Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Br J Cancer ; 117(7): 947-953, 2017 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-28850564

RESUMO

BACKGROUND: Sunitinib is one of the first-line standard treatments for metastatic clear cell renal cell carcinoma (ccRCC) with a median time to progression shorter than 1 year. The objective is to discover predictive markers of response to adapt the treatment at diagnosis. METHODS: Prospective phase 2 multi-centre trials were conducted in ccRCC patients initiating sunitinib (54 patients) or bevacizumab (45 patients) in the first-line metastatic setting (SUVEGIL and TORAVA trials). The plasmatic level of CXCL7 at baseline was correlated with progression-free survival (PFS). RESULTS: The cut-off value of CXCL7 for PFS was 250 ng ml-1. Patients with CXCL7 plasmatic levels above the cut-off at baseline (250 ng ml-1) had a significantly longer PFS (hazard ratio 0.323 (95% confidence interval 0.147-0.707), P=0.001). These results were confirmed in a retrospective validation cohort. The levels of CXCL7 did not influence PFS of the bevacizumab-treated patients. CONCLUSIONS: CXCL7 may be considered as a predictive marker of sunitinib efficacy for ccRCC patients.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/sangue , Carcinoma de Células Renais/tratamento farmacológico , Indóis/uso terapêutico , Neoplasias Renais/sangue , Neoplasias Renais/tratamento farmacológico , Pirróis/uso terapêutico , beta-Tromboglobulina/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bevacizumab/administração & dosagem , Biomarcadores Tumorais/sangue , Carcinoma de Células Renais/secundário , Carcinoma de Células Renais/cirurgia , Intervalo Livre de Doença , Feminino , Humanos , Neoplasias Renais/patologia , Neoplasias Renais/cirurgia , Células Matadoras Naturais , Linfócitos do Interstício Tumoral , Macrófagos , Masculino , Camundongos , Pessoa de Meia-Idade , Gradação de Tumores , Transplante de Neoplasias , Nefrectomia , Neutrófilos , Estudos Prospectivos , Estudos Retrospectivos , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Sunitinibe , Taxa de Sobrevida
2.
Cell Stem Cell ; 29(5): 856-868.e5, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523143

RESUMO

After birth, the intestine undergoes major changes to shift from an immature proliferative state to a functional intestinal barrier. By combining inducible lineage tracing and transcriptomics in mouse models, we identify a prodifferentiation PDGFRαHigh intestinal stromal lineage originating from postnatal LTßR+ perivascular stromal progenitors. The genetic blockage of this lineage increased the intestinal stem cell pool while decreasing epithelial and immune maturation at weaning age, leading to reduced postnatal growth and dysregulated repair responses. Ablating PDGFRα in the LTBR stromal lineage demonstrates that PDGFRα has a major impact on the lineage fate and function, inducing a transcriptomic switch from prostemness genes, such as Rspo3 and Grem1, to prodifferentiation factors, including BMPs, retinoic acid, and laminins, and on spatial organization within the crypt-villus and repair responses. Our results show that the PDGFRα-induced transcriptomic switch in intestinal stromal cells is required in the first weeks after birth to coordinate postnatal intestinal maturation and function.


Assuntos
Intestinos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Diferenciação Celular/fisiologia , Mecanismos de Defesa , Mucosa Intestinal , Receptor beta de Linfotoxina , Camundongos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Células-Tronco
3.
Cancers (Basel) ; 13(10)2021 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-34065348

RESUMO

Signaling, proliferation, and inflammation are dependent on K63-linked ubiquitination-conjugation of a chain of ubiquitin molecules linked via lysine 63. However, very little information is currently available about how K63-linked ubiquitination is subverted in cancer. The present study provides, for the first time, evidence that cadmium (Cd), a widespread environmental carcinogen, is a potent activator of K63-linked ubiquitination, independently of oxidative damage, activation of ubiquitin ligase, or proteasome impairment. We show that Cd induces the formation of protein aggregates that sequester and inactivate cylindromatosis (CYLD) and selective autophagy, two tumor suppressors that deubiquitinate and degrade K63-ubiquitinated proteins, respectively. The aggregates are constituted of substrates of selective autophagy-SQSTM1, K63-ubiquitinated proteins, and mitochondria. These protein aggregates also cluster double-membrane remnants, which suggests an impairment in autophagosome maturation. However, failure to eliminate these selective cargos is not due to alterations in the general autophagy process, as degradation of long-lived proteins occurs normally. We propose that the simultaneous disruption of CYLD and selective autophagy by Cd feeds a vicious cycle that further amplifies K63-linked ubiquitination and downstream activation of the NF-κB pathway, processes that support cancer progression. These novel findings link together impairment of selective autophagy, K63-linked ubiquitination, and carcinogenesis.

4.
Commun Biol ; 4(1): 166, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33547392

RESUMO

Polo-like kinase 1 (Plk1) expression is inversely correlated with survival advantages in many cancers. However, molecular mechanisms that underlie Plk1 expression are poorly understood. Here, we uncover a hypoxia-regulated mechanism of Plk1-mediated cancer metastasis and drug resistance. We demonstrated that a HIF-2-dependent regulatory pathway drives Plk1 expression in clear cell renal cell carcinoma (ccRCC). Mechanistically, HIF-2 transcriptionally targets the hypoxia response element of the Plk1 promoter. In ccRCC patients, high expression of Plk1 was correlated to poor disease-free survival and overall survival. Loss-of-function of Plk1 in vivo markedly attenuated ccRCC growth and metastasis. High Plk1 expression conferred a resistant phenotype of ccRCC to targeted therapeutics such as sunitinib, in vitro, in vivo, and in metastatic ccRCC patients. Importantly, high Plk1 expression was defined in a subpopulation of ccRCC patients that are refractory to current therapies. Hence, we propose a therapeutic paradigm for improving outcomes of ccRCC patients.


Assuntos
Carcinoma de Células Renais , Proteínas de Ciclo Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Renais , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Estudos de Coortes , Embrião não Mamífero , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Metástase Neoplásica , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Regulação para Cima/genética , Peixe-Zebra , Quinase 1 Polo-Like
5.
Med Sci (Paris) ; 35(2): 132-137, 2019 Feb.
Artigo em Francês | MEDLINE | ID: mdl-30774080

RESUMO

The lymphatic system is made up of vessels that drain interstitial fluids throughout the body. The circulation of the lymph (liquid in the lymphatic system) in the lymphatic vessels is unidirectional: tissues to the lymph nodes and then to the veins. Ganglia are mechanical filters but also immune barriers that can block the progression of certain pathogens as well as cancer cells. However, most studies on the lymphatic system and cancer highlight the role of the lymphatic network in metastatic dissemination as tumor cells use this network to reach other organs. However, recent studies describe a beneficial role of the lymphatic system and of the vascular endothelial growth factor C (VEGF-C) which is one of the main factors responsible for the development of lymphatic vessels in cancer. In this review, we will illustrate this ambivalent and emerging role of VEGF-C and the lymphatic system in cancer aggressiveness.


Assuntos
Carcinogênese , Linfangiogênese , Metástase Neoplásica , Fator C de Crescimento do Endotélio Vascular/fisiologia , Animais , Carcinogênese/genética , Carcinogênese/imunologia , Humanos , Linfangiogênese/genética , Linfangiogênese/imunologia , Vasos Linfáticos/fisiologia , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Metástase Neoplásica/imunologia , Metástase Neoplásica/patologia , Evasão Tumoral/fisiologia
6.
Theranostics ; 9(4): 1181-1199, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30867824

RESUMO

Lysosomotropic agents such as sunitinib, lapatinib, and chloroquine belong to a drug family that is being used more frequently to treat advanced cancers. Sunitinib is standard care for metastatic renal cell carcinomas (mRCC) and lapatinib is used for trastuzumab/pertuzumab-refractory cancers. However, patients ineluctably relapse with a delay varying from a few months to a few years. To improve reactivity prior to relapse it is essential to identify the mechanisms leading to such variability. We showed previously that sunitinib became sequestered in lysosomes because of its basic pKa. Methods: Modifications to gene expression in response to sunitinib and in sunitinib resistant cells were analyzed by transcriptomic and proteomic analysis. ROS production was evaluated by FACS. Nuclear Factor kappa B (NFkB)-dependent transcriptional regulation of inflammatory gene expression was evaluated with a reporter gene. Correlation of CXCL5 with survival was analyzed with an online available data base (TCGA) and using a cohort of patients enrolled in the SUVEGIL clinical trial (NCT00943839). Results: We now show that sunitinib sequestration in lysosomes induced an incomplete autophagic process leading to activation of the NFkB inflammatory pathway. We defined a subset of inflammatory cytokines that were up-regulated by the drug either after an acute or chronic stimulus. One of the most up-regulated genes in sunitinib-resistant cells was the CXCL5 cytokine. CXCL5 was also induced in RCC by chloroquine and in a model of HER2 positive breast cancer cell lines after acute or chronic treatment with lapatinib. CXCL5 correlated to shorter survival in RCC and to the most aggressive forms of breast cancers. The levels of CXCL5 present in the plasma of patients treated with sunitinib were predictive of the efficacy of sunitinib but not of the VEGF-directed antibody bevacizumab. Conclusion: This translational study identified CXCL5 as a biomarker of efficacy of lysosomotropic drugs, a potential asset for personalized medicine.


Assuntos
Antineoplásicos/farmacologia , Autofagia , Neoplasias da Mama/tratamento farmacológico , Quimiocina CXCL5/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias Renais/tratamento farmacológico , Sunitinibe/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Humanos , Inflamação , Proteômica , Sunitinibe/uso terapêutico , Análise de Sobrevida
7.
Theranostics ; 9(3): 661-675, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809300

RESUMO

Hypoxic zones are common features of metastatic tumors. Due to inactivation of the von Hippel-Lindau gene (VHL), renal cell carcinomas (RCC) show constitutive stabilization of the alpha subunit of the hypoxia-inducible factor (HIF). Thus, RCC represents a model of chronic hypoxia. Development of the lymphatic network is dependent on vascular endothelial growth factor C (VEGFC) and lies at the front line of metastatic spreading. Here, we addressed the role of VEGFC in RCC aggressiveness and the regulation of its expression in hypoxia. Methods: Transcriptional and post transcriptional regulation of VEGFC expression was evaluated by qPCR and with reporter genes. The involvement of HIF was evaluated using a siRNA approach. Experimental RCC were performed with immuno-competent/deficient mice using human and mouse cells knocked-out for the VEGFC gene by a CRISPR/Cas9 method. The VEGFC axis was analyzed with an online available data base (TCGA) and using an independent cohort of patients. Results: Hypoxia induced VEGFC protein expression but down-regulated VEGFC gene transcription and mRNA stability. Increased proliferation, migration, over-activation of the AKT signaling pathway and enhanced expression of mesenchymal markers characterized VEGFC-/- cells. VEGFC-/- cells did not form tumors in immuno-deficient mice but developed aggressive tumors in immuno-competent mice. These tumors showed down-regulation of markers of activated lymphocytes and M1 macrophages, and up-regulation of M2 macrophages markers and programmed death ligand 1 (PDL1). Over-expression of lymphangiogenic genes including VEGFC was linked to increased disease-free and overall survival in patients with non-metastatic tumors, whereas its over-expression correlated with decreased progression-free and overall survival of metastatic patients. Conclusion: Our study revisited the admitted dogma linking VEGFC to tumor aggressiveness. We conclude that targeting VEGFC for therapy must be considered with caution.


Assuntos
Carcinoma de Células Renais/patologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Camundongos , Transplante de Neoplasias , Transplante Heterólogo
8.
Cancer Res ; 77(5): 1212-1226, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28087600

RESUMO

Sunitinib is an antiangiogenic therapy given as a first-line treatment for renal cell carcinoma (RCC). While treatment improves progression-free survival, most patients relapse. We hypothesized that patient relapse can stem from the development of a lymphatic network driven by the production of the main growth factor for lymphatic endothelial cells, VEGFC. In this study, we found that sunitinib can stimulate vegfc gene transcription and increase VEGFC mRNA half-life. In addition, sunitinib activated p38 MAPK, which resulted in the upregulation/activity of HuR and inactivation of tristetraprolin, two AU-rich element-binding proteins. Sunitinib stimulated a VEGFC-dependent development of lymphatic vessels in experimental tumors. This may explain our findings of increased lymph node invasion and new metastatic sites in 30% of sunitinib-treated patients and increased lymphatic vessels found in 70% of neoadjuvant treated patients. In summary, a therapy dedicated to destroying tumor blood vessels induced the development of lymphatic vessels, which may have contributed to the treatment failure. Cancer Res; 77(5); 1212-26. ©2017 AACR.


Assuntos
Inibidores da Angiogênese/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Indóis/farmacologia , Pirróis/farmacologia , Fator C de Crescimento do Endotélio Vascular/biossíntese , Inibidores da Angiogênese/efeitos adversos , Animais , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Feminino , Humanos , Indóis/efeitos adversos , Linfangiogênese/efeitos dos fármacos , Metástase Linfática , Camundongos , Camundongos Nus , Pirróis/efeitos adversos , Sunitinibe , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Curr Cancer Drug Targets ; 15(8): 684-702, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26452384

RESUMO

Survival rates of patients with metastatic or recurrent cancers have remained virtually unchanged during the past 30 years. This fact makes the need for new therapeutic options even more urgent. An attractive option would be to target autophagy, an essential quality control process that degrades toxic aggregates, damaged organelles, and signaling proteins, and acts as a tumor suppressor pathway of tumor initiation. Conversely, other fascinating observations suggest that autophagy supports cancer progression, relapse, metastasis, dormancy and resistance to therapy. This review provides an overview of the contradictory roles that autophagy plays in cancer initiation and progression and discusses the promises and challenges of current strategies that target autophagy for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Autofagia , Metástase Neoplásica/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias/tratamento farmacológico , Testes Imediatos , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Humanos , Metástase Neoplásica/patologia , Recidiva Local de Neoplasia/patologia
10.
Autophagy ; 10(2): 201-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24300375

RESUMO

Degradation of signaling proteins is one of the most powerful tumor-suppressive mechanisms by which a cell can control its own growth, its survival, and its motility. Emerging evidence suggests that autophagy limits several signaling pathways by degrading kinases, downstream components, and transcription factors; however, this often occurs under stressful conditions. Our recent studies revealed that constitutive autophagy temporally and spatially controls the RHOA pathway. Specifically, inhibition of autophagosome degradation induces the accumulation of the GTP-bound form of RHOA. The active RHOA is sequestered via SQSTM1/p62 within autolysosomes, and accordingly fails to localize to the spindle midbody or to the cell surface, as we demonstrate herein. As a result, all RHOA-downstream responses are deregulated, thus driving cytokinesis failure, aneuploidy and motility, three processes that directly have an impact upon cancer progression. We therefore propose that autophagy acts as a degradative brake for RHOA signaling and thereby controls cell proliferation, migration, and genome stability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia/fisiologia , Proliferação de Células , Proteínas de Choque Térmico/metabolismo , Transdução de Sinais/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Autofagia/genética , Movimento Celular/fisiologia , Células Cultivadas , Citocinese/genética , Citocinese/fisiologia , Camundongos , Fagossomos/metabolismo , Proteína Sequestossoma-1
11.
Autophagy ; 9(10): 1629-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24004837

RESUMO

A fundamental issue in cell biology is how the activation of a signaling pathway should lead to the appropriate cell response. Because of their oncogenic potential, the abundance, the duration and the localization of key signaling proteins must be carefully controlled. Negative feedback loops that combine transcription and protein-protein interactions are among the strategies by which a cell can turn off signaling. Our recent studies in Cancer Research and Autophagy show that degradation of key active proteins such as RHOA-GTP by constitutive autophagy represents one safeguard mechanism that limits signaling in a spatially and temporally restricted manner for faithful cytokinesis and directed migration. As a result, all autophagy compromises drive cytokinesis failure, aneuploidy, and motility-three processes that directly have an impact upon cancer progression. We therefore propose the term "signalphagy" to indicate a dedicated type of macroautophagy that degrades and thereby maintains the appropriate level of active signaling proteins to achieve tumor suppression.


Assuntos
Autofagia/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais/fisiologia , Animais , Movimento Celular/fisiologia , Citocinese/fisiologia , Humanos , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa