Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Semin Cancer Biol ; 78: 104-123, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33979673

RESUMO

Disseminated tumor cells (DTCs) spread systemically yet distinct patterns of metastasis indicate a range of tissue susceptibility to metastatic colonization. Distinctions between permissive and suppressive tissues are still being elucidated at cellular and molecular levels. Although there is a growing appreciation for the role of the microenvironment in regulating metastatic success, we have a limited understanding of how diverse tissues regulate DTC dormancy, the state of reversible quiescence and subsequent awakening thought to contribute to delayed relapse. Several themes of microenvironmental regulation of dormancy are beginning to emerge, including vascular association, co-option of pre-existing niches, metabolic adaptation, and immune evasion, with tissue-specific nuances. Conversely, DTC awakening is often associated with injury or inflammation-induced activation of the stroma, promoting a proliferative environment with DTCs following suit. We review what is known about tissue-specific regulation of tumor dormancy on a tissue-by-tissue basis, profiling major metastatic organs including the bone, lung, brain, liver, and lymph node. An aerial view of the barriers to metastatic growth may reveal common targets and dependencies to inform the therapeutic prevention of relapse.


Assuntos
Suscetibilidade a Doenças , Neoplasias/etiologia , Neoplasias/patologia , Microambiente Tumoral , Animais , Gerenciamento Clínico , Humanos , Recidiva
2.
Nature ; 527(7578): 329-35, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26524530

RESUMO

Ever since Stephen Paget's 1889 hypothesis, metastatic organotropism has remained one of cancer's greatest mysteries. Here we demonstrate that exosomes from mouse and human lung-, liver- and brain-tropic tumour cells fuse preferentially with resident cells at their predicted destination, namely lung fibroblasts and epithelial cells, liver Kupffer cells and brain endothelial cells. We show that tumour-derived exosomes uptaken by organ-specific cells prepare the pre-metastatic niche. Treatment with exosomes from lung-tropic models redirected the metastasis of bone-tropic tumour cells. Exosome proteomics revealed distinct integrin expression patterns, in which the exosomal integrins α6ß4 and α6ß1 were associated with lung metastasis, while exosomal integrin αvß5 was linked to liver metastasis. Targeting the integrins α6ß4 and αvß5 decreased exosome uptake, as well as lung and liver metastasis, respectively. We demonstrate that exosome integrin uptake by resident cells activates Src phosphorylation and pro-inflammatory S100 gene expression. Finally, our clinical data indicate that exosomal integrins could be used to predict organ-specific metastasis.


Assuntos
Encéfalo/metabolismo , Exossomos/metabolismo , Integrinas/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Metástase Neoplásica/patologia , Metástase Neoplásica/prevenção & controle , Tropismo , Animais , Biomarcadores/metabolismo , Encéfalo/citologia , Linhagem Celular Tumoral , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes src , Humanos , Integrina alfa6beta1/metabolismo , Integrina alfa6beta4/antagonistas & inibidores , Integrina alfa6beta4/metabolismo , Cadeias beta de Integrinas/metabolismo , Integrina beta4/metabolismo , Integrinas/antagonistas & inibidores , Células de Kupffer/citologia , Células de Kupffer/metabolismo , Fígado/citologia , Pulmão/citologia , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Fosforilação , Receptores de Vitronectina/antagonistas & inibidores , Receptores de Vitronectina/metabolismo , Proteínas S100/genética
3.
Adv Exp Med Biol ; 1100: 19-45, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30411258

RESUMO

Anti-angiogenic therapy was conceived originally as a silver bullet able to maintain tumor dormancy indefinitely. By targeting new blood vessel formation, anti-angiogenic agents were expected to suppress the growth of any type of primary or metastatic tumor, independent of their subtype or genetic landscape. However, more that 20 years after the first anti-angiogenic preclinical trial, the astonishing inhibition of metastatic outgrowth originally observed in mouse models never translated into clinics. Indeed, whereas anti-angiogenic agents (sometimes) prolong progression-free survival, they fail to impact overall survival, particularly in breast cancer. This observation revealed to be true in early- and advanced-stage breast cancer patients treated either in adjuvant or neo-adjuvant settings, suggesting that the effect of anti-angiogenic therapy on repressing growth of overt metastases - and also on preventing outgrowth of disseminated tumor cells and micrometastases - is limited. What are the reasons underlying this failure? And, more importantly, is there still room for improvement?


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias da Mama/tratamento farmacológico , Recidiva Local de Neoplasia/prevenção & controle , Animais , Humanos , Imunoterapia , Camundongos , Metástase Neoplásica/prevenção & controle , Neovascularização Patológica/prevenção & controle , Falha de Tratamento
4.
Nature ; 540(7634): 528-529, 2016 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-27974802
5.
Development ; 140(2): 343-52, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23250208

RESUMO

Epithelial cell invasion through the extracellular matrix (ECM) is a crucial step in branching morphogenesis. The mechanisms by which the mammary epithelium integrates cues from the ECM with intracellular signaling in order to coordinate invasion through the stroma to make the mammary tree are poorly understood. Because the cell membrane-bound matrix metalloproteinase Mmp14 is known to play a key role in cancer cell invasion, we hypothesized that it could also be centrally involved in integrating signals for mammary epithelial cells (MECs) to navigate the collagen 1 (CL-1)-rich stroma of the mammary gland. Expression studies in nulliparous mice that carry a NLS-lacZ transgene downstream of the Mmp14 promoter revealed that Mmp14 is expressed in MECs at the tips of the branches. Using both mammary organoids and 3D organotypic cultures, we show that MMP activity is necessary for invasion through dense CL-1 (3 mg/ml) gels, but dispensable for MEC branching in sparse CL-1 (1 mg/ml) gels. Surprisingly, however, Mmp14 without its catalytic activity was still necessary for branching. Silencing Mmp14 prevented cell invasion through CL-1 and disrupted branching altogether; it also reduced integrin ß1 (Itgb1) levels and attenuated MAPK signaling, disrupting Itgb1-dependent invasion/branching within CL-1 gels. FRET imaging revealed that Mmp14 associates directly with Itgb1. We identified a domain of Mmp14 that is required for modulating the levels of Itgb1, MEC signaling and the rate of invasion within CL-1. These results shed light on hitherto undescribed non-proteolytic activities of Mmp14 that are necessary for the Itgb1-dependent biochemical and mechanical signals that regulate branching in the mammary epithelium.


Assuntos
Membrana Celular/metabolismo , Integrina beta1/metabolismo , Sistema de Sinalização das MAP Quinases , Metaloproteinase 14 da Matriz/metabolismo , Animais , Domínio Catalítico , Colágeno/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Inativação Gênica , Lentivirus/metabolismo , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Transgenes
6.
Trends Cancer ; 10(4): 275-276, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38521653

RESUMO

Seeking to define early events that regulate disseminated tumor cell (DTC) fate upon their arrival to the lung, Jakab et al. reach the surprising conclusion that dormancy is determined by a cell autonomous poised epigenetic state that renders DTCs responsive to angiocrine Wnt signaling.


Assuntos
Epigênese Genética , Humanos
7.
Cancer Cell ; 42(1): 119-134.e12, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-38194912

RESUMO

The period between "successful" treatment of localized breast cancer and the onset of distant metastasis can last many years, representing an unexploited window to eradicate disseminated disease and prevent metastases. We find that the source of recurrence-disseminated tumor cells (DTCs) -evade endogenous immunity directed against tumor neoantigens. Although DTCs downregulate major histocompatibility complex I, this does not preclude recognition by conventional T cells. Instead, the scarcity of interactions between two relatively rare populations-DTCs and endogenous antigen-specific T cells-underlies DTC persistence. This scarcity is overcome by any one of three immunotherapies that increase the number of tumor-specific T cells: T cell-based vaccination, or adoptive transfer of T cell receptor or chimeric antigen receptor T cells. Each approach achieves robust DTC elimination, motivating discovery of MHC-restricted and -unrestricted DTC antigens that can be targeted with T cell-based immunotherapies to eliminate the reservoir of metastasis-initiating cells in patients.


Assuntos
Neoplasias da Mama , Linfócitos T , Humanos , Feminino , Evasão da Resposta Imune , Transferência Adotiva , Neoplasias da Mama/terapia , Imunoterapia
8.
Am J Pathol ; 180(6): 2249-56, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22542846

RESUMO

Intact organ structure is essential in maintaining tissue specificity and cellular differentiation. Small physiological or genetic variations lead to changes in microanatomy that, if persistent, could have functional consequences and may easily be masked by the heterogeneity of tissue anatomy. Current imaging techniques rely on histological, two-dimensional sections requiring sample manipulation that are essentially two dimensional. We have developed a method for three-dimensional imaging of whole-mount, unsectioned mammalian tissues to elucidate subtle and detailed micro- and macroanatomies in adult organs and embryos. We analyzed intact or dissected organ whole mounts with laser scanning-based tissue autofluorescence/fluorescence imaging (LS-TAFI). We obtained clear visualization of microstructures within murine mammary glands and mammary tumors and other organs without the use of immunostaining and without probes or fluorescent reporter genes. Combining autofluorescence with reflected light signals from chromophore-stained tissues allowed identification of individual cells within three-dimensional structures of whole-mounted organs. This technique could be useful for rapid diagnosis of human clinical samples and possibly the effect of subtle variations such as low dose radiation.


Assuntos
Imageamento Tridimensional/métodos , Lasers , Glândulas Mamárias Animais/anatomia & histologia , Neoplasias Mamárias Experimentais/patologia , Animais , Corantes , Feminino , Fluorescência , Pulmão/anatomia & histologia , Glândulas Mamárias Animais/embriologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Confocal/métodos , Pâncreas/anatomia & histologia , beta-Galactosidase
9.
Immunohorizons ; 7(1): 64-70, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36637515

RESUMO

The human ubiquitin C promoter (UBC)-driven GFP-transgenic mouse (UBC-GFP) transgene integration site was mapped recently to chromosome 17, linked closely to the MHC locus. In this study, we demonstrate a functional consequence of this insertion site in the backcrossed UBC-GFP BALB/c congenic strain [CByJ.B6-Tg(UBC-GFP) 30Scha/J]: rejection of transplanted "syngeneic" 4T1 mammary tumor cells. Rejection of BALB/c-derived 4T1 cells is in all likelihood a consequence of MHC mismatch due to stable inheritance of C57BL/6-derived H-2b (rather than prototypical H-2d) by the BALB/c UBC-GFP strain. These data are a valuable resource to researchers who have previously employed the UBC-GFP congenic strain for attempted syngeneic MHC-matched and allogenic MHC-mismatched studies, as their data likely require reinterpretation. Further, this study reemphasizes the impact of mapping transgene integration sites of commonly used mouse strains as a way of increasing scientific rigor and reproducibility.


Assuntos
Alelos , Camundongos , Animais , Humanos , Reprodutibilidade dos Testes , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
10.
Mol Oncol ; 16(1): 130-147, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34058066

RESUMO

Dormant, disseminated tumor cells (DTCs) are thought to be the source of breast cancer metastases several years or even decades after initial treatment. To date, a selective therapy that leads to their elimination has not been discovered. While dormant DTCs resist chemotherapy, evidence suggests that this resistance is driven not by their lack of proliferation, but by their engagement of the surrounding microenvironment, via integrin-ß1-mediated interactions. Because integrin-ß1-targeted agents have not been translated readily to the clinic, signaling nodes downstream of integrin-ß1 could serve as attractive therapeutic targets in order to sensitize dormant DTCs to therapy. By probing a number of kinases downstream of integrin-ß1, we determined that PI3K inhibition with either a tool compounds or a compound (PF-05212384; aka Gedatolisib) in clinical trials robustly sensitizes quiescent breast tumor cells seeded in organotypic bone marrow cultures to chemotherapy. These results motivated the preclinical study of whether Gedatolisib-with or without genotoxic therapy-would reduce DTC burden and prevent metastases. Despite promising results in organotypic culture, Gedatolisib failed to reduce DTC burden or delay, reduce or prevent metastasis in murine models of either triple-negative or estrogen receptor-positive breast cancer dissemination and metastasis. This result held true whether analyzing Gedatolisib on its own (vs. vehicle-treated animals) or in combination with dose-dense doxorubicin and cyclophosphamide (vs. animals treated only with dose-dense chemotherapies). These data suggest that PI3K is not the node downstream of integrin-ß1 that confers chemotherapeutic resistance to DTCs. More broadly, they cast doubt on the strategy to target PI3K in order to eliminate DTCs and prevent breast cancer metastasis.


Assuntos
Neoplasias da Mama , Fosfatidilinositol 3-Quinases , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Feminino , Humanos , Integrinas , Camundongos , Morfolinas , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Serina-Treonina Quinases TOR , Triazinas , Microambiente Tumoral
11.
Nat Cell Biol ; 24(4): 538-553, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35411081

RESUMO

Skeletal muscle has long been recognized as an inhospitable site for disseminated tumour cells (DTCs). Yet its antimetastatic nature has eluded a thorough mechanistic examination. Here, we show that DTCs traffic to and persist within skeletal muscle in mice and in humans, which raises the question of how this tissue suppresses colonization. Results from mouse and organotypic culture models along with metabolomic profiling suggested that skeletal muscle imposes a sustained oxidative stress on DTCs that impairs their proliferation. Functional studies demonstrated that disrupting reduction-oxidation homeostasis via chemogenetic induction of reactive oxygen species slowed proliferation in a more fertile organ: the lung. Conversely, enhancement of the antioxidant potential of tumour cells through ectopic expression of catalase in the tumour or host mitochondria allowed robust colonization of skeletal muscle. These findings reveal a profound metabolic bottleneck imposed on DTCs and sustained by skeletal muscle. A thorough understanding of this biology could reveal previously undocumented DTC vulnerabilities that can be exploited to prevent metastasis in other more susceptible tissues.


Assuntos
Neoplasias , Estresse Oxidativo , Animais , Camundongos , Músculo Esquelético/metabolismo , Neoplasias/metabolismo , Oxirredução , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo
12.
Cancer Cell ; 40(8): 787-791, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35944497

RESUMO

Metastasis, the major cause of cancer death, represents one of the major challenges in oncology. Scientists are still trying to understand the biological basis underlying the dissemination and outgrowth of tumor cells, why these cells can remain dormant for years, how they become resistant to the immune system or cytotoxic effects of systemic therapy, and how they interact with their new microenvironment. We asked experts to discuss some of the unknowns, advances, and areas of opportunity related to cancer metastasis.


Assuntos
Neoplasias , Microambiente Tumoral , Humanos , Sistema Imunitário/patologia , Metástase Neoplásica/patologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia
13.
Nat Cancer ; 3(1): 25-42, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35121993

RESUMO

Although dormancy is thought to play a key role in the metastasis of breast tumor cells to the brain, our knowledge of the molecular mechanisms regulating disseminated tumor cell (DTC) dormancy in this organ is limited. Here using serial intravital imaging of dormant and metastatic triple-negative breast cancer lines, we identify escape from the single-cell or micrometastatic state as the rate-limiting step towards brain metastasis. We show that every DTC occupies a vascular niche, with quiescent DTCs residing on astrocyte endfeet. At these sites, astrocyte-deposited laminin-211 drives DTC quiescence by inducing the dystroglycan receptor to associate with yes-associated protein, thereby sequestering it from the nucleus and preventing its prometastatic functions. These findings identify a brain-specific mechanism of DTC dormancy and highlight the need for a more thorough understanding of tumor dormancy to develop therapeutic approaches that prevent brain metastasis.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Astrócitos/metabolismo , Encéfalo/metabolismo , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Laminina/metabolismo , Microambiente Tumoral
14.
Elife ; 112022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36511483

RESUMO

Advanced prostate malignancies are a leading cause of cancer-related deaths in men, in large part due to our incomplete understanding of cellular drivers of disease progression. We investigate prostate cancer cell dynamics at single-cell resolution from disease onset to the development of androgen independence in an in vivo murine model. We observe an expansion of a castration-resistant intermediate luminal cell type that correlates with treatment resistance and poor prognosis in human patients. Moreover, transformed epithelial cells and associated fibroblasts create a microenvironment conducive to pro-tumorigenic immune infiltration, which is partially androgen responsive. Androgen-independent prostate cancer leads to significant diversification of intermediate luminal cell populations characterized by a range of androgen signaling activity, which is inversely correlated with proliferation and mRNA translation. Accordingly, distinct epithelial populations are exquisitely sensitive to translation inhibition, which leads to epithelial cell death, loss of pro-tumorigenic signaling, and decreased tumor heterogeneity. Our findings reveal a complex tumor environment largely dominated by castration-resistant luminal cells and immunosuppressive infiltrates.


Assuntos
Androgênios , Neoplasias da Próstata , Masculino , Humanos , Camundongos , Animais , Próstata/metabolismo , Neoplasias da Próstata/patologia , Orquiectomia , Dinâmica Populacional , Receptores Androgênicos/metabolismo , Progressão da Doença , Microambiente Tumoral
15.
Exp Cell Res ; 316(5): 813-25, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20067788

RESUMO

During angiogenesis, endothelial cells (ECs) degrade their surrounding extracellular matrix (ECM) to facilitate invasion. How interactions between ECs and other cells within their microenvironment facilitate this process is only partially understood. We have utilized a tractable 3D co-culture model to investigate the proteolytic mechanisms by which pre-committed or more highly committed mesenchymal cells stimulate capillary formation. On their own, ECs invade their surrounding matrix, but do not form capillaries. However, in the presence of either mesenchymal stem cells (MSCs) or fibroblasts, ECs form polarized, tubular structures that are intimately associated with mesenchymal cells. Further, ECs up-regulate gene expression of several extracellular proteases upon co-culture with either mesenchymal cell type. The administration of both broad spectrum and specific protease inhibitors demonstrated that MSC-stimulated capillary formation relied solely on membrane-type matrix metalloproteinases (MT-MMPs) while fibroblast-mediated sprouting proceeded independent of MMP inhibition unless the plasminogen activator/plasmin axis was inhibited in concert. While other studies have established a role for the ECM itself in dictating proteolysis and matrix degradation during capillary morphogenesis, the present study illustrates that heterotypic cellular interactions within the microenvironment can direct the proteolytic mechanisms required for capillary formation.


Assuntos
Capilares/crescimento & desenvolvimento , Mesoderma/citologia , Morfogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Capilares/anatomia & histologia , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Hidrólise , Inibidores de Proteases/metabolismo , RNA/genética , RNA/metabolismo
16.
Annu Rev Pathol ; 16: 409-432, 2021 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-33276706

RESUMO

Over the last four decades, the cancer biology field has concentrated on cellular and microenvironmental drivers of metastasis. Despite this focus, mortality rates upon diagnosis of metastatic disease remain essentially unchanged. Would a small change in perspective help? Knowing what constitutes an inhospitable, rather than hospitable, microenvironment could provide the inspiration necessary to develop better therapies and preventative strategies. In this review, we canvas the literature for hints about what characteristics four common antimetastatic niches-skeletal muscle, spleen, thyroid, and yellow bone marrow-have in common. We posit that thorough molecular and mechanistic characterization of antimetastatic tissues may inspire reimagined therapies that inhibit metastatic development and/or progression in an enduring manner.


Assuntos
Invasividade Neoplásica/patologia , Neoplasias/patologia , Microambiente Tumoral , Animais , Humanos
17.
Am J Pathol ; 174(6): 1996-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19465642

RESUMO

This Commentary highlights two articles in this issue of the American Journal of Pathology, discussing the implications of stromal expression of caveolin-1 in breast cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Caveolina 1/metabolismo , Matriz Extracelular , Feminino , Humanos
18.
Biotechnol Bioeng ; 107(6): 1020-8, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20672286

RESUMO

Stem cell niches are composed of numerous microenvironmental features, including soluble and insoluble factors, cues from other cells, and the extracellular matrix (ECM), which collectively serve to maintain stem cell quiescence and promote their ability to support tissue homeostasis. A hallmark of many adult stem cell niches is their proximity to the vasculature in vivo, a feature common to neural stem cells, mesenchymal stem cells (MSCs) from bone marrow and adipose tissue, hematopoietic stem cells, and many tumor stem cells. In this study, we describe a novel 3D microfluidic device (MFD) as a model system in which to study the molecular regulation of perivascular stem cell niches. Endothelial cells (ECs) suspended within 3D fibrin gels patterned in the device adjacent to stromal cells (either fibroblasts or bone marrow-derived MSCs) executed a morphogenetic process akin to vasculogenesis, forming a primitive vascular plexus and maturing into a robust capillary network with hollow well-defined lumens. Both MSCs and fibroblasts formed pericytic associations with the ECs but promoted capillary morphogenesis with distinct kinetics. Biochemical assays within the niche revealed that the perivascular association of MSCs required interaction between their α6ß1 integrin receptor and EC-deposited laminin. These studies demonstrate the potential of this physiologically relevant ex vivo model system to study how proximity to blood vessels may influence stem cell multipotency.


Assuntos
Microfluídica/métodos , Células Endoteliais , Matriz Extracelular/metabolismo , Humanos , Técnicas de Cultura de Órgãos/métodos , Células-Tronco
19.
Nat Cell Biol ; 21(2): 238-250, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30664790

RESUMO

The presence of disseminated tumour cells (DTCs) in bone marrow is predictive of poor metastasis-free survival of patients with breast cancer with localized disease. DTCs persist in distant tissues despite systemic administration of adjuvant chemotherapy. Many assume that this is because the majority of DTCs are quiescent. Here, we challenge this notion and provide evidence that the microenvironment of DTCs protects them from chemotherapy, independent of cell cycle status. We show that chemoresistant DTCs occupy the perivascular niche (PVN) of distant tissues, where they are protected from therapy by vascular endothelium. Inhibiting integrin-mediated interactions between DTCs and the PVN, driven partly by endothelial-derived von Willebrand factor and vascular cell adhesion molecule 1, sensitizes DTCs to chemotherapy. Importantly, chemosensitization is achieved without inducing DTC proliferation or exacerbating chemotherapy-associated toxicities, and ultimately results in prevention of bone metastasis. This suggests that prefacing adjuvant therapy with integrin inhibitors is a viable clinical strategy to eradicate DTCs and prevent metastasis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Feminino , Integrinas/metabolismo , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Paclitaxel/administração & dosagem
20.
Nat Cell Biol ; 21(11): 1403-1412, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31685984

RESUMO

The development of effective therapies against brain metastasis is currently hindered by limitations in our understanding of the molecular mechanisms driving it. Here we define the contributions of tumour-secreted exosomes to brain metastatic colonization and demonstrate that pre-conditioning the brain microenvironment with exosomes from brain metastatic cells enhances cancer cell outgrowth. Proteomic analysis identified cell migration-inducing and hyaluronan-binding protein (CEMIP) as elevated in exosomes from brain metastatic but not lung or bone metastatic cells. CEMIP depletion in tumour cells impaired brain metastasis, disrupting invasion and tumour cell association with the brain vasculature, phenotypes rescued by pre-conditioning the brain microenvironment with CEMIP+ exosomes. Moreover, uptake of CEMIP+ exosomes by brain endothelial and microglial cells induced endothelial cell branching and inflammation in the perivascular niche by upregulating the pro-inflammatory cytokines encoded by Ptgs2, Tnf and Ccl/Cxcl, known to promote brain vascular remodelling and metastasis. CEMIP was elevated in tumour tissues and exosomes from patients with brain metastasis and predicted brain metastasis progression and patient survival. Collectively, our findings suggest that targeting exosomal CEMIP could constitute a future avenue for the prevention and treatment of brain metastasis.


Assuntos
Neoplasias Encefálicas/genética , Exossomos/metabolismo , Regulação Neoplásica da Expressão Gênica , Hialuronoglucosaminidase/genética , Neovascularização Patológica/genética , Microambiente Tumoral/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quimiocina CCL1/genética , Quimiocina CCL1/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Exossomos/patologia , Humanos , Hialuronoglucosaminidase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Metástase Neoplásica , Neovascularização Patológica/metabolismo , Neovascularização Patológica/mortalidade , Neovascularização Patológica/patologia , Transdução de Sinais , Análise de Sobrevida , Carga Tumoral , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA