Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Prostate ; 75(3): 255-65, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25327758

RESUMO

BACKGROUND: Accumulating evidence suggests that chronic prostatic inflammation may lead to prostate cancer development. Growth differentiation factor-15 (GDF-15) is highly expressed in the prostate and has been associated with inflammation and tumorigenesis. METHODS: To examine the relationship between GDF-15 and prostatic inflammation, GDF-15 expression was measured by immunohistochemical (IHC) staining in human prostatectomy specimens containing inflammation. The relationship between GDF-15 and specific inflammatory cells was determined using non-biased computer image analysis. To provide insight into a potential suppressive role for GDF-15 in inflammation, activation of inflammatory mediator nuclear factor of kappa B (NFκB) was measured in PC3 cells. RESULTS: GDF-15 expression in luminal epithelial cells was decreased with increasing inflammation severity, suggesting an inverse association between GDF-15 and inflammation. Quantification of IHC staining by image analysis for GDF-15 and inflammatory cell markers revealed an inverse correlation between GDF-15 and CD3+, CD4+, CD8+, CD68+, and inos+ leukocytes. GDF-15 suppressed NFκB activity in luciferase reporter assays. Expression of the NFκB target, interleukin 8 (IL-8), was downregulated by GDF-15. CONCLUSIONS: The inverse relationship between GDF-15 and inflammation demonstrates a novel expression pattern for GDF-15 in the human prostate and suppression of NFκB activity may shed light on a potential mechanism for this inverse correlation.


Assuntos
Fator 15 de Diferenciação de Crescimento/metabolismo , NF-kappa B/metabolismo , Próstata/metabolismo , Prostatite/metabolismo , Antígenos CD/metabolismo , Atrofia/metabolismo , Atrofia/patologia , Humanos , Masculino , Próstata/patologia , Prostatite/patologia
2.
J Cell Biochem ; 114(6): 1424-33, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23280549

RESUMO

Growth differentiation factor-15 (GDF-15) and the CCN family member, connective tissue growth factor (CCN2), are associated with cardiac disease, inflammation, and cancer. The precise role and signaling mechanism for these factors in normal and diseased tissues remains elusive. Here we demonstrate an interaction between GDF-15 and CCN2 using yeast two-hybrid assays and have mapped the domain of interaction to the von Willebrand factor type C domain of CCN2. Biochemical pull down assays using secreted GDF-15 and His-tagged CCN2 produced in PC-3 prostate cancer cells confirmed a direct interaction between these proteins. To investigate the functional consequences of this interaction, in vitro angiogenesis assays were performed. We demonstrate that GDF-15 blocks CCN2-mediated tube formation in human umbilical vein endothelial (HUVEC) cells. To examine the molecular mechanism whereby GDF-15 inhibits CCN2-mediated angiogenesis, activation of αV ß3 integrins and focal adhesion kinase (FAK) was examined. CCN2-mediated FAK activation was inhibited by GDF-15 and was accompanied by a decrease in αV ß3 integrin clustering in HUVEC cells. These results demonstrate, for the first time, a novel signaling pathway for GDF-15 through interaction with the matricellular signaling molecule CCN2. Furthermore, antagonism of CCN2 mediated angiogenesis by GDF-15 may provide insight into the functional role of GDF-15 in disease states.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fator 15 de Diferenciação de Crescimento/fisiologia , Neovascularização Patológica/metabolismo , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Integrina alfaVbeta3/metabolismo , Neoplasias/irrigação sanguínea , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
3.
Nat Commun ; 11(1): 5079, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-33033234

RESUMO

Tumor heterogeneity and lack of knowledge about resistant cell states remain a barrier to targeted cancer therapies. Basal cell carcinomas (BCCs) depend on Hedgehog (Hh)/Gli signaling, but can develop mechanisms of Smoothened (SMO) inhibitor resistance. We previously identified a nuclear myocardin-related transcription factor (nMRTF) resistance pathway that amplifies noncanonical Gli1 activity, but characteristics and drivers of the nMRTF cell state remain unknown. Here, we use single cell RNA-sequencing of patient tumors to identify three prognostic surface markers (LYPD3, TACSTD2, and LY6D) which correlate with nMRTF and resistance to SMO inhibitors. The nMRTF cell state resembles transit-amplifying cells of the hair follicle matrix, with AP-1 and TGFß cooperativity driving nMRTF activation. JNK/AP-1 signaling commissions chromatin accessibility and Smad3 DNA binding leading to a transcriptional program of RhoGEFs that facilitate nMRTF activity. Importantly, small molecule AP-1 inhibitors selectively target LYPD3+/TACSTD2+/LY6D+ nMRTF human BCCs ex vivo, opening an avenue for improving combinatorial therapies.


Assuntos
Carcinoma Basocelular/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Cromatina/metabolismo , DNA de Neoplasias/metabolismo , Resistencia a Medicamentos Antineoplásicos , Matriz Extracelular/metabolismo , Ontologia Genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Folículo Piloso/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Proteína Smad3/metabolismo , Transativadores/metabolismo , Regulação para Cima
4.
J Invest Dermatol ; 139(7): 1439-1448, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30707899

RESUMO

Basal cell carcinomas (BCCs) rely on Hedgehog (HH) pathway growth signal amplification by the microtubule-based organelle, the primary cilium. Despite naive tumor responsiveness to Smoothened inhibitors (Smoi), resistance in advanced tumors remains common. Although the resistant BCCs usually maintain HH pathway activation, squamous cell carcinomas with Ras/MAPK pathway activation also arise, and the molecular basis of tumor type and pathway selection are still obscure. Here, we identify the primary cilium as a critical determinant controlling tumor pathway switching. Strikingly, Smoothened inhibitor-resistant BCCs have an increased mutational load in ciliome genes, resulting in reduced primary cilia and HH pathway activation compared with naive or Gorlin syndrome patient BCCs. Gene set enrichment analysis of resistant BCCs with a low HH pathway signature showed increased Ras/MAPK pathway activation. Tissue analysis confirmed an inverse relationship between primary cilia presence and Ras/MAPK activation, and primary cilia removal in BCCs potentiated Ras/MAPK pathway activation. Moreover, activating Ras in HH-responsive cell lines conferred resistance to both canonical (vismodegib) and noncanonical (atypical protein kinase C and MRTF inhibitors) HH pathway inhibitors and conferred sensitivity to MAPK inhibitors. Our results provide insights into BCC treatment and identify the primary cilium as an important lineage gatekeeper, preventing HH-to-Ras/MAPK pathway switching.


Assuntos
Síndrome do Nevo Basocelular/metabolismo , Carcinoma Basocelular/metabolismo , Cílios/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Hedgehog/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas ras/metabolismo , Anilidas/uso terapêutico , Antineoplásicos/uso terapêutico , Síndrome do Nevo Basocelular/genética , Síndrome do Nevo Basocelular/patologia , Carcinogênese , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Proteínas Hedgehog/antagonistas & inibidores , Humanos , Mutação/genética , Piridinas/uso terapêutico , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
5.
Nat Med ; 24(3): 271-281, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29400712

RESUMO

Hedgehog pathway-dependent cancers can escape Smoothened (SMO) inhibition through mutations in genes encoding canonical hedgehog pathway components; however, around 50% of drug-resistant basal cell carcinomas (BCCs) lack additional variants of these genes. Here we use multidimensional genomics analysis of human and mouse drug-resistant BCCs to identify a noncanonical hedgehog activation pathway driven by the transcription factor serum response factor (SRF). Active SRF along with its coactivator megakaryoblastic leukemia 1 (MKL1) binds DNA near hedgehog target genes and forms a previously unknown protein complex with the hedgehog transcription factor glioma-associated oncogene family zinc finger-1 (GLI1), causing amplification of GLI1 transcriptional activity. We show that cytoskeletal activation through Rho and the formin family member Diaphanous (mDia) is required for SRF-MKL-driven GLI1 activation and for tumor cell viability. Remarkably, nuclear MKL1 staining served as a biomarker in tumors from mice and human subjects to predict tumor responsiveness to MKL inhibitors, highlighting the therapeutic potential of targeting this pathway. Thus, our study illuminates, for the first time, cytoskeletal-activation-driven transcription as a personalized therapeutic target for combatting drug-resistant malignancies.


Assuntos
Carcinoma Basocelular/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Fator de Resposta Sérica/genética , Transativadores/genética , Proteína GLI1 em Dedos de Zinco/genética , Animais , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Proteínas Hedgehog , Humanos , Camundongos , Complexos Multiproteicos/genética , Transdução de Sinais , Ativação Transcricional
6.
Cell Stem Cell ; 20(2): 149-150, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28157493

RESUMO

Whether invasive tumor phenotypes like EMT arise from oncogenic drivers or from priming of the pre-tumor cell of origin remains unknown. In this issue of Cell Stem Cell, Latil et al. (2017) show that the pre-tumor niche establishes a chromatin state predisposing squamous cell carcinomas to undergo EMT and metastasis, suggesting that the pre-tumor epigenome has prognostic value.


Assuntos
Carcinoma de Células Escamosas , Solo , Epigenômica , Humanos , Prognóstico , Células-Tronco
7.
PLoS One ; 11(12): e0168031, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28030567

RESUMO

The Hedgehog pathway is a potent regulator of cellular growth and plays a central role in the development of many cancers including basal cell carcinoma (BCC). The majority of BCCs arise from mutations in the Patched receptor resulting in constitutive activation of the Hedgehog pathway. Secondary driver mutations promote BCC oncogenesis and occur frequently due to the high mutational burden resulting from sun exposure of the skin. Here, we uncover novel secondary mutations in Suppressor of Fused (SUFU), the major negative regulator of the Hedgehog pathway. SUFU normally binds to a Hedgehog transcriptional activator, GLI1, in order to prevent it from initiating transcription of Hedgehog target genes. We sequenced tumor-normal pairs from patients with early sporadic BCCs. This resulted in the discovery of nine mutations in SUFU, which were functionally investigated to determine whether they help drive BCC formation. Our results show that four of the SUFU mutations inappropriately activate the Hedgehog pathway, suggesting they may act as driver mutations for BCC development. Indeed, all four of the loss of function SUFU variants were found to disrupt its binding to GLI, leading to constitutive pathway activation. Our results from functional characterization of these mutations shed light on SUFU's role in Hedgehog signaling, tumor progression, and highlight a way in which BCCs can arise.


Assuntos
Carcinoma Basocelular/metabolismo , Proteínas Hedgehog/metabolismo , Mutação , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Animais , Carcinogênese , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Proliferação de Células , Progressão da Doença , Células HEK293 , Humanos , Camundongos , Modelos Moleculares , Células NIH 3T3 , Conformação Proteica , Proteínas Repressoras/química , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Adulto Jovem , Proteína GLI1 em Dedos de Zinco/metabolismo
8.
Cancer Cell ; 27(3): 342-53, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25759020

RESUMO

Advanced basal cell carcinomas (BCCs) frequently acquire resistance to Smoothened (SMO) inhibitors through unknown mechanisms. Here we identify SMO mutations in 50% (22 of 44) of resistant BCCs and show that these mutations maintain Hedgehog signaling in the presence of SMO inhibitors. Alterations include four ligand binding pocket mutations defining sites of inhibitor binding and four variants conferring constitutive activity and inhibitor resistance, illuminating pivotal residues that ensure receptor autoinhibition. In the presence of a SMO inhibitor, tumor cells containing either class of SMO mutants effectively outcompete cells containing the wild-type SMO. Finally, we show that both classes of SMO variants respond to aPKC-ι/λ or GLI2 inhibitors that operate downstream of SMO, setting the stage for the clinical use of GLI antagonists.


Assuntos
Anilidas/uso terapêutico , Carcinoma Basocelular/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Piridinas/uso terapêutico , Receptores Acoplados a Proteínas G/genética , Neoplasias Cutâneas/tratamento farmacológico , Anilidas/química , Sítios de Ligação , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Análise Mutacional de DNA , Exoma , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Modelos Moleculares , Mutação , Estrutura Terciária de Proteína , Piridinas/química , Receptores Acoplados a Proteínas G/química , Análise de Sequência de DNA , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Receptor Smoothened
9.
Cancer Res ; 75(17): 3623-35, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26130651

RESUMO

Aberrant Shh signaling promotes tumor growth in diverse cancers. The importance of Shh signaling is particularly evident in medulloblastoma and basal cell carcinoma (BCC), where inhibitors targeting the Shh pathway component Smoothened (Smo) show great therapeutic promise. However, the emergence of drug resistance limits long-term efficacy, and the mechanisms of resistance remain poorly understood. Using new medulloblastoma models, we identify two distinct paradigms of resistance to Smo inhibition. Sufu mutations lead to maintenance of the Shh pathway in the presence of Smo inhibitors. Alternatively activation of the RAS-MAPK pathway circumvents Shh pathway dependency, drives tumor growth, and enhances metastatic behavior. Strikingly, in BCC patients treated with Smo inhibitor, squamous cell cancers with RAS/MAPK activation emerged from the antecedent BCC tumors. Together, these findings reveal a critical role of the RAS-MAPK pathway in drug resistance and tumor evolution of Shh pathway-dependent tumors.


Assuntos
Carcinoma Basocelular/genética , Proteínas Hedgehog/genética , Meduloblastoma/genética , Receptores Acoplados a Proteínas G/biossíntese , Proteínas ras/genética , Anilidas/administração & dosagem , Animais , Carcinoma Basocelular/tratamento farmacológico , Carcinoma Basocelular/patologia , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/patologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Piridinas/administração & dosagem , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/biossíntese
10.
Cold Spring Harb Perspect Med ; 4(7): a013581, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24985127

RESUMO

Basal cell carcinomas (BCCs) are very common epithelial cancers that depend on the Hedgehog pathway for tumor growth. Traditional therapies such as surgical excision are effective for most patients with sporadic BCC; however, better treatment options are needed for cosmetically sensitive or advanced and metastatic BCC. The first approved Hedgehog antagonist targeting the membrane receptor Smoothened, vismodegib, shows remarkable effectiveness on both syndromic and nonsyndromic BCCs. However, drug-resistant tumors frequently develop, illustrating the need for the development of next-generation Hedgehog antagonists targeting pathway components downstream from Smoothened. In this article, we will summarize available BCC treatment options and discuss the development of next-generation antagonists.


Assuntos
Anilidas/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma Basocelular/tratamento farmacológico , Proteínas Hedgehog/antagonistas & inibidores , Piridinas/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Descoberta de Drogas , Resistencia a Medicamentos Antineoplásicos , Humanos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptor Smoothened , Tecnologia Farmacêutica/tendências , Resultado do Tratamento
11.
Nat Med ; 20(7): 732-40, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24973920

RESUMO

Hedgehog signaling drives oncogenesis in several cancers, and strategies targeting this pathway have been developed, most notably through inhibition of Smoothened (SMO). However, resistance to Smoothened inhibitors occurs by genetic changes of Smoothened or other downstream Hedgehog components. Here we overcome these resistance mechanisms by modulating GLI transcription through inhibition of bromo and extra C-terminal (BET) bromodomain proteins. We show that BRD4 and other BET bromodomain proteins regulate GLI transcription downstream of SMO and suppressor of fused (SUFU), and chromatin immunoprecipitation studies reveal that BRD4 directly occupies GLI1 and GLI2 promoters, with a substantial decrease in engagement of these sites after treatment with JQ1, a small-molecule inhibitor targeting BRD4. Globally, genes associated with medulloblastoma-specific GLI1 binding sites are downregulated in response to JQ1 treatment, supporting direct regulation of GLI activity by BRD4. Notably, patient- and GEMM (genetically engineered mouse model)-derived Hedgehog-driven tumors (basal cell carcinoma, medulloblastoma and atypical teratoid rhabdoid tumor) respond to JQ1 even when harboring genetic lesions rendering them resistant to Smoothened antagonists. Altogether, our results reveal BET proteins as critical regulators of Hedgehog pathway transcriptional output and nominate BET bromodomain inhibitors as a strategy for treating Hedgehog-driven tumors with emerged or a priori resistance to Smoothened antagonists.


Assuntos
Epigênese Genética , Proteínas Hedgehog/genética , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , Animais , Azepinas/farmacologia , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Ligantes , Camundongos , Neoplasias Experimentais/patologia , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
12.
J Invest Dermatol ; 133(5): 1131-3, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23594533

RESUMO

The tumor suppressor Patched1 (Ptch1) possesses well-described roles in regulating sonic hedgehog (SHH) signaling in the skin and preventing the formation of basal cell carcinomas (BCCs). In this issue, Kang et al. extend their previous work to show that a naturally occurring allele of Ptch1 found in FVB mice promotes early squamous cell carcinoma (SCC) growth without aberrant activation of the SHH pathway. The study reveals new roles for Ptch1 that lie at the nexus between BCC and SCC formation.


Assuntos
Carcinoma de Células Escamosas/fisiopatologia , Desenvolvimento Fetal/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Queratina-14/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Neoplasias Cutâneas/fisiopatologia , Animais , Receptores Patched , Receptor Patched-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA