Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters








Publication year range
1.
bioRxiv ; 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39345453

ABSTRACT

During persistent antigen stimulation, PD-1 + CD8 T cells are maintained by progenitor exhausted PD-1 + TCF-1 + CD8 T cells (Tpex). Tpex respond to PD-1 blockade, and regulation of Tpex differentiation into more functional Tex is of major interest for cancer immunotherapies. Tpex express high levels of Inducible Costimulator (ICOS), but the role of ICOS for PD-1 + CD8 T cell responses has not been addressed. In chronic infection, ICOS-deficiency increased both number and quality of virus-specific CD8 T cells, with accumulation of effector-like Tex due to enhanced survival. Mechanistically, loss of ICOS signaling potentiated FoxO1 activity and memory-like features of Tpex. In mice with established chronic infection, ICOS-Ligand blockade resulted in expansion of effector-like Tex and reduction in viral load. In a mouse model of hepatocellular carcinoma, ICOS inhibition improved cytokine production by tumor-specific PD-1 + CD8 T cells and delayed tumor growth. Overall, we show that ICOS limits CD8 T cell responses during chronic antigen exposure.

2.
bioRxiv ; 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39229151

ABSTRACT

Multimeric SWI/SNF chromatin remodelers assemble into discrete conformations with unique complex functionalities difficult to dissect. Distinct cancers harbor mutations in specific subunits, altering the chromatin landscape, such as the PBAF-specific component ARID2 in melanoma. Here, we performed comprehensive epigenomic profiling of SWI/SNF complexes and their associated chromatin states in melanoma and melanocytes and uncovered a subset of PBAF-exclusive regions that coexist with PRC2 and repressive chromatin. Time-resolved approaches revealed that PBAF regions are generally less sensitive to ATPase-mediated remodeling than BAF sites. Moreover, PBAF/PRC2-bound loci are enriched for REST, a transcription factor that represses neuronal genes. In turn, absence of ARID2 and consequent PBAF complex disruption hinders the ability of REST to bind and inactivate its targets, leading to upregulation of synaptic transcripts. Remarkably, this gene signature is conserved in melanoma patients with ARID2 mutations. In sum, we demonstrate a unique role for PBAF in generating accessibility for a silencing transcription factor at repressed chromatin, with important implications for disease.

3.
Nat Cell Biol ; 25(9): 1332-1345, 2023 09.
Article in English | MEDLINE | ID: mdl-37605008

ABSTRACT

MacroH2A has established tumour suppressive functions in melanoma and other cancers, but an unappreciated role in the tumour microenvironment. Using an autochthonous, immunocompetent mouse model of melanoma, we demonstrate that mice devoid of macroH2A variants exhibit increased tumour burden compared with wild-type counterparts. MacroH2A-deficient tumours accumulate immunosuppressive monocytes and are depleted of functional cytotoxic T cells, characteristics consistent with a compromised anti-tumour response. Single cell and spatial transcriptomics identify increased dedifferentiation along the neural crest lineage of the tumour compartment and increased frequency and activation of cancer-associated fibroblasts following macroH2A loss. Mechanistically, macroH2A-deficient cancer-associated fibroblasts display increased myeloid chemoattractant activity as a consequence of hyperinducible expression of inflammatory genes, which is enforced by increased chromatin looping of their promoters to enhancers that gain H3K27ac. In summary, we reveal a tumour suppressive role for macroH2A variants through the regulation of chromatin architecture in the tumour stroma with potential implications for human melanoma.


Subject(s)
Cancer-Associated Fibroblasts , Histones , Melanoma , Animals , Mice , Chromatin/genetics , Gene Expression , Histones/genetics , Melanoma/genetics , Tumor Microenvironment/genetics
4.
Sci Immunol ; 8(86): eadg0878, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37624910

ABSTRACT

During persistent antigen stimulation, such as in chronic infections and cancer, CD8 T cells differentiate into a hypofunctional programmed death protein 1-positive (PD-1+) exhausted state. Exhausted CD8 T cell responses are maintained by precursors (Tpex) that express the transcription factor T cell factor 1 (TCF-1) and high levels of the costimulatory molecule CD28. Here, we demonstrate that sustained CD28 costimulation is required for maintenance of antiviral T cells during chronic infection. Low-level CD28 engagement preserved mitochondrial fitness and self-renewal of Tpex, whereas stronger CD28 signaling enhanced glycolysis and promoted Tpex differentiation into TCF-1neg exhausted CD8 T cells (Tex). Furthermore, enhanced differentiation by CD28 engagement did not reduce the Tpex pool. Together, these findings demonstrate that continuous CD28 engagement is needed to sustain PD-1+ CD8 T cells and suggest that increasing CD28 signaling promotes Tpex differentiation into more functional effector-like Tex, possibly without compromising long-term responses.


Subject(s)
CD28 Antigens , T Cell Transcription Factor 1 , T Cell Transcription Factor 1/genetics , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes , Cell Differentiation , Transcription Factors
5.
Sci Adv ; 8(48): eabo0876, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36459552

ABSTRACT

MacroH2A variants have been linked to inhibition of metastasis through incompletely understood mechanisms. Here, we reveal that solitary dormant disseminated cancer cells (DCCs) display increased levels of macroH2A variants in head and neck squamous cell carcinoma PDX in vivo models and patient samples compared to proliferating primary or metastatic lesions. We demonstrate that dormancy-inducing transforming growth factor-ß2 and p38α/ß pathways up-regulate macroH2A expression and that macroH2A variant overexpression is sufficient to induce DCC dormancy and suppress metastasis in vivo. Notably, inducible expression of the macroH2A2 variant in vivo suppresses metastasis via a reversible growth arrest of DCCs. This state does not require the dormancy-regulating transcription factors DEC2 and NR2F1; instead, transcriptomic analysis reveals that macroH2A2 overexpression inhibits cell cycle and oncogenic signaling programs, while up-regulating dormancy and senescence-associated inflammatory cytokines. We conclude that the macroH2A2-enforced dormant phenotype results from tapping into transcriptional programs of both quiescence and senescence to limit metastatic outgrowth.


Subject(s)
Head and Neck Neoplasms , Histones , Humans , Carcinogenesis , Cell Division , Cell Cycle , Head and Neck Neoplasms/genetics
6.
Cell Rep ; 39(1): 110637, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385731

ABSTRACT

ARID2 is the most recurrently mutated SWI/SNF complex member in melanoma; however, its tumor-suppressive mechanisms in the context of the chromatin landscape remain to be elucidated. Here, we model ARID2 deficiency in melanoma cells, which results in defective PBAF complex assembly with a concomitant genomic redistribution of the BAF complex. Upon ARID2 depletion, a subset of PBAF and shared BAF-PBAF-occupied regions displays diminished chromatin accessibility and associated gene expression, while BAF-occupied enhancers gain chromatin accessibility and expression of genes linked to the process of invasion. As a function of altered accessibility, the genomic occupancy of melanoma-relevant transcription factors is affected and significantly correlates with the observed transcriptional changes. We further demonstrate that ARID2-deficient cells acquire the ability to colonize distal organs in multiple animal models. Taken together, our results reveal a role for ARID2 in mediating BAF and PBAF subcomplex chromatin dynamics with consequences for melanoma metastasis.


Subject(s)
Chromosomal Proteins, Non-Histone , Melanoma , Transcription Factors , Animals , Chromatin , Chromatin Assembly and Disassembly , Gene Expression Regulation , Humans , Melanoma/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Nat Rev Cancer ; 21(4): 257-275, 2021 04.
Article in English | MEDLINE | ID: mdl-33568791

ABSTRACT

Cancer is a complex disease characterized by loss of cellular homeostasis through genetic and epigenetic alterations. Emerging evidence highlights a role for histone variants and their dedicated chaperones in cancer initiation and progression. Histone variants are involved in processes as diverse as maintenance of genome integrity, nuclear architecture and cell identity. On a molecular level, histone variants add a layer of complexity to the dynamic regulation of transcription, DNA replication and repair, and mitotic chromosome segregation. Because these functions are critical to ensure normal proliferation and maintenance of cellular fate, cancer cells are defined by their capacity to subvert them. Hijacking histone variants and their chaperones is emerging as a common means to disrupt homeostasis across a wide range of cancers, particularly solid tumours. Here we discuss histone variants and histone chaperones as tumour-promoting or tumour-suppressive players in the pathogenesis of cancer.


Subject(s)
Histone Chaperones/metabolism , Histones/metabolism , Neoplasms/metabolism , Chromatin/metabolism , Histone Chaperones/genetics , Histones/genetics , Humans , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Neoplasms/genetics
8.
Cancer Cell ; 36(5): 512-527.e9, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31631027

ABSTRACT

ATRX alterations occur at high frequency in neuroblastoma of adolescents and young adults. Particularly intriguing are the large N-terminal deletions of ATRX (Alpha Thalassemia/Mental Retardation, X-linked) that generate in-frame fusion (IFF) proteins devoid of key chromatin interaction domains, while retaining the SWI/SNF-like helicase region. We demonstrate that ATRX IFF proteins are redistributed from H3K9me3-enriched chromatin to promoters of active genes and identify REST as an ATRX IFF target whose activation promotes silencing of neuronal differentiation genes. We further show that ATRX IFF cells display sensitivity to EZH2 inhibitors, due to derepression of neurogenesis genes, including a subset of REST targets. Taken together, we demonstrate that ATRX structural alterations are not loss-of-function and put forward EZH2 inhibitors as a potential therapy for ATRX IFF neuroblastoma.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Neuroblastoma/drug therapy , Repressor Proteins/genetics , X-linked Nuclear Protein/genetics , Animals , Base Sequence/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Male , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/surgery , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Promoter Regions, Genetic , Protein Domains/genetics , Sequence Deletion , X-linked Nuclear Protein/metabolism , Xenograft Model Antitumor Assays
9.
Nat Struct Mol Biol ; 25(10): 958-970, 2018 10.
Article in English | MEDLINE | ID: mdl-30291361

ABSTRACT

The histone variant macroH2A occupies large repressive domains throughout the genome; however, mechanisms underlying its precise deposition remain poorly understood. Here, we characterize de novo chromatin deposition of macroH2A2 using temporal genomic profiling in murine-derived fibroblasts devoid of all macroH2A isoforms. We find that macroH2A2 is first pervasively deposited genome wide at both steady-state domains and adjacent transcribed regions, the latter of which are subsequently pruned, establishing mature macroH2A2 domains. Pruning of macroH2A2 can be counteracted by chemical inhibition of transcription. Further, locus-specific transcriptional manipulation reveals that gene activation depletes pre-existing macroH2A2, while silencing triggers ectopic macroH2A2 accumulation. We demonstrate that the FACT (facilitates chromatin transcription) complex is required for macroH2A2 pruning within transcribed chromatin. Taken together, we have identified active chromatin as a boundary for macroH2A domains through a transcription-associated 'pruning' mechanism that establishes and maintains the faithful genomic localization of macroH2A variants.


Subject(s)
Chromatin/metabolism , Histones/physiology , Transcription, Genetic , Animals , Chromatin/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation , Histones/chemistry , Histones/metabolism , Male , Mice , Models, Molecular
10.
Mol Cell ; 68(4): 731-744.e9, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149598

ABSTRACT

Bromodomain and extraterminal domain inhibitors (BETi) represent promising therapeutic agents for metastatic melanoma, yet their mechanism of action remains unclear. Here we interrogated the transcriptional effects of BETi and identified AMIGO2, a transmembrane molecule, as a BET target gene essential for melanoma cell survival. AMIGO2 is upregulated in melanoma cells and tissues compared to human melanocytes and nevi, and AMIGO2 silencing in melanoma cells induces G1/S arrest followed by apoptosis. We identified the pseudokinase PTK7 as an AMIGO2 interactor whose function is regulated by AMIGO2. Epigenomic profiling and genome editing revealed that AMIGO2 is regulated by a melanoma-specific BRD2/4-bound promoter and super-enhancer configuration. Upon BETi treatment, BETs are evicted from these regulatory elements, resulting in AMIGO2 silencing and changes in PTK7 proteolytic processing. Collectively, this study uncovers mechanisms underlying the therapeutic effects of BETi in melanoma and reveals the AMIGO2-PTK7 axis as a targetable pathway for metastatic melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Enhancer Elements, Genetic , Melanoma/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Male , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Genes Dev ; 31(5): 463-480, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28356341

ABSTRACT

In mammals, centromere definition involves the histone variant CENP-A (centromere protein A), deposited by its chaperone, HJURP (Holliday junction recognition protein). Alterations in this process impair chromosome segregation and genome stability, which are also compromised by p53 inactivation in cancer. Here we found that CENP-A and HJURP are transcriptionally up-regulated in p53-null human tumors. Using an established mouse embryonic fibroblast (MEF) model combining p53 inactivation with E1A or HRas-V12 oncogene expression, we reproduced a similar up-regulation of HJURP and CENP-A. We delineate functional CDE/CHR motifs within the Hjurp and Cenpa promoters and demonstrate their roles in p53-mediated repression. To assess the importance of HJURP up-regulation in transformed murine and human cells, we used a CRISPR/Cas9 approach. Remarkably, depletion of HJURP leads to distinct outcomes depending on their p53 status. Functional p53 elicits a cell cycle arrest response, whereas, in p53-null transformed cells, the absence of arrest enables the loss of HJURP to induce severe aneuploidy and, ultimately, apoptotic cell death. We thus tested the impact of HJURP depletion in pre-established allograft tumors in mice and revealed a major block of tumor progression in vivo. We discuss a model in which an "epigenetic addiction" to the HJURP chaperone represents an Achilles' heel in p53-deficient transformed cells.


Subject(s)
Autoantigens/metabolism , Cell Transformation, Neoplastic/genetics , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Oncogenes/genetics , Amino Acid Motifs/genetics , Animals , Autoantigens/genetics , Cell Line , Cells, Cultured , Centromere Protein A , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation/genetics , DNA-Binding Proteins/genetics , Female , Gene Deletion , Genomic Instability/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal
12.
Annu Rev Cell Dev Biol ; 30: 615-46, 2014.
Article in English | MEDLINE | ID: mdl-25288118

ABSTRACT

Within the nucleus, the interplay between lineage-specific transcription factors and chromatin dynamics defines cellular identity. Control of this interplay is necessary to properly balance stability and plasticity during the development and entire life span of multicellular organisms. Here, we present our current knowledge of the contribution of histone H3 variants to chromatin dynamics during development. We review the network of histone chaperones that governs their deposition timing and sites of incorporation and highlight how their distinct distribution impacts genome organization and function. We integrate the importance of H3 variants in the context of nuclear reprogramming and cell differentiation, and, using the centromere as a paradigm, we describe a case in which the identity of a given genomic locus is propagated across different cell types. Finally, we compare development to changes in stress and disease. Both physiological and pathological settings underline the importance of H3 dynamics for genome and chromatin integrity.


Subject(s)
Epigenesis, Genetic/physiology , Histone Code , Histones/physiology , Molecular Chaperones/physiology , Amino Acid Sequence , Animals , Blastocyst , Cell Lineage , Chromatin Assembly and Disassembly , Conserved Sequence , Epigenesis, Genetic/genetics , Fertilization , Gametogenesis/genetics , Histones/chemistry , Histones/genetics , Humans , Molecular Chaperones/genetics , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity
13.
J Cell Sci ; 127(Pt 15): 3347-59, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24906798

ABSTRACT

Correct chromosome segregation requires a unique chromatin environment at centromeres and in their vicinity. Here, we address how the deposition of canonical H2A and H2A.Z histone variants is controlled at pericentric heterochromatin (PHC). Whereas in euchromatin newly synthesized H2A and H2A.Z are deposited throughout the cell cycle, we reveal two discrete waves of deposition at PHC - during mid to late S phase in a replication-dependent manner for H2A and during G1 phase for H2A.Z. This G1 cell cycle restriction is lost when heterochromatin features are altered, leading to the accumulation of H2A.Z at the domain. Interestingly, compromising PHC integrity also impacts upon neighboring centric chromatin, increasing the amount of centromeric CENP-A without changing the timing of its deposition. We conclude that the higher-order chromatin structure at the pericentric domain influences dynamics at the nucleosomal level within centromeric chromatin. The two different modes of rearrangement of the PHC during the cell cycle provide distinct opportunities to replenish one or the other H2A variant, highlighting PHC integrity as a potential signal to regulate the deposition timing and stoichiometry of histone variants at the centromere.


Subject(s)
Cell Cycle , Histones/metabolism , 3T3 Cells , Animals , Autoantigens/genetics , Autoantigens/metabolism , Centromere/genetics , Centromere Protein A , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , Heterochromatin , Histones/genetics , Mice , Multiprotein Complexes/metabolism
14.
Neuro Oncol ; 16(9): 1196-209, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24558021

ABSTRACT

BACKGROUND: Inactivation of the NF2 gene predisposes to neurofibromatosis type II and the development of schwannomas. In vitro studies have shown that loss of NF2 leads to the induction of mitogenic signaling mediated by receptor tyrosine kinases (RTKs), MAP kinase, AKT, or Hippo pathways. The goal of our study was to evaluate the expression and activity of these signaling pathways in human schwannomas in order to identify new potential therapeutic targets. METHODS: Large sets of human schwannomas, totaling 68 tumors, were analyzed using complementary proteomic approaches. RTK arrays identified the most frequently activated RTKs. The correlation between the expression and activity of signaling pathways and proliferation of tumor cells using Ki67 marker was investigated by reverse-phase protein array (RRPA). Finally, immunohistochemistry was used to evaluate the expression pattern of signaling effectors in the tumors. RESULTS: We showed that Her2, Her3, PDGFRß, Axl, and Tie2 are frequently activated in the tumors. Furthermore, RRPA demonstrated that Ki67 levels are linked to YAP, p-Her3, and PDGFRß expression levels. In addition, Her2, Her3, and PDGFRß are transcriptional targets of Yes-associated protein (YAP) in schwannoma cells in culture. Finally, we observed that the expression of these signaling effectors is very variable between tumors. CONCLUSIONS: Tumor cell proliferation in human schwannomas is linked to a signaling network controlled by the Hippo effector YAP. Her2, Her3, PDGFRß, Axl, and Tie2, as well as YAP, represent potentially valuable therapeutic targets. However, the variability of their expression between tumors may result in strong differences in the response to targeted therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain Neoplasms/metabolism , Neurilemmoma/metabolism , Neurofibromatosis 2/metabolism , Phosphoproteins/metabolism , Signal Transduction , Cell Proliferation , Female , Humans , Male , Proteomics , Transcription Factors , YAP-Signaling Proteins
15.
Trends Genet ; 29(11): 630-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23830582

ABSTRACT

Animal development and lifetime potential exploit a balance between the stability and plasticity of cellular identity. Within the nucleus, this is controlled by an interplay involving lineage-specific transcription factors and chromatin dynamics. Histone H3 variants contribute to chromatin dynamics through the timing and sites of their incorporation, promoted by dedicated histone chaperones. Moreover, their individual modifications and binding partners provide distinct features at defined genomic loci. We highlight here the importance of the H3.3 replacement variant for the nuclear reprogramming that occurs during gametogenesis, fertilization, and germline establishment. Furthermore, we describe how the recently characterized H3.3 dynamics associated with gastrulation, myogenesis, or neurogenesis underline the role of chromatin changes in cell differentiation. Finally, we discuss the challenges of maintaining centromeric identity through propagation of the centromeric CenH3 variant in different cell types. Future challenges will be to gain a comprehensive picture of H3 variants and their chaperones during development and differentiation.


Subject(s)
Gametogenesis/genetics , Histones/genetics , Animals , Cell Cycle , Cell Nucleus/genetics , Centromere/genetics , Chromatin/genetics , Chromatin/metabolism , Fertilization/genetics , Gametogenesis/physiology , Germ Cells/metabolism , Histones/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL