Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Res ; 83(20): 3354-3367, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37548552

ABSTRACT

Metastatic cancer is largely incurable and is the main cause of cancer-related deaths. The metastatic microenvironment facilitates formation of metastases. Cancer-associated fibroblasts (CAF) are crucial players in generating a hospitable metastatic niche by mediating an inflammatory microenvironment. Fibroblasts also play a central role in modifying the architecture and stiffness of the extracellular matrix (ECM). Resolving the early changes in the metastatic niche could help identify approaches to inhibit metastatic progression. Here, we demonstrate in mouse models of spontaneous breast cancer pulmonary metastasis that fibrotic changes and rewiring of lung fibroblasts occurred at premetastatic stages, suggesting systemic influence by the primary tumor. Activin A (ActA), a TGFß superfamily member, was secreted from breast tumors and its levels in the blood were highly elevated in tumor-bearing mice. ActA upregulated the expression of profibrotic factors in lung fibroblasts, leading to enhanced collagen deposition in the lung premetastatic niche. ActA signaling was functionally important for lung metastasis, as genetic targeting of ActA in breast cancer cells significantly attenuated lung metastasis and improved survival. Moreover, high levels of ActA in human patients with breast cancer were associated with lung metastatic relapse and poor survival. This study uncovers a novel mechanism by which breast cancer cells systemically rewire the stromal microenvironment in the metastatic niche to facilitate pulmonary metastasis. SIGNIFICANCE: ActA mediates cross-talk between breast cancer cells and cancer-associated fibroblasts in the lung metastatic niche that enhances fibrosis and metastasis, implicating ActA as a potential therapeutic target to inhibit metastatic relapse.

2.
Trends Cell Biol ; 32(2): 110-123, 2022 02.
Article in English | MEDLINE | ID: mdl-34479765

ABSTRACT

Modification of the extracellular matrix (ECM) is a critical aspect of developing a metastasis-supportive organ niche. Recent work investigating ECM changes that facilitate metastasis has revealed ways in which different metastatic organ niches are similar as well as the distinct characteristics that make them unique. In this review, we present recent findings regarding how ECM modifications support metastasis in four frequent metastatic sites: the lung, liver, bone, and brain. We discuss ways in which these modifications are shared between metastatic organs as well as features specific to each location. We also discuss areas of technical innovation that could be advantageous to future research and areas of inquiry that merit further investigation.


Subject(s)
Neoplasms , Tumor Microenvironment , Brain/pathology , Extracellular Matrix , Humans , Lung/pathology , Neoplasm Metastasis/pathology , Neoplasms/pathology
3.
PLoS Genet ; 15(5): e1008020, 2019 05.
Article in English | MEDLINE | ID: mdl-31125342

ABSTRACT

Breast cancer is the second leading cause of cancer-related deaths in the United States, with the majority of these deaths due to metastatic lesions rather than the primary tumor. Thus, a better understanding of the etiology of metastatic disease is crucial for improving survival. Using a haplotype mapping strategy in mouse and shRNA-mediated gene knockdown, we identified Rnaseh2c, a scaffolding protein of the heterotrimeric RNase H2 endoribonuclease complex, as a novel metastasis susceptibility factor. We found that the role of Rnaseh2c in metastatic disease is independent of RNase H2 enzymatic activity, and immunophenotyping and RNA-sequencing analysis revealed engagement of the T cell-mediated adaptive immune response. Furthermore, the cGAS-Sting pathway was not activated in the metastatic cancer cells used in this study, suggesting that the mechanism of immune response in breast cancer is different from the mechanism proposed for Aicardi-Goutières Syndrome, a rare interferonopathy caused by RNase H2 mutation. These results suggest an important novel, non-enzymatic role for RNASEH2C during breast cancer progression and add Rnaseh2c to a panel of genes we have identified that together could determine patients with high risk for metastasis. These results also highlight a potential new target for combination with immunotherapies and may contribute to a better understanding of the etiology of Aicardi-Goutières Syndrome autoimmunity.


Subject(s)
Adaptive Immunity , Autoimmune Diseases of the Nervous System/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Nervous System Malformations/genetics , Ribonuclease H/genetics , Animals , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/mortality , Autoimmune Diseases of the Nervous System/pathology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lymphatic Metastasis , Mice , Mice, Nude , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Nervous System Malformations/immunology , Nervous System Malformations/mortality , Nervous System Malformations/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/immunology , Sequence Analysis, RNA , Signal Transduction , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology
4.
Nat Rev Cancer ; 18(4): 211-223, 2018 04.
Article in English | MEDLINE | ID: mdl-29422598

ABSTRACT

Tumour heterogeneity poses a substantial problem for the clinical management of cancer. Somatic evolution of the cancer genome results in genetically distinct subclones in the primary tumour with different biological properties and therapeutic sensitivities. The problem of heterogeneity is compounded in metastatic disease owing to the complexity of the metastatic process and the multiple biological hurdles that the tumour cell must overcome to establish a clinically overt metastatic lesion. New advances in sequencing technology and clinical sample acquisition are providing insights into the phylogenetic relationship of metastases and primary tumours at the level of somatic tumour genetics while also illuminating fundamental mechanisms of the metastatic process. In addition to somatically acquired genetic heterogeneity in the tumour cells, inherited population-based genetic heterogeneity can profoundly modify metastatic biology and further complicate the development of effective, broadly applicable antimetastatic therapies. Here, we examine how genetic heterogeneity impacts metastatic disease and the implications of current knowledge for future research endeavours and therapeutic interventions.


Subject(s)
Genetic Heterogeneity , Neoplasm Metastasis/genetics , Neoplasms/pathology , Animals , Genetic Predisposition to Disease , Humans , Mice , Neoplasm Metastasis/therapy , Neoplasms/genetics , Neoplasms, Experimental/genetics
5.
Article in English | MEDLINE | ID: mdl-33842931

ABSTRACT

Breast cancer is one of the leading causes of cancer-related mortality of women in the United States. Since the majority of cancer deaths are due to metastases rather than the primary tumor, a better understanding of the biological mechanisms that lead to metastatic disease is critical to reduce breast cancer associated mortality. Current adjuvant therapies use the same broadly cytotoxic and targeted strategies against metastases as are used against the primary tumor. However, resistance to chemotherapy due to the cellular dormancy, high genotypic and phenotypic heterogeneity between primary tumor and metastases as well as among individual metastases, and the limitations in detection of disseminated tumor cells and micrometastases significantly hinder the efficiency of currently available therapies. While it is crucial to directly address the issue of metastatic dormancy and evaluate for anti-metastatic therapy the relevance of molecular targets chosen based on primary tumor profiling, it is also imperative to address metastasis-specific mechanisms of growth and survival that are likely to be distinct from those of the primary tumor. We believe that a three-pronged approach to therapy will be necessary to deal with progressive disease: blocking of further dissemination after diagnosis; eradication of disseminated tumor cells and prevention of the dormant-to-proliferative switch of those remaining; and elimination of established metastatic tumors. The implementation of this strategy requires a greater depth of knowledge of metastasis driver and maintenance genes and suggests the need for a "Metastasis Genome Atlas" project to complement the current investigations into cancer genomic landscapes.

SELECTION OF CITATIONS
SEARCH DETAIL