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.
Nat Commun ; 13(1): 6579, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36323660

ABSTRACT

The limited efficacy of immune checkpoint inhibitor treatment in triple-negative breast cancer (TNBC) patients is attributed to sparse or unresponsive tumor-infiltrating lymphocytes, but the mechanisms that lead to a therapy resistant tumor immune microenvironment are incompletely known. Here we show a strong correlation between MYC expression and loss of immune signatures in human TNBC. In mouse models of TNBC proficient or deficient of breast cancer type 1 susceptibility gene (BRCA1), MYC overexpression dramatically decreases lymphocyte infiltration in tumors, along with immune signature remodelling. MYC-mediated suppression of inflammatory signalling induced by BRCA1/2 inactivation is confirmed in human TNBC cell lines. Moreover, MYC overexpression prevents the recruitment and activation of lymphocytes in both human and mouse TNBC co-culture models. Chromatin-immunoprecipitation-sequencing reveals that MYC, together with its co-repressor MIZ1, directly binds promoters of multiple interferon-signalling genes, resulting in their downregulation. MYC overexpression thus counters tumor growth inhibition by a Stimulator of Interferon Genes (STING) agonist via suppressing induction of interferon signalling. Together, our data reveal that MYC suppresses innate immunity and facilitates tumor immune escape, explaining the poor immunogenicity of MYC-overexpressing TNBCs.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Interferons , Lymphocytes, Tumor-Infiltrating , Signal Transduction , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment/genetics , Proto-Oncogene Proteins c-myc/metabolism
2.
Cell Cycle ; 18(16): 1830-1848, 2019 08.
Article in English | MEDLINE | ID: mdl-31260383

ABSTRACT

Recent studies have shown that genomic instability in tumor cells leads to activation of inflammatory signaling through the cGAS/STING pathway. In this review, we describe multiple ways by which genomic instability leads to cGAS/STING-mediated inflammatory signaling, as well as the consequences for tumor development and the tumor microenvironment. Also, we elaborate on how tumor cells have apparently evolved to escape the immune surveillance mechanisms that are triggered by cGAS/STING signaling. Finally, we describe how cGAS/STING-mediated inflammatory signaling can be therapeutically targeted to improve therapy responses.


Subject(s)
Genomic Instability , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/genetics , Animals , Cytosol/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Humans , Immunotherapy/methods , Inflammation/metabolism , Membrane Proteins/metabolism , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Nucleotidyltransferases/metabolism , Signal Transduction/drug effects , Tumor Escape/genetics , Tumor Microenvironment/genetics
3.
Nat Commun ; 10(1): 100, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626869

ABSTRACT

Loss of BRCA2 affects genome stability and is deleterious for cellular survival. Using a genome-wide genetic screen in near-haploid KBM-7 cells, we show that tumor necrosis factor-alpha (TNFα) signaling is a determinant of cell survival upon BRCA2 inactivation. Specifically, inactivation of the TNF receptor (TNFR1) or its downstream effector SAM68 rescues cell death induced by BRCA2 inactivation. BRCA2 inactivation leads to pro-inflammatory cytokine production, including TNFα, and increases sensitivity to TNFα. Enhanced TNFα sensitivity is not restricted to BRCA2 inactivation, as BRCA1 or FANCD2 inactivation, or hydroxyurea treatment also sensitizes cells to TNFα. Mechanistically, BRCA2 inactivation leads to cGAS-positive micronuclei and results in a cell-intrinsic interferon response, as assessed by quantitative mass-spectrometry and gene expression profiling, and requires ASK1 and JNK signaling. Combined, our data reveals that micronuclei induced by loss of BRCA2 instigate a cGAS/STING-mediated interferon response, which encompasses re-wired TNFα signaling and enhances TNFα sensitivity.


Subject(s)
BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Survival/physiology , Inflammation/metabolism , Nucleotidyltransferases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Gene Deletion , Humans , Nucleotidyltransferases/genetics , Signal Transduction
4.
Nat Commun ; 8: 15981, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28714471

ABSTRACT

Mutations in homologous recombination (HR) genes BRCA1 and BRCA2 predispose to tumorigenesis. HR-deficient cancers are hypersensitive to Poly (ADP ribose)-polymerase (PARP) inhibitors, but can acquire resistance and relapse. Mechanistic understanding how PARP inhibition induces cytotoxicity in HR-deficient cancer cells is incomplete. Here we find PARP inhibition to compromise replication fork stability in HR-deficient cancer cells, leading to mitotic DNA damage and consequent chromatin bridges and lagging chromosomes in anaphase, frequently leading to cytokinesis failure, multinucleation and cell death. PARP-inhibitor-induced multinucleated cells fail clonogenic outgrowth, and high percentages of multinucleated cells are found in vivo in remnants of PARP inhibitor-treated Brca2-/-;p53-/- and Brca1-/-;p53-/- mammary mouse tumours, suggesting that mitotic progression promotes PARP-inhibitor-induced cell death. Indeed, enforced mitotic bypass through EMI1 depletion abrogates PARP-inhibitor-induced cytotoxicity. These findings provide insight into the cytotoxic effects of PARP inhibition, and point at combination therapies to potentiate PARP inhibitor treatment of HR-deficient tumours.


Subject(s)
Anaphase/drug effects , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Cytokinesis/drug effects , DNA Damage/drug effects , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Experimental/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , HeLa Cells , Humans , Mice , Mice, Knockout , Mitosis/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Recombinational DNA Repair/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
5.
Expert Opin Drug Discov ; 12(6): 565-581, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28425306

ABSTRACT

INTRODUCTION: DNA double-strand breaks (DSBs) are toxic DNA lesions that can be repaired by non-homologous end-joining (NHEJ) or homologous recombination (HR). Mutations in HR genes elicit a predisposition to cancer; yet, they also result in increased sensitivity to certain DNA damaging agents and poly (ADP-ribose) polymerase (PARP) inhibitors. To optimally implement PARP inhibitor treatment, it is important that patients with HR-deficient tumors are adequately selected. Areas covered: Herein, the authors describe the HR pathway mechanistically and review the treatment of HR-deficient cancers, with a specific focus on PARP inhibition for BRCA1/2-mutated breast and ovarian cancer. In addition, mechanisms of acquired PARP inhibitor resistance are discussed. Furthermore, combination therapies with PARP inhibitors are reviewed, in the context of both HR-deficient and HR-proficient tumors and methods for proper patient selection are also discussed. Expert opinion: Currently, only patients with germline or somatic BRCA1/2 mutations are eligible for PARP inhibitor treatment and only a proportion of patients respond. Patients with HR-deficient tumors caused by other (epi)genetic events may also benefit from PARP inhibitor treatment. Ideally, selection of eligible patients for PARP inhibitor treatment include a functional HR read-out, in which cancer cells are interrogated for their ability to perform HR repair and maintain replication fork stability.


Subject(s)
Molecular Targeted Therapy , Patient Selection , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA Breaks, Double-Stranded , Drug Design , Female , Genetic Predisposition to Disease , Humans , Mutation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Recombinational DNA Repair/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...