Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 15(1): 1870, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38467607

RESUMO

Myelin regeneration (remyelination) is essential to prevent neurodegeneration in demyelinating diseases such as Multiple Sclerosis, however, its efficiency declines with age. Regulatory T cells (Treg) recently emerged as critical players in tissue regeneration, including remyelination. However, the effect of ageing on Treg-mediated regenerative processes is poorly understood. Here, we show that expansion of aged Treg does not rescue age-associated remyelination impairment due to an intrinsically diminished capacity of aged Treg to promote oligodendrocyte differentiation and myelination in male and female mice. This decline in regenerative Treg functions can be rescued by a young environment. We identified Melanoma Cell Adhesion Molecule 1 (MCAM1) and Integrin alpha 2 (ITGA2) as candidates of Treg-mediated oligodendrocyte differentiation that decrease with age. Our findings demonstrate that ageing limits the neuroregenerative capacity of Treg, likely limiting their remyelinating therapeutic potential in aged patients, and describe two mechanisms implicated in Treg-driven remyelination that may be targetable to overcome this limitation.


Assuntos
Remielinização , Humanos , Masculino , Feminino , Camundongos , Animais , Idoso , Remielinização/fisiologia , Linfócitos T Reguladores/metabolismo , Oligodendroglia/fisiologia , Diferenciação Celular/fisiologia , Bainha de Mielina/metabolismo , Envelhecimento , Sistema Nervoso Central
2.
Int J Cancer ; 133(10): 2315-24, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23681936

RESUMO

The extracellular protease ADAMTS1 (A disintegrin and metalloprotease with thrombospondin repeats 1) has been described as an anti-angiogenic molecule and its role as a putative tumor protective molecule has also been suggested. Here, we have used a tumor xenograft model to determine the role of ADAMTS1 in tumor growth and angiogenesis. Increasing levels of the protease led to the complete inhibition of tumor growth. In an attempt to elucidate the mechanism of action of this protease, we focused our attention on its proteolytic activity on nidogens, one of the main components of the vascular basement membrane. The increased expression of ADAMTS1 was accompanied by increased proteolysis of nidogen-1 and -2 and their almost complete removal from vascular structures, together with major morphological alterations of tumor blood vessels and a decreased vessel density. The clinical relevance of this work is supported by our observations that ADAMTS1 expression is decreased in breast tumor specimens when compared with healthy tissue. Our studies also reveal that the cleavage of nidogen-1 and -2 is partially inhibited in human tumor samples. Moreover, the deposition of both nidogens surrounding vascular structures is drastically altered, implying a possible reduction in the maintenance of vessel integrity. Our studies reflect the requirement to explore the functional interactions between proteases and specific substrates in cancer biology.


Assuntos
Proteínas ADAM/metabolismo , Neoplasias da Mama/genética , Moléculas de Adesão Celular/metabolismo , Genes Supressores de Tumor , Glicoproteínas de Membrana/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS1 , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular/genética , Linhagem Celular , Regulação para Baixo , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/fisiologia , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteólise , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
3.
Oncogene ; 42(36): 2659-2672, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37516803

RESUMO

Melanoma is an aggressive form of skin cancer due to its high metastatic abilities and resistance to therapies. Melanoma cells reside in a heterogeneous tumour microenvironment that acts as a crucial regulator of its progression. Snail1 is an epithelial-to-mesenchymal transition transcription factor expressed during development and reactivated in pathological situations including fibrosis and cancer. In this work, we show that Snail1 is activated in the melanoma microenvironment, particularly in fibroblasts. Analysis of mouse models that allow stromal Snail1 depletion and therapeutic Snail1 blockade indicate that targeting Snail1 in the tumour microenvironment decreases melanoma growth and lung metastatic burden, extending mice survival. Transcriptomic analysis of melanoma-associated fibroblasts and analysis of the tumours indicate that stromal Snail1 induces melanoma growth by promoting an immunosuppressive microenvironment and a decrease in anti-tumour immunity. This study unveils a novel role of Snail1 in melanoma biology and supports its potential as a therapeutic target.


Assuntos
Melanoma , Neoplasias Cutâneas , Microambiente Tumoral , Animais , Camundongos , Transição Epitelial-Mesenquimal , Terapia de Imunossupressão , Melanoma/genética , Neoplasias Cutâneas/genética , Fatores de Transcrição da Família Snail/antagonistas & inibidores , Fatores de Transcrição da Família Snail/imunologia , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/genética
4.
Cancers (Basel) ; 15(9)2023 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-37173906

RESUMO

Melanoma is one of the deadliest forms of cancer. Most melanoma deaths are caused by distant metastases in several organs, especially the brain, the so-called melanoma brain metastases (MBMs). However, the precise mechanisms that sustain the growth of MBMs remain elusive. Recently, the excitatory neurotransmitter glutamate has been proposed as a brain-specific, pro-tumorigenic signal for various types of cancers, but how neuronal glutamate shuttling onto metastases is regulated remains unknown. Here, we show that the cannabinoid CB1 receptor (CB1R), a master regulator of glutamate output from nerve terminals, controls MBM proliferation. First, in silico transcriptomic analysis of cancer-genome atlases indicated an aberrant expression of glutamate receptors in human metastatic melanoma samples. Second, in vitro experiments conducted on three different melanoma cell lines showed that the selective blockade of glutamatergic NMDA receptors, but not AMPA or metabotropic receptors, reduces cell proliferation. Third, in vivo grafting of melanoma cells in the brain of mice selectively devoid of CB1Rs in glutamatergic neurons increased tumour cell proliferation in concert with NMDA receptor activation, whereas melanoma cell growth in other tissue locations was not affected. Taken together, our findings demonstrate an unprecedented regulatory role of neuronal CB1Rs in the MBM tumour microenvironment.

5.
Nat Metab ; 5(9): 1544-1562, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37563469

RESUMO

Resistance of melanoma to targeted therapy and immunotherapy is linked to metabolic rewiring. Here, we show that increased fatty acid oxidation (FAO) during prolonged BRAF inhibitor (BRAFi) treatment contributes to acquired therapy resistance in mice. Targeting FAO using the US Food and Drug Administration-approved and European Medicines Agency-approved anti-anginal drug ranolazine (RANO) delays tumour recurrence with acquired BRAFi resistance. Single-cell RNA-sequencing analysis reveals that RANO diminishes the abundance of the therapy-resistant NGFRhi neural crest stem cell subpopulation. Moreover, by rewiring the methionine salvage pathway, RANO enhances melanoma immunogenicity through increased antigen presentation and interferon signalling. Combination of RANO with anti-PD-L1 antibodies strongly improves survival by increasing antitumour immune responses. Altogether, we show that RANO increases the efficacy of targeted melanoma therapy through its effects on FAO and the methionine salvage pathway. Importantly, our study suggests that RANO could sensitize BRAFi-resistant tumours to immunotherapy. Since RANO has very mild side-effects, it might constitute a therapeutic option to improve the two main strategies currently used to treat metastatic melanoma.


Assuntos
Melanoma , Estados Unidos , Animais , Camundongos , Ranolazina/farmacologia , Ranolazina/uso terapêutico , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Imunoterapia , Inibidores de Proteínas Quinases/farmacologia , Metionina
6.
Methods Mol Biol ; 1731: 179-192, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29318554

RESUMO

The relevance of tumor vasculature has been extensively recognized, and it is still the focus of numerous lines of research for basic, translational, and clinical scientists. Indeed, the knowledge of some of its regulatory mechanisms has provoked the generation of ongoing cancer therapies. Within the context of the tumor microenvironment, the information that the analysis of the vasculature provides is very valuable, and it might reveal not just its quality and the response against a specific therapy but also its close relationship with neighboring stromal and tumor players.Studies during last decades already supported the contribution of extracellular proteases in neovascularization events, including ADAMTS. However, deeper analyses are still required to better understand the modulation of their proteolytic activity in the tumor microenvironment. Future studies will clearly benefit from existing and ongoing genetically modified mouse models.Here we emphasize the use of syngeneic models to study the vasculature during tumor progression, supported by their intact immunocompetent capacities and also by the range of possibilities to play with engineered mice and with modified tumor cells. Although various high-tech and sophisticated approaches have already been reported to evaluate tumor neovascularization, here we describe a simple and easily reproduced methodology based in the immunofluorescence detection of vascular-specific molecules. A final in silico analysis guarantees an unbiased quantification of tumor vasculature under different conditions.


Assuntos
Proteína ADAMTS1/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Neoplasias/patologia , Neovascularização Patológica/patologia , Proteína ADAMTS1/genética , Animais , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/genética , Software , Microambiente Tumoral
7.
Sci Rep ; 8(1): 13103, 2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30166561

RESUMO

Recent advances have emphasized the relevance of studying the extracellular microenvironment given its main contribution to tissue homeostasis and disease. Within this complex scenario, we have studied the extracellular protease ADAMTS1 (a disintegrin and metalloprotease with thrombospondin motif 1), implicated in vascularization and development, with reported anti- and pro-tumorigenic activities. In this work we performed a detailed study of the vasculature and substrates in adult organs of wild type and Adamts1-deficient mice. In addition to the expected alterations of organs like kidney, heart and aorta, we found that the lack of ADAMTS1 differently affects lymphocyte and myeloid populations in the spleen and bone marrow. The study of the substrate versican also revealed its alteration in the absence of the protease. With such premises, we challenged our mice with subcutaneous B16F1 syngeneic tumours and closely evaluated the immune repertoire in the tumours but also in the distant spleen and bone marrow. Our results confirmed a pro-inflammatory landscape in the absence of ADAMTS1, correlating with tumour blockade, supporting its novel role as a modulator of the immune cell response.


Assuntos
Proteína ADAMTS1/metabolismo , Inflamação/imunologia , Inflamação/patologia , Neoplasias/imunologia , Neoplasias/patologia , Proteína ADAMTS1/genética , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Proliferação de Células , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/metabolismo , Especificidade de Órgãos , Baço/metabolismo , Baço/patologia , Especificidade por Substrato , Versicanas/metabolismo
8.
Oncotarget ; 7(23): 34507-19, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27120788

RESUMO

The matrix metalloprotease ADAMTS1 (A Disintegrin And Metalloprotease with ThromboSpondin repeats 1) has been involved in tumorigenesis although its contributions appeared ambiguous. To understand the multifaceted actions of this protease, it is still required a deeper knowledge of its implication in heterogeneous tumor-stroma interactions. Using a syngeneic B16F1 melanoma model in wild type and ADAMTS1 knockout mice we found distinct stroma versus tumor functions for this protease. Genetic deletion of ADAMTS1 in the host microenvironment resulted in a drastic decrease of tumor growth and metastasis. However, the downregulation of tumor ADAMTS1 did not uncover relevant effects. Reduced tumors in ADAMTS1 KO mice displayed a paradoxical increase in vascular density and vascular-related genes; a detailed characterization revealed an impaired vasculature, along with a minor infiltration of macrophages. In addition, ex-vivo assays supported a chief role for ADAMTS1 in vascular sprouting, and melanoma xenografts showed a relevant induction of its expression in stroma compartments. These findings provide the first genetic evidence that supports the pro-tumorigenic role of stromal ADAMTS1.


Assuntos
Proteína ADAMTS1/genética , Melanoma Experimental/patologia , Melanoma/patologia , Neovascularização Patológica/patologia , Neoplasias Uveais/patologia , Proteína ADAMTS1/metabolismo , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo , Deleção de Genes , Células HEK293 , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Matrix Biol ; 44-46: 38-45, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25698314

RESUMO

ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) proteases comprise the most recently discovered branch of the extracellular metalloenzymes. Research during the last 15years, uncovered their association with a variety of physiological and pathological processes including blood coagulation, tissue repair, fertility, arthritis and cancer. Importantly, a frequent feature of ADAMTS enzymes relates to their effects on vascular-related phenomena, including angiogenesis. Their specific roles in vascular biology have been clarified by information on their expression profiles and substrate specificity. Through their catalytic activity, ADAMTS proteases modify rather than degrade extracellular proteins. They predominantly target proteoglycans and glycoproteins abundant in the basement membrane, therefore their broad contributions to the vasculature should not come as a surprise. Furthermore, in addition to their proteolytic functions, non-enzymatic roles for ADAMTS have also been identified expanding our understanding on the multiple activities of these enzymes in vascular-related processes.


Assuntos
Proteínas ADAM/química , Proteínas ADAM/metabolismo , Neovascularização Patológica , Neovascularização Fisiológica , Animais , Coagulação Sanguínea , Glicoproteínas/metabolismo , Humanos , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA