Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Neurobiol ; 59(2): 1214-1229, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34962634

RESUMO

Lafora disease (LD) is a fatal childhood-onset dementia characterized by the extensive accumulation of glycogen aggregates-the so-called Lafora Bodies (LBs)-in several organs. The accumulation of LBs in the brain underlies the neurological phenotype of the disease. LBs are composed of abnormal glycogen and various associated proteins, including p62, an autophagy adaptor that participates in the aggregation and clearance of misfolded proteins. To study the role of p62 in the formation of LBs and its participation in the pathology of LD, we generated a mouse model of the disease (malinKO) lacking p62. Deletion of p62 prevented LB accumulation in skeletal muscle and cardiac tissue. In the brain, the absence of p62 altered LB morphology and increased susceptibility to epilepsy. These results demonstrate that p62 participates in the formation of LBs and suggest that the sequestration of abnormal glycogen into LBs is a protective mechanism through which it reduces the deleterious consequences of its accumulation in the brain.


Assuntos
Doença de Lafora , Animais , Modelos Animais de Doenças , Glicogênio/metabolismo , Corpos de Inclusão/metabolismo , Doença de Lafora/genética , Camundongos , Camundongos Knockout , Proteína Sequestossoma-1
2.
Nat Commun ; 11(1): 6335, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33303745

RESUMO

Most breast cancers exhibit low immune infiltration and are unresponsive to immunotherapy. We hypothesized that inhibition of the receptor activator of nuclear factor-κB (RANK) signaling pathway may enhance immune activation. Here we report that loss of RANK signaling in mouse tumor cells increases leukocytes, lymphocytes, and CD8+ T cells, and reduces macrophage and neutrophil infiltration. CD8+ T cells mediate the attenuated tumor phenotype observed upon RANK loss, whereas neutrophils, supported by RANK-expressing tumor cells, induce immunosuppression. RANKL inhibition increases the anti-tumor effect of immunotherapies in breast cancer through a tumor cell mediated effect. Comparably, pre-operative single-agent denosumab in premenopausal early-stage breast cancer patients from the Phase-II D-BEYOND clinical trial (NCT01864798) is well tolerated, inhibits RANK pathway and increases tumor infiltrating lymphocytes and CD8+ T cells. Higher RANK signaling activation in tumors and serum RANKL levels at baseline predict these immune-modulatory effects. No changes in tumor cell proliferation (primary endpoint) or other secondary endpoints are observed. Overall, our preclinical and clinical findings reveal that tumor cells exploit RANK pathway as a mechanism to evade immune surveillance and support the use of RANK pathway inhibitors to prime luminal breast cancer for immunotherapy.


Assuntos
Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunidade , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Adulto , Animais , Neoplasias da Mama/sangue , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Denosumab/farmacologia , Denosumab/uso terapêutico , Feminino , Humanos , Terapia de Imunossupressão , Imunoterapia , Mediadores da Inflamação/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Modelos Biológicos , Células Mieloides/imunologia , Estadiamento de Neoplasias , Neutrófilos/imunologia , Ligante RANK/sangue , Ligante RANK/metabolismo
5.
Nat Commun ; 9(1): 4333, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30323191

RESUMO

The original version of this Article incorrectly gave a publication date of 8 October 2018; this should have been 28 September 2018. This has now been corrected in the PDF and HTML versions of the Article.

6.
Nat Commun ; 9(1): 3191, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30266909

RESUMO

Huntington's disease is a progressive neurodegenerative disorder caused by polyglutamine-expanded mutant huntingtin (mHTT). Here, we show that the deubiquitinase Usp12 rescues mHTT-mediated neurodegeneration in Huntington's disease rodent and patient-derived human neurons, and in Drosophila. The neuroprotective role of Usp12 may be specific amongst related deubiquitinases, as the closely related homolog Usp46 does not suppress mHTT-mediated toxicity. Mechanistically, we identify Usp12 as a potent inducer of neuronal autophagy. Usp12 overexpression accelerates autophagic flux and induces an approximately sixfold increase in autophagic structures as determined by ultrastructural analyses, while suppression of endogenous Usp12 slows autophagy. Surprisingly, the catalytic activity of Usp12 is not required to protect against neurodegeneration or induce autophagy. These findings identify the deubiquitinase Usp12 as a regulator of neuronal proteostasis and mHTT-mediated neurodegeneration.


Assuntos
Autofagia/genética , Neurônios/metabolismo , Neuroproteção/genética , Ubiquitina Tiolesterase/genética , Animais , Células Cultivadas , Drosophila melanogaster , Regulação da Expressão Gênica , Células HEK293 , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Mutação , Neurônios/citologia , Neurônios/ultraestrutura , Interferência de RNA , Ratos , Ubiquitina Tiolesterase/metabolismo
7.
Stem Cell Reports ; 8(5): 1392-1407, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28457887

RESUMO

Taxanes are a mainstay of treatment for breast cancer, but resistance often develops followed by metastatic disease and mortality. Aiming to reveal the mechanisms underlying taxane resistance, we used breast cancer patient-derived orthoxenografts (PDX). Mimicking clinical behavior, triple-negative breast tumors (TNBCs) from PDX models were more sensitive to docetaxel than luminal tumors, but they progressively acquired resistance upon continuous drug administration. Mechanistically, we found that a CD49f+ chemoresistant population with tumor-initiating ability is present in sensitive tumors and expands during the acquisition of drug resistance. In the absence of the drug, the resistant CD49f+ population shrinks and taxane sensitivity is restored. We describe a transcriptional signature of resistance, predictive of recurrent disease after chemotherapy in TNBC. Together, these findings identify a CD49f+ population enriched in tumor-initiating ability and chemoresistance properties and evidence a drug holiday effect on the acquired resistance to docetaxel in triple-negative breast cancer.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Integrina alfa6/metabolismo , Taxoides/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Células Cultivadas , Docetaxel , Feminino , Humanos , Integrina alfa6/genética , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/transplante , Taxoides/farmacologia , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Res ; 76(19): 5857-5869, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27480274

RESUMO

RANK expression is associated with poor prognosis in breast cancer even though its therapeutic potential remains unknown. RANKL and its receptor RANK are downstream effectors of the progesterone signaling pathway. However, RANK expression is enriched in hormone receptor negative adenocarcinomas, suggesting additional roles for RANK signaling beyond its hormone-dependent function. Here, to explore the role of RANK signaling once tumors have developed, we use the mouse mammary tumor virus-Polyoma Middle T (MMTV-PyMT), which mimics RANK and RANKL expression patterns seen in human breast adenocarcinomas. Complementary genetic and pharmacologic approaches demonstrate that therapeutic inhibition of RANK signaling drastically reduces the cancer stem cell pool, decreases tumor and metastasis initiation, and enhances sensitivity to chemotherapy. Mechanistically, genome-wide expression analyses show that anti-RANKL therapy promotes lactogenic differentiation of tumor cells. Moreover, RANK signaling in tumor cells negatively regulates the expression of Ap2 transcription factors, and enhances the Wnt agonist Rspo1 and the Sca1-population, enriched in tumor-initiating cells. In addition, we found that expression of TFAP2B and the RANK inhibitor, OPG, in human breast cancer correlate and are associated with relapse-free tumors. These results support the use of RANKL inhibitors to reduce recurrence and metastasis in breast cancer patients based on its ability to induce tumor cell differentiation. Cancer Res; 76(19); 5857-69. ©2016 AACR.


Assuntos
Neoplasias Mamárias Experimentais/prevenção & controle , Recidiva Local de Neoplasia/prevenção & controle , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Ataxina-1/análise , Diferenciação Celular/efeitos dos fármacos , Docetaxel , Feminino , Humanos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/efeitos dos fármacos , Ligante RANK/antagonistas & inibidores , Ligante RANK/farmacologia , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Taxoides/farmacologia , Fator de Transcrição AP-2/fisiologia
9.
Stem Cells ; 34(4): 1027-39, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26695351

RESUMO

Prolactin and progesterone both orchestrate the proliferation and differentiation of the mammary gland during gestation. Differentiation of milk secreting alveoli depends on the presence of prolactin receptor, the downstream Jak2-Stat5 pathway and the transcription factor Elf5. A strict regulation of Rank signaling is essential for the differentiation of the mammary gland and in particular for alveolar commitment. Impaired alveologenesis and lactation failure are observed in both, knockout and Rank overexpressing mice; however, the underlying molecular mechanism responsible for these phenotypes remains largely unknown. Using genome-wide expression analyses and functional studies, we show here that Rankl (RL) exposure leads to impaired secretory differentiation of alveolar cells not only in MMTV-RANK but also in wild-type (WT) mammary acini. Conversely, pharmacological blockage of Rank signaling at midgestation in WT mice leads to precocious and exacerbated lactogenesis. Mechanistically, RL negatively regulates Stat5 phosphorylation and Elf5 expression at the onset of lactogenesis. Continuous RL exposure leads to the expansion of basal and bipotent cells in WT and MMTV-RANK acini. Overall, we demonstrate that enhanced Rank signaling impairs secretory differentiation during pregnancy by inhibition of the prolactin/p-Stat5 pathway.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Prolactina/genética , Ligante RANK/genética , Fator de Transcrição STAT5/genética , Fatores de Transcrição/genética , Animais , Proliferação de Células/genética , Proteínas de Ligação a DNA/biossíntese , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Janus Quinase 2/biossíntese , Janus Quinase 2/genética , Lactação/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Knockout , Gravidez , Progesterona/genética , Progesterona/metabolismo , Prolactina/metabolismo , Ligante RANK/biossíntese , Fator de Transcrição STAT5/biossíntese , Transdução de Sinais , Fatores de Transcrição/biossíntese
10.
Stem Cells ; 31(9): 1954-65, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23766243

RESUMO

Receptor Activator of NF-kappa B (RANK) pathway controls mammary gland development in mice but its role in mammary stem cell fate remains undefined. We show that constitutive RANK signaling expands luminal and basal mammary compartments including mammary stem and luminal progenitor cell pools and interferes with the generation of CD61+ and Sca1+ luminal cells and Elf5 expression. Impaired mammary cell commitment upon RANK overexpression leads to the accumulation of progenitors including K14+K8+ bipotent cells and the formation of heterogeneous tumors containing hyperplastic basal, luminal, and progenitor cells. RANK expression increases in wild-type mammary epithelia with age and parity, and spontaneous preneoplastic lesions express RANK and accumulate K14+K8+ cells. In human breast tumors, high RANK expression levels are also associated with altered mammary differentiation. These results suggest that increased RANK signaling interferes with mammary cell commitment, contributing to breast carcinogenesis.


Assuntos
Carcinogênese/patologia , Linhagem da Célula , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Envelhecimento/patologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinogênese/genética , Compartimento Celular , Diferenciação Celular , Forma Celular , Epitélio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Queratinas/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Camundongos , Modelos Biológicos , Paridade , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Gravidez , Receptor Ativador de Fator Nuclear kappa-B/genética , Células-Tronco/metabolismo
11.
Genes Chromosomes Cancer ; 51(12): 1133-43, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22927297

RESUMO

Epigenetic alterations, such as CpG islands methylation and histone modifications, are recognized key characteristics of cancer. Glycogenes are a group of genes which epigenetic status was found to be changed in several tumors. In this study, we determined promoter methylation status of the glycogene beta-1,4-galactosyltransferase 1 (B4GALT1) in colorectal cancer patients. Methylation status of B4GALT1 was assessed in 130 colorectal adenocarcinomas, 13 adenomas, and in paired normal tissue using quantitative methylation specific PCR (QMSP). B4GALT1 mRNA expression was evaluated in methylated/unmethylated tumor and normal specimens. We also investigated microsatellite stability and microsatellite instability status and KRAS/BRAF mutations. Discriminatory power of QMSP was assessed by receiving operating curve (ROC) analysis on a training set of 24 colorectal cancers and paired mucosa. The area under the ROC curve (AUC) was 0.737 (95% confidence interval [CI]:0.591-0.881, P = 0.005) with an optimal cutoff value of 2.07 yielding a 54% sensitivity (95% CI: 35.1%-72.1%) and a specificity of 91.7% (95% CI: 74.1%-97.7%). These results were confirmed in an independent validation set where B4GALT1 methylation was detected in 52/106 patients. An inverse correlation was observed between methylation and B4GALT1 mRNA expression levels (r = -0.482, P = 0.037). Significant differences in methylation levels and frequencies was demonstrated in invasive lesions as compared with normal mucosa (P = 0.0001) and in carcinoma samples as compared with adenoma (P = 0.009). B4GALT1 methylation is a frequent and specific event in colorectal cancer and correlates with downregulation of mRNA expression. These results suggest that the glycogene B4GALT1 represent a valuable candidate biomarker of invasive phenotype of colorectal cancer.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Galactosiltransferases/genética , Regiões Promotoras Genéticas , Idoso , Metilação de DNA , Feminino , Galactosiltransferases/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Fenótipo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , RNA Mensageiro/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
12.
Cancer Res ; 72(11): 2879-88, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22496457

RESUMO

Paracrine signaling through receptor activator of NF-κB (RANK) pathway mediates the expansion of mammary epithelia that occurs during pregnancy, and activation of RANK pathway promotes mammary tumorigenesis in mice. In this study we extend these previous data to human cells and show that the RANK pathway promotes the development of mammary stem cells and breast cancer. Overexpression of RANK (FL-RANK) in a panel of tumoral and normal human mammary cells induces the expression of breast cancer stem and basal/stem cell markers. High levels of RANK in untransformed MCF10A cells induce changes associated with both stemness and transformation, including mammary gland reconstitution, epithelial-mesenchymal transition (EMT), increased migration, and anchorage-independent growth. In addition, spheroids of RANK overexpressing MCF10A cells display disrupted acinar formation, impair growth arrest and polarization, and luminal filling. RANK overexpression in tumor cells with nonfunctional BRCA1 enhances invasiveness in acinar cultures and increases tumorigenesis and metastasis in immunodeficient mice. High levels of RANK were found in human primary breast adenocarcinomas that lack expression of the hormone receptors, estrogen and progesterone, and in tumors with high pathologic grade and proliferation index; high RANK/RANKL expression was significantly associated with metastatic tumors. Together, our findings show that RANK promotes tumor initiation, progression, and metastasis in human mammary epithelial cells by increasing the population of CD44(+)CD24(-) cells, inducing stemness and EMT. These results suggest that RANK expression in primary breast cancer associates with poor prognosis.


Assuntos
Neoplasias da Mama/etiologia , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Animais , Proteína BRCA1/fisiologia , Neoplasias da Mama/patologia , Antígeno CD24/análise , Linhagem Celular Tumoral , Movimento Celular , Humanos , Receptores de Hialuronatos/análise , Camundongos , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Ligante RANK/análise , Receptor Ativador de Fator Nuclear kappa-B/análise
13.
Breast Cancer Res Treat ; 129(3): 947-54, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21541702

RESUMO

Intracellular signaling mediated by the receptor activator of nuclear factor-κB [Rank, encoded by the tumor necrosis factor receptor superfamily, member 11a (Tnfrsf11a) gene] is fundamental for mammary gland development in mice, regulating the expansion of stem and progenitor cell compartments. Conversely, Rank overexpression in mice promotes abnormal proliferation and impairs differentiation, leading to an increased incidence of tumorigenesis. Here, we show that a common genetic variant near the 5'-end of TNFRSF11A, rs7226991, is associated with breast cancer risk in the general population and among carriers of mutations in the breast cancer 2, early onset (BRCA2) gene. Akin to the results of the Cancer and Genetics Markers of Susceptibility initiative, combined analysis of rs7226991 in two Spanish case-control studies (1,365 controls and 1,323 cases in total) revealed a significant association with risk: odds ratio (OR) = 0.88, 95% confidence interval (CI) 0.78-0.98, P (trend) = 0.025. Subsequent examination of BRCA1 (n = 1,017) and BRCA2 (n = 885) mutation carriers revealed a consistent association in the latter group: weighted hazard ratio ((w)HR) = 0.70; 95% CI 0.55-0.88; and P (trend) = 0.003; compared to BRCA1 mutation carriers, (w)HR = 0.91; 95% CI 0.76-1.10; and P (trend) = 0.33. The results of this study need to be replicated in other populations and with larger numbers of BRCA1/2 mutation carriers.


Assuntos
Neoplasias da Mama/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Proteína BRCA1/genética , Proteína BRCA2/genética , Estudos de Coortes , Feminino , Predisposição Genética para Doença , Heterozigoto , Humanos , Mutação , Razão de Chances
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...