochemical properties, and target genes. GLI3 acts as the main repressor of the pathway in the absence of HH ligands, whereas, in their presence, GLI2 is the main HH effector that drives the expression of GLI1 (Briscoe and Therond, 2013).

6.
Int J Biochem Cell Biol ; 98: 75-81, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29544895

RESUMO

The ubiquitous distribution of both Hippo and TGF-ß signaling cascade components and their critical implication in tissue homeostasis and disease has led to the discovery of a remarkable slew of interesting and unique features regarding their functional crosstalks. Upstream cellular cues regulating the Hippo pathway, including cell-cell contacts and apico-basal cell polarity have been well characterized. Herein, we provide an overview of the published models of compartmentalized signaling crosstalk mechanisms between Hippo signaling and the TGF-ß/SMAD pathway. How cell polarity impacts the interaction between the two pathways is discussed, together with the specifics of cytoplasmic and nuclear events implicating SMADs and YAP/TAZ, leading to contextual regulation of target gene expression.


Assuntos
Polaridade Celular , Células Epiteliais/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Epiteliais/citologia , Via de Sinalização Hippo , Humanos , Transdução de Sinais
7.
Oncotarget ; 8(49): 86447-86462, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29156807

RESUMO

More efficient therapies that target multiple molecular mechanisms are needed for the treatment of incurable bone metastases. Halofuginone is a plant alkaloid-derivative with antiangiogenic and antiproliferative effects. Here we demonstrate that halofuginone is an effective therapy for the treatment of bone metastases, through multiple actions that include inhibition of TGFß and BMP-signaling. Halofuginone blocked TGF-ß-signaling in MDA-MB-231 and PC3 cells showed by inhibition of TGF-ß-induced Smad-reporter, phosphorylation of Smad-proteins, and expression of TGF-ß-regulated metastatic genes. Halofuginone increased inhibitory Smad7-mRNA and reduced TGF-ß-receptor II protein. Proline supplementation but not Smad7-knockdown reversed halofuginone-inhibition of TGF-ß-signaling. Halofuginone also decreased BMP-signaling. Treatment of MDA-MB-231 and PC3 cells with halofuginone reduced the BMP-Smad-reporter (BRE)4, Smad1/5/8-phosphorylation and mRNA of the BMP-regulated gene Id-1. Halofuginone decreased immunostaining of phospho-Smad2/3 and phospho-Smad1/5/8 in cancer cells in vivo. Furthermore, halofuginone decreased tumor-take and growth of orthotopic-tumors. Mice with breast or prostate bone metastases treated with halofuginone had significantly less osteolysis than control mice. Combined treatment with halofuginone and zoledronic-acid significantly reduced osteolytic area more than either treatment alone. Thus, halofuginone reduces breast and prostate cancer bone metastases in mice and combined with treatment currently approved by the FDA is an effective treatment for this devastating complication of breast and prostate-cancer.

8.
Dev Cell ; 32(5): 640-51, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25758862

RESUMO

Cell-cell contacts inhibit cell growth and proliferation in part by activating the Hippo pathway that drives the phosphorylation and nuclear exclusion of the transcriptional coactivators YAP and TAZ. Cell density and Hippo signaling have also been reported to block transforming growth factor ß (TGF-ß) responses, based on the ability of phospho-YAP/TAZ to sequester TGF-ß-activated SMAD complexes in the cytoplasm. Herein, we provide evidence that epithelial cell polarization interferes with TGF-ß signaling well upstream and independent of cytoplasmic YAP/TAZ. Rather, polarized basolateral presentation of TGF-ß receptors I and II deprives apically delivered TGF-ß of access to its receptors. Basolateral ligand delivery nonetheless remains entirely effective to induce TGF-ß responses. These data demonstrate that cell-type-specific inhibition of TGF-ß signaling by cell density is restricted to polarized epithelial cells and reflects the polarized distribution of TGF-ß receptors, which thus affects SMAD activation irrespective of Hippo pathway activation.


Assuntos
Citoplasma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Aciltransferases , Western Blotting , Contagem de Células , Proteínas de Ciclo Celular , Proliferação de Células , Células Cultivadas , Imunofluorescência , Via de Sinalização Hippo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Proteínas Nucleares/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética
9.
Arch Dermatol Res ; 307(4): 351-64, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740152

RESUMO

With aging, epidermal homeostasis and barrier function are disrupted. In a previous study, we analyzed the transcriptomic response of young skin epidermis after stratum corneum removal, and obtained a global kinetic view of the molecular processes involved in barrier function recovery. In the present study, the same analysis was performed in aged skin in order to better understand the defects which occur with aging. Thirty healthy male volunteers (67 ± 4 years old) were involved. Tape-strippings were carried out on the inner face of one forearm, the other unstripped forearm serving as control. At 2, 6, 18, 30 and 72 h after stripping, TEWL measurements were taken, and epidermis samples were collected. Total RNA was extracted and analyzed using DermArray(®) cDNA microarrays. The results highlighted that barrier function recovery and overall kinetics of gene expression were delayed following stripping in aged skin. Indeed, the TEWL measurements showed that barrier recovery in the young group appeared to be dramatically significant during the overall kinetics, while there were no significant evolution in the aged group until 30 h. Moreover, gene expression analysis revealed that the number of modulated genes following tape stripping increased as a function of time and reached a peak at 6 h after tape stripping in young skin, while it was at 30 h in aged skin, showing that cellular activity linked to the repair process may be engaged earlier in young epidermis than in aged epidermis. A total of 370 genes were modulated in the young group. In the aged group, 382 genes were modulated, whose 184 were also modulated in the young group. Only eight genes that were modulated in both groups were significantly differently modulated. The characterization of these genes into 15 functional families helped to draw a scenario for the aging process affecting epidermal repair capacity.


Assuntos
Epiderme/fisiologia , Expressão Gênica/fisiologia , Envelhecimento da Pele/fisiologia , Cicatrização/genética , Fatores Etários , Idoso , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade
10.
J Invest Dermatol ; 134(1): 123-132, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23897276

RESUMO

YAP and its paralog protein TAZ are downstream effectors of the Hippo pathway. Both are amplified in many human cancers and promote cell proliferation and epithelial-mesenchymal transition. Little is known about the status of the Hippo pathway in cutaneous melanoma. We profiled Hippo pathway component expression in a panel of human melanoma cell lines and melanocytic lesions, and characterized the capacity of YAP and TAZ to control melanoma cell behavior. YAP and TAZ immuno-staining in human samples revealed mixed cytoplasmic and nuclear staining for both proteins in benign nevi and superficial spreading melanoma. TAZ was expressed at higher levels than YAP1/2 in all cell lines and in those with high invasive potential. Stable YAP or TAZ knockdown dramatically reduced the expression of the classical Hippo target CCN2/connective-tissue growth factor (CTGF), as well as anchorage-independent growth, capacity to invade Matrigel, and ability form lung metastases in mice following tail-vein injection. YAP knockdown also reduced invasion in a model of skin reconstruct. Inversely, YAP overexpression increased melanoma cell invasiveness, associated with increased TEA domain-dependent transcription and CCN2/CTGF expression. Together, these results demonstrate that both YAP and TAZ contribute to the invasive and metastatic capacity of melanoma cells and may represent worthy targets for therapeutic intervention.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Melanoma/metabolismo , Fosfoproteínas/metabolismo , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/metabolismo , Aciltransferases , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Via de Sinalização Hippo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma/patologia , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética , Proteínas de Sinalização YAP
11.
Pigment Cell Melanoma Res ; 26(6): 861-73, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23890107

RESUMO

In melanoma cells, high expression of the transcription factor GLI2 is associated with increased invasive potential and loss of E-cadherin expression, an event reminiscent of the epithelial-to-mesenchymal transition (EMT). Herein, we provide evidence that GLI2 represses E-cadherin gene (CDH1) expression in melanoma cells via distinct mechanisms, enhancing transcription of the EMT-activator ZEB1 and cooperative repression of CDH1 gene transcription via direct binding of both GLI2 and ZEB1 to two closely positioned Kruppel-like factor-binding sites within the CDH1 promoter. GLI2 silencing rescued CDH1 expression except in melanoma cell lines in which the CDH1 promoter was hypermethylated. Proximity ligation assays identified GLI2-ZEB1 complexes in melanoma cell nuclei, proportional to endogenous GLI2 and ZEB1 expression, and whose accumulation was enhanced by the classical EMT inducer TGF-ß. These data identify GLI2 as a critical modulator of the cadherin switch in melanoma, a molecular process that is critical for metastatic spread of the disease.


Assuntos
Caderinas/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Melanoma/genética , Proteínas Nucleares/metabolismo , Neoplasias Cutâneas/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Antígenos CD , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Humanos , Melanoma/patologia , Modelos Biológicos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição da Família Snail , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Homeobox 1 de Ligação a E-box em Dedo de Zinco , Proteína Gli2 com Dedos de Zinco
12.
Ann Dermatol ; 25(2): 135-44, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23717002

RESUMO

Transforming growth factor-ß (TGF-ß) is a pleiotropic growth factor with broad tissue distribution that plays critical roles during embryonic development, normal tissue homeostasis, and cancer. While its cytostatic activity on normal epithelial cells initially defined TGF-ß signaling as a tumor suppressor pathway, there is ample evidence indicating that TGF-ß is a potent pro-tumorigenic agent, acting via autocrine and paracrine mechanisms to promote peri-tumoral angiogenesis, together with tumor cell migration, immune escape, and dissemination to metastatic sites. This review summarizes the current knowledge on the implication of TGF-ß signaling in melanoma.

13.
Pharmacol Ther ; 137(2): 183-99, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23063491

RESUMO

Recent advances in the field of cancer therapeutics come from the development of drugs that specifically recognize validated oncogenic or pro-metastatic targets. The latter may be mutated proteins with altered function, such as kinases that become constitutively active, or critical components of growth factor signaling pathways, whose deregulation leads to aberrant malignant cell proliferation and dissemination to metastatic sites. We herein focus on the description of the overlapping activities of two important developmental pathways often exacerbated in cancer, namely Transforming Growth Factor-ß (TGF-ß) and Hedgehog (HH) signaling, with a special emphasis on the unifying oncogenic role played by GLI1/2 transcription factors. The latter are the main effectors of the canonical HH pathway, yet are direct target genes of TGF-ß/SMAD signal transduction. While tumor-suppressor in healthy and pre-malignant tissues, TGF-ß is often expressed at high levels in tumors and contributes to tumor growth, escape from immune surveillance, invasion and metastasis. HH signaling regulates cell proliferation, differentiation and apoptosis, and aberrant HH signaling is found in a variety of cancers. We discuss the current knowledge on HH and TGF-ß implication in cancer including cancer stem cell biology, as well as the current state, both successes and failures, of targeted therapeutics aimed at blocking either of these pathways in the pre-clinical and clinical settings.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Hedgehog/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador alfa/antagonistas & inibidores , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Ensaios Clínicos como Assunto , Proteínas Hedgehog/genética , Humanos , Neoplasias/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , Fator de Crescimento Transformador alfa/genética
14.
Cancer Res ; 72(23): 6247-56, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23002206

RESUMO

TGF-ß derived from bone fuels melanoma bone metastases by inducing tumor secretion of prometastatic factors that act on bone cells to change the skeletal microenvironment. Halofuginone is a plant alkaloid derivative that blocks TGF-ß signaling with antiangiogenic and antiproliferative properties. Here, we show for the first time that halofuginone therapy decreases development and progression of bone metastasis caused by melanoma cells through the inhibition of TGF-ß signaling. Halofuginone treatment of human melanoma cells inhibited cell proliferation, phosphorylation of SMAD proteins in response to TGF-ß, and TGF-ß-induced SMAD-driven transcription. In addition, halofuginone reduced expression of TGF-ß target genes that enhance bone metastases, including PTHrP, CTGF, CXCR4, and IL11. Also, cell apoptosis was increased in response to halofuginone. In nude mice inoculated with 1205 Lu melanoma cells, a preventive protocol with halofuginone inhibited bone metastasis. The beneficial effects of halofuginone treatment were comparable with those observed with other anti-TGF-ß strategies, including systemic administration of SD208, a small-molecule inhibitor of TGF-ß receptor I kinase, or forced overexpression of Smad7, a negative regulator of TGF-ß signaling. Furthermore, mice with established bone metastases treated with halofuginone had significantly less osteolysis than mice receiving placebo assessed by radiography. Thus, halofuginone is also effective in reducing the progression of melanoma bone metastases. Moreover, halofuginone treatment reduced melanoma metastasis to the brain, showing the potential of this novel treatment against cancer metastasis.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Melanoma/tratamento farmacológico , Piperidinas/farmacologia , Quinazolinonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Expressão Gênica , Humanos , Melanoma/metabolismo , Melanoma/patologia , Melanoma/secundário , Camundongos , Camundongos Nus , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
15.
FEBS Lett ; 586(14): 2016-25, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22609357

RESUMO

Hedgehog (HH) and TGF-ß signals control various aspects of embryonic development and cancer progression. While their canonical signal transduction cascades have been well characterized, there is increasing evidence that these pathways are able to exert overlapping activities that challenge efficient therapeutic targeting. We herein review the current knowledge on HH signaling and summarize the recent findings on the crosstalks between the HH and TGF-ß pathways in cancer.


Assuntos
Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Transição Epitelial-Mesenquimal , Fibrose/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Ligantes , Camundongos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Processamento Pós-Transcricional do RNA , Ratos , Transdução de Sinais , Transativadores/metabolismo , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
16.
J Biol Chem ; 287(22): 17996-8004, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22496449

RESUMO

The melanocyte-specific transcription factor M-MITF is involved in numerous aspects of melanoblast lineage biology including pigmentation, survival, and migration. It plays complex roles at all stages of melanoma progression and metastasis. We established previously that GLI2, a Kruppel-like transcription factor that acts downstream of Hedgehog signaling, is a direct transcriptional target of the TGF-ß/SMAD pathway and contributes to melanoma progression, exerting antagonistic activities against M-MITF to control melanoma cell invasiveness. Herein, we dissected the molecular mechanisms underlying both TGF-ß and GLI2-driven M-MITF gene repression. Using transient cell transfection experiments with M-MITF promoter constructs, chromatin immunoprecipitation, site-directed mutagenesis, and electrophoretic mobility shift assays, we identified a GLI2 binding site within the -334/-296 region of the M-MITF promoter, critical for GLI2-driven transcriptional repression. This region is, however, not needed for inhibition of M-MITF promoter activity by TGF-ß. We determined that TGF-ß rapidly repressed protein kinase A activity, thus reducing both phospho-cAMP-response element-binding protein (CREB) levels and CREB-dependent transcription of the M-MITF promoter. Increased GLI2 binding to its cognate cis-element, associated with reduced CREB-dependent transcription, allowed maximal inhibition of the M-MITF promoter via two distinct mechanisms.


Assuntos
Fatores de Transcrição Kruppel-Like/fisiologia , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/genética , Proteínas Nucleares/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , DNA de Neoplasias/genética , Progressão da Doença , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica , Humanos , Fator de Transcrição Associado à Microftalmia/fisiologia , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica , Proteína Gli2 com Dedos de Zinco
17.
Pigment Cell Melanoma Res ; 25(3): 343-53, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22336146

RESUMO

There is growing evidence that the metastatic spread of melanoma is driven not by a linear increase in tumorigenic aggressiveness, but rather by switching back and forth between two different phenotypes of metastatic potential. In vitro these phenotypes are respectively defined by the characteristics of strong proliferation/weak invasiveness and weak proliferation/strong invasiveness. Melanoma cell phenotype is tightly linked to gene expression. Taking advantage of this, we have developed a gene expression-based tool for predicting phenotype called Heuristic Online Phenotype Prediction. We demonstrate the predictive utility of this tool by comparing phenotype-specific signatures with measurements of characteristics of melanoma phenotype-specific biology in different melanoma cell lines and short-term cultures. We further show that 86% of 536 tested melanoma lines and short-term cultures are significantly associated with the phenotypes we describe. These findings reinforce the concept that a two-state system, as described by the phenotype switching model, underlies melanoma progression.


Assuntos
Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Perfilação da Expressão Gênica , Melanoma/classificação , Neoplasias Cutâneas/classificação , Proliferação de Células , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos/genética , Estudos de Associação Genética , Humanos , Melanoma/genética , Melanoma/patologia , Análise em Microsséries , Invasividade Neoplásica , Fenótipo , Reprodutibilidade dos Testes , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Distribuição Tecidual , Células Tumorais Cultivadas/classificação
18.
Cancer Res ; 71(17): 5606-10, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21862631

RESUMO

The Hedgehog (HH) and TGF-ß signaling pathways represent essential regulators of cell proliferation and differentiation during embryogenesis. Pathway deregulation is a characteristic of various cancers. Recently, evidence for a convergence of these pathways at the level of the GLI2 transcription factor in the context of tumor initiation and progression to metastasis has emerged. This short review summarizes recent knowledge about GLI2 function and mechanisms of action downstream of TGF-ß in cancer.


Assuntos
Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias/patologia , Proteínas Nucleares/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Progressão da Doença , Feminino , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/secundário , Metástase Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Proteína Gli2 com Dedos de Zinco
19.
Pigment Cell Melanoma Res ; 24(5): 932-43, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21801332

RESUMO

We recently identified GLI2, the most active of GLI transcription factors, as a direct TGF-ß/SMAD target, whose expression in melanoma cells is associated with increased invasiveness and metastatic capacity. In this work, we provide evidence that high GLI2 expression is inversely correlated with that of the melanocyte-specific transcription factor M-microphthalmia transcription factor (M-MITF) and associated transcriptional program. GLI2-expressing cell lines were characterized by the loss of M-MITF-dependent melanocytic differentiation markers and reduced pigmentation. The balance between M-MITF and GLI2 expression did not correlate with the presence or absence of BRAF-activating mutations, but rather was controlled by two distinct pathways: the TGF-ß pathway, which favors GLI2 expression, and the protein kinase A (PKA)/cAMP pathway, which pushes the balance toward high M-MITF expression. Furthermore, overexpression and knockdown experiments demonstrated that GLI2 and M-MITF reciprocally repress each other's expression and control melanoma cell invasion in an opposite manner. These findings thus identify GLI2 as a critical transcription factor antagonizing M-MITF function to promote melanoma cell phenotypic plasticity and invasive behavior.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , Melanoma/genética , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fatores de Transcrição Kruppel-Like/genética , Melanoma/fisiopatologia , Camundongos , Camundongos Nus , Fator de Transcrição Associado à Microftalmia/genética , Invasividade Neoplásica , Proteínas Nucleares/genética , Pigmentação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Cutâneas/fisiopatologia , Proteínas Smad/metabolismo , Tela Subcutânea/efeitos dos fármacos , Tela Subcutânea/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína Gli2 com Dedos de Zinco
20.
Mol Cancer ; 10(1): 2, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-21211030

RESUMO

BACKGROUND: SKI and SnoN proteins have been shown to inhibit TGF-ß signaling, acting both as transcriptional co-repressors in the cell nucleus, and as sequestrators of SMAD proteins in the cytoplasm. TGF-ß, on the other hand, induces rapid, proteasome-mediated, degradation of both proteins. How elevated SKI and SnoN protein levels co-exist with active autocrine TGF-ß signaling in cancer cells is yet to be understood. RESULTS: In this study, we found elevated SKI and SnoN protein levels in a panel of melanoma cell lines, as compared to normal melanocytes. There was no correlation between SKI protein content and the capacity of melanoma cells to invade Matrigel™, to form subcutaneous tumors, or to metastasize to bone after intracardiac inoculation into nude mice. Nor did we find a correlation between SKI expression and histopathological staging of human melanoma. TGF-ß induced a rapid and dose-dependent degradation of SKI protein, associated with SMAD3/4 specific transcriptional response and induction of pro-metastatic target genes, partially prevented by pharmacologic blockade of proteasome activity. SKI knockdown in 1205Lu melanoma cells did not alter their invasive capacity or transcriptional responses to TGF-ß, and did not allow p21 expression in response to TGF-ß or reveal any growth inhibitory activity of TGF-ß. CONCLUSIONS: Despite high expression in melanoma cells, the role of SKI in melanoma remains elusive: SKI does not efficiently interfere with the pro-oncogenic activities of TGF-ß, unless stabilized by proteasome blockade. Its highly labile nature makes it an unlikely target for therapeutic intervention.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melanoma/patologia , Proteínas Proto-Oncogênicas/metabolismo , Neoplasias Cutâneas/patologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Humanos , Leupeptinas/farmacologia , Melanoma/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Inibidores de Proteassoma , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Neoplasias Cutâneas/metabolismo , Ativação Transcricional , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...