Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Nature ; 620(7976): 1080-1088, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37612508

ABSTRACT

Chromosomal instability (CIN) is a driver of cancer metastasis1-4, yet the extent to which this effect depends on the immune system remains unknown. Using ContactTracing-a newly developed, validated and benchmarked tool to infer the nature and conditional dependence of cell-cell interactions from single-cell transcriptomic data-we show that CIN-induced chronic activation of the cGAS-STING pathway promotes downstream signal re-wiring in cancer cells, leading to a pro-metastatic tumour microenvironment. This re-wiring is manifested by type I interferon tachyphylaxis selectively downstream of STING and a corresponding increase in cancer cell-derived endoplasmic reticulum (ER) stress response. Reversal of CIN, depletion of cancer cell STING or inhibition of ER stress response signalling abrogates CIN-dependent effects on the tumour microenvironment and suppresses metastasis in immune competent, but not severely immune compromised, settings. Treatment with STING inhibitors reduces CIN-driven metastasis in melanoma, breast and colorectal cancers in a manner dependent on tumour cell-intrinsic STING. Finally, we show that CIN and pervasive cGAS activation in micronuclei are associated with ER stress signalling, immune suppression and metastasis in human triple-negative breast cancer, highlighting a viable strategy to identify and therapeutically intervene in tumours spurred by CIN-induced inflammation.


Subject(s)
Chromosomal Instability , Disease Progression , Neoplasms , Humans , Benchmarking , Cell Communication , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Tumor Microenvironment , Interferon Type I/immunology , Neoplasm Metastasis , Endoplasmic Reticulum Stress , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology
2.
Nature ; 607(7918): 366-373, 2022 07.
Article in English | MEDLINE | ID: mdl-35705809

ABSTRACT

Chromosomal instability (CIN) drives cancer cell evolution, metastasis and therapy resistance, and is associated with poor prognosis1. CIN leads to micronuclei that release DNA into the cytoplasm after rupture, which triggers activation of inflammatory signalling mediated by cGAS and STING2,3. These two proteins are considered to be tumour suppressors as they promote apoptosis and immunosurveillance. However, cGAS and STING are rarely inactivated in cancer4, and, although they have been implicated in metastasis5, it is not known why loss-of-function mutations do not arise in primary tumours4. Here we show that inactivation of cGAS-STING signalling selectively impairs the survival of triple-negative breast cancer cells that display CIN. CIN triggers IL-6-STAT3-mediated signalling, which depends on the cGAS-STING pathway and the non-canonical NF-κB pathway. Blockade of IL-6 signalling by tocilizumab, a clinically used drug that targets the IL-6 receptor (IL-6R), selectively impairs the growth of cultured triple-negative breast cancer cells that exhibit CIN. Moreover, outgrowth of chromosomally instable tumours is significantly delayed compared with tumours that do not display CIN. Notably, this targetable vulnerability is conserved across cancer types that express high levels of IL-6 and/or IL-6R in vitro and in vivo. Together, our work demonstrates pro-tumorigenic traits of cGAS-STING signalling and explains why the cGAS-STING pathway is rarely inactivated in primary tumours. Repurposing tocilizumab could be a strategy to treat cancers with CIN that overexpress IL-6R.


Subject(s)
Chromosomal Instability , Interleukin-6 , Membrane Proteins , Nucleotidyltransferases , Triple Negative Breast Neoplasms , Antibodies, Monoclonal, Humanized/pharmacology , Cell Survival/drug effects , Chromosomal Instability/genetics , Drug Repositioning , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , NF-kappa B/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
3.
Dev Cell ; 56(17): 2440-2454.e6, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34352223

ABSTRACT

Mitotic errors lead to aneuploidy, a condition of karyotype imbalance, frequently found in cancer cells. Alterations in chromosome copy number induce a wide variety of cellular stresses, including genome instability. Here, we show that cancer cells might exploit aneuploidy-induced genome instability and the resulting gene copy-number changes to survive under conditions of selective pressure, such as chemotherapy. Resistance to chemotherapeutic drugs was dictated by the acquisition of recurrent karyotypes, indicating that gene dosage might play a role in driving chemoresistance. Thus, our study establishes a causal link between aneuploidy-driven changes in gene copy number and chemoresistance and might explain why some chemotherapies fail to succeed.


Subject(s)
Aneuploidy , Chromosomal Instability/genetics , Drug Resistance/genetics , Drug Therapy , Gene Dosage/genetics , Drug Therapy/methods , Genomic Instability/genetics , Humans , Karyotype
4.
Cells ; 8(10)2019 10 10.
Article in English | MEDLINE | ID: mdl-31658669

ABSTRACT

Chromosomal instability (CIN) is an intricate phenomenon that is often found in human cancer, characterized by persisting errors in chromosome segregation. This ongoing chromosome mis-segregation results in structural and numerical chromosomal abnormalities that have been widely described to promote tumor evolution. In addition to being a driver of tumor evolution, recent evidence demonstrates CIN to be the central node of the crosstalk between a tumor and its surrounding microenvironment, as mediated by the cGAS-STING pathway. The role that cGAS-STING signaling exerts on CIN tumors is both complex and paradoxical. On one hand, the cGAS-STING axis promotes the clearance of CIN tumors through recruitment of immune cells, thus suppressing tumor progression. On the other hand, the cGAS-STING pathway has been described to be the major regulator in the promotion of metastasis of CIN tumors. Here, we review this dual role of the cGAS-STING pathway in the context of chromosomal instability and discuss the potential therapeutic implications of cGAS-STING signaling for targeting CIN tumors.


Subject(s)
Chromosomal Instability , Neoplasms/genetics , Signal Transduction , Animals , Disease Progression , Humans , Membrane Proteins/metabolism , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL