Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Commun ; 13(1): 7182, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36418296

ABSTRACT

The association between loss of BRCA1/2 and a homologous recombination deficiency phenotype is lineage dependent. In BRCA-associated cancers such as breast, ovarian, pancreas and prostate, this phenotype confers sensitivity to PARP inhibitors and platinum-therapies. Somatic reversion mutations restoring BRCA1/2 function mediate resistance, and have exclusively been reported in BRCA-associated tumors. In this study, we analyze matched tumor and normal sequencing from 31,927 patients and identify 846 (2.7%) patients with germline BRCA1/2 variants across 43 different cancer types, including 11 with somatic reversion mutations. While nine are in BRCA-associated tumors, we find two reversion mutations in non-BRCA-associated histologies, namely lung and esophagogastric adenocarcinomas. Both were detected following platinum therapy. Whole exome sequencing confirms the homologous recombination deficiency phenotype of these tumors. While reversion mutations arise in all BRCA-associated cancer types, here we show that reversion mutations arising post-platinum in non-BRCA associated histologies, while rare, may indicate BRCA1/2 mediated tumorigenesis.


Subject(s)
Adenocarcinoma , Platinum , Humans , Male , BRCA1 Protein/genetics , Germ Cells , Mutation , Phenotype , BRCA2 Protein/genetics
2.
Nature ; 608(7924): 795-802, 2022 08.
Article in English | MEDLINE | ID: mdl-35978189

ABSTRACT

Although p53 inactivation promotes genomic instability1 and presents a route to malignancy for more than half of all human cancers2,3, the patterns through which heterogenous TP53 (encoding human p53) mutant genomes emerge and influence tumorigenesis remain poorly understood. Here, in a mouse model of pancreatic ductal adenocarcinoma that reports sporadic p53 loss of heterozygosity before cancer onset, we find that malignant properties enabled by p53 inactivation are acquired through a predictable pattern of genome evolution. Single-cell sequencing and in situ genotyping of cells from the point of p53 inactivation through progression to frank cancer reveal that this deterministic behaviour involves four sequential phases-Trp53 (encoding mouse p53) loss of heterozygosity, accumulation of deletions, genome doubling, and the emergence of gains and amplifications-each associated with specific histological stages across the premalignant and malignant spectrum. Despite rampant heterogeneity, the deletion events that follow p53 inactivation target functionally relevant pathways that can shape genomic evolution and remain fixed as homogenous events in diverse malignant populations. Thus, loss of p53-the 'guardian of the genome'-is not merely a gateway to genetic chaos but, rather, can enable deterministic patterns of genome evolution that may point to new strategies for the treatment of TP53-mutant tumours.


Subject(s)
Carcinogenesis , Disease Progression , Genes, p53 , Genome , Loss of Heterozygosity , Pancreatic Neoplasms , Tumor Suppressor Protein p53 , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Evolution, Molecular , Gene Deletion , Genes, p53/genetics , Genome/genetics , Mice , Models, Genetic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
4.
Nat Commun ; 13(1): 1450, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304457

ABSTRACT

The efficacy of the highly selective RET inhibitor selpercatinib is now established in RET-driven cancers, and we sought to characterize the molecular determinants of response and resistance. We find that the pre-treatment genomic landscape does not shape the variability of treatment response except for rare instances of RAS-mediated primary resistance. By contrast, acquired selpercatinib resistance is driven by MAPK pathway reactivation by one of two distinct routes. In some patients, on- and off-target pathway reactivation via secondary RET solvent front mutations or MET amplifications are evident. In other patients, rare RET-wildtype tumor cell populations driven by an alternative mitogenic driver are selected for by treatment. Multiple distinct mechanisms are often observed in the same patient, suggesting polyclonal resistance may be common. Consequently, sequential RET-directed therapy may require combination treatment with inhibitors targeting alternative MAPK effectors, emphasizing the need for prospective characterization of selpercatinib-treated tumors at the time of monotherapy progression.


Subject(s)
Lung Neoplasms , Thyroid Neoplasms , Humans , Lung , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Prospective Studies , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics
5.
Sci Adv ; 8(7): eabi7127, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35179962

ABSTRACT

The contribution of epigenetic dysregulation to metastasis remains understudied. Through a meta-analysis of gene expression datasets followed by a mini-screen, we identified Plant Homeodomain Finger protein 8 (PHF8), a histone demethylase of the Jumonji C protein family, as a previously unidentified prometastatic gene in melanoma. Loss- and gain-of-function approaches demonstrate that PHF8 promotes cell invasion without affecting proliferation in vitro and increases dissemination but not subcutaneous tumor growth in vivo, thus supporting its specific contribution to the acquisition of metastatic potential. PHF8 requires its histone demethylase activity to enhance melanoma cell invasion. Transcriptomic and epigenomic analyses revealed that PHF8 orchestrates a molecular program that directly controls the TGFß signaling pathway and, as a consequence, melanoma invasion and metastasis. Our findings bring a mechanistic understanding of epigenetic regulation of metastatic fitness in cancer, which may pave the way for improved therapeutic interventions.


Subject(s)
Histone Demethylases , Melanoma , Cell Proliferation , Epigenesis, Genetic , Histone Demethylases/genetics , Histone Demethylases/metabolism , Homeodomain Proteins/genetics , Humans , Melanoma/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
6.
Cell Stem Cell ; 29(2): 298-314.e9, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35045331

ABSTRACT

Clonal hematopoiesis (CH) is an aging-associated condition characterized by the clonal outgrowth of pre-leukemic cells that acquire specific mutations. Although individuals with CH are healthy, they are at an increased risk of developing myeloid malignancies, suggesting that additional alterations are needed for the transition from a pre-leukemia stage to frank leukemia. To identify signaling states that cooperate with pre-leukemic cells, we used an in vivo RNAi screening approach. One of the most prominent genes identified was the ubiquitin ligase TRAF6. Loss of TRAF6 in pre-leukemic cells results in overt myeloid leukemia and is associated with MYC-dependent stem cell signatures. TRAF6 is repressed in a subset of patients with myeloid malignancies, suggesting that subversion of TRAF6 signaling can lead to acute leukemia. Mechanistically, TRAF6 ubiquitinates MYC, an event that does not affect its protein stability but rather represses its functional activity by antagonizing an acetylation modification.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/pathology , Mutation , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
7.
Stem Cell Reports ; 15(6): 1233-1245, 2020 12 08.
Article in English | MEDLINE | ID: mdl-32976761

ABSTRACT

Methylation of histone 3 at lysine 9 (H3K9) constitutes a roadblock for cellular reprogramming. Interference with methyltransferases or activation of demethylases by the cofactor ascorbic acid (AA) facilitates the derivation of induced pluripotent stem cells (iPSCs), but possible interactions between specific methyltransferases and AA treatment remain insufficiently explored. We show that chemical inhibition of the methyltransferases EHMT1 and EHMT2 counteracts iPSC formation in an enhanced reprogramming system in the presence of AA, an effect that is dependent on EHMT1. EHMT inhibition during enhanced reprogramming is associated with rapid loss of H3K9 dimethylation, inefficient downregulation of somatic genes, and failed mesenchymal-to-epithelial transition. Furthermore, transient EHMT inhibition during reprogramming yields iPSCs that fail to efficiently give rise to viable mice upon blastocyst injection. Our observations establish novel functions of H3K9 methyltransferases and suggest that a functional balance between AA-stimulated enzymes and EHMTs supports efficient and less error-prone iPSC reprogramming to pluripotency.


Subject(s)
Cellular Reprogramming , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/enzymology , Animals , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/cytology , Methylation , Mice
8.
Clin Cancer Res ; 26(7): 1624-1632, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31871300

ABSTRACT

PURPOSE: TRK inhibitors achieve marked tumor-agnostic efficacy in TRK fusion-positive cancers and consequently are now an established standard of care. Little is known, however, about the demographics, outcomes, response to alternative standard therapies, or genomic characteristics of TRK fusion-positive cancers. EXPERIMENTAL DESIGN: Utilizing a center-wide screening program involving more than 26,000 prospectively sequenced patients, genomic and clinical data from all cases with TRK fusions were extracted. An integrated analysis was performed of genomic, therapeutic, and phenomic outcomes. RESULTS: We identified 76 cases with confirmed TRK fusions (0.28% overall prevalence) involving 48 unique rearrangements and 17 cancer types. The presence of a TRK fusion was associated with depletion of concurrent oncogenic drivers (P < 0.001) and lower tumor mutation burden (P < 0.001), with the exception of colorectal cancer where TRK fusions cooccur with microsatellite instability (MSI-H). Longitudinal profiling in a subset of patients indicated that TRK fusions were present in all sampled timepoints in 82% (14/17) of cases. Progression-free survival on first-line therapy, excluding TRK inhibitors, administered for advanced disease was 9.6 months [95% confidence interval (CI), 4.8-13.2]. The best overall response rate achieved with chemotherapy containing-regimens across all lines of therapy was 63% (95% CI, 41-81). Among 12 patients treated with checkpoint inhibitors, a patient with MSI-H colorectal cancer had the only observed response. CONCLUSIONS: TRK fusion-positive cancers can respond to alternative standards of care, although efficacy of immunotherapy in the absence of other predictive biomarkers (MSI-H) appears limited. TRK fusions are present in tumors with simple genomes lacking in concurrent drivers that may partially explain the tumor-agnostic efficacy of TRK inhibitors.


Subject(s)
Membrane Glycoproteins/genetics , Mutation , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Proteins/genetics , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Child , Child, Preschool , Female , Genomics , Humans , Infant , Infant, Newborn , Male , Microsatellite Instability , Middle Aged , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/genetics , Proteins/antagonists & inhibitors , Proteins/metabolism , Young Adult
9.
Nat Immunol ; 20(9): 1196-1207, 2019 09.
Article in English | MEDLINE | ID: mdl-31406379

ABSTRACT

The response to systemic infection and injury requires the rapid adaptation of hematopoietic stem cells (HSCs), which proliferate and divert their differentiation toward the myeloid lineage. Significant interest has emerged in understanding the signals that trigger the emergency hematopoietic program. However, the mechanisms that halt this response of HSCs, which is critical to restore homeostasis, remain unknown. Here we reveal that the E3 ubiquitin ligase Speckle-type BTB-POZ protein (SPOP) restrains the inflammatory activation of HSCs. In the absence of Spop, systemic inflammation proceeded in an unresolved manner, and the sustained response in the HSCs resulted in a lethal phenotype reminiscent of hyper-inflammatory syndrome or sepsis. Our proteomic studies decipher that SPOP restricted inflammation by ubiquitinating the innate signal transducer myeloid differentiation primary response protein 88 (MYD88). These findings unearth an HSC-intrinsic post-translational mechanism that is essential for reestablishing homeostasis after emergency hematopoiesis.


Subject(s)
Inflammation/immunology , Leukocytosis/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophils/immunology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Female , HEK293 Cells , Hematopoiesis/immunology , Humans , Male , Mice , Neutrophils/cytology , Ubiquitin-Protein Ligase Complexes , Ubiquitin-Protein Ligases/metabolism
10.
Cancer Discov ; 9(7): 890-909, 2019 07.
Article in English | MEDLINE | ID: mdl-31048321

ABSTRACT

The BCL2 family plays important roles in acute myeloid leukemia (AML). Venetoclax, a selective BCL2 inhibitor, has received FDA approval for the treatment of AML. However, drug resistance ensues after prolonged treatment, highlighting the need for a greater understanding of the underlying mechanisms. Using a genome-wide CRISPR/Cas9 screen in human AML, we identified genes whose inactivation sensitizes AML blasts to venetoclax. Genes involved in mitochondrial organization and function were significantly depleted throughout our screen, including the mitochondrial chaperonin CLPB. We demonstrated that CLPB is upregulated in human AML, it is further induced upon acquisition of venetoclax resistance, and its ablation sensitizes AML to venetoclax. Mechanistically, CLPB maintains the mitochondrial cristae structure via its interaction with the cristae-shaping protein OPA1, whereas its loss promotes apoptosis by inducing cristae remodeling and mitochondrial stress responses. Overall, our data suggest that targeting mitochondrial architecture may provide a promising approach to circumvent venetoclax resistance. SIGNIFICANCE: A genome-wide CRISPR/Cas9 screen reveals genes involved in mitochondrial biological processes participate in the acquisition of venetoclax resistance. Loss of the mitochondrial protein CLPB leads to structural and functional defects of mitochondria, hence sensitizing AML cells to apoptosis. Targeting CLPB synergizes with venetoclax and the venetoclax/azacitidine combination in AML in a p53-independent manner.See related commentary by Savona and Rathmell, p. 831.This article is highlighted in the In This Issue feature, p. 813.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mitochondria/drug effects , Mitochondria/genetics , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Resistance, Neoplasm , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/metabolism , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Mitochondria/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays
11.
Nat Med ; 24(8): 1157-1166, 2018 08.
Article in English | MEDLINE | ID: mdl-30038221

ABSTRACT

Cellular transformation is accompanied by extensive rewiring of many biological processes leading to augmented levels of distinct types of cellular stress, including proteotoxic stress. Cancer cells critically depend on stress-relief pathways for their survival. However, the mechanisms underlying the transcriptional initiation and maintenance of the oncogenic stress response remain elusive. Here, we show that the expression of heat shock transcription factor 1 (HSF1) and the downstream mediators of the heat shock response is transcriptionally upregulated in T cell acute lymphoblastic leukemia (T-ALL). Hsf1 ablation suppresses the growth of human T-ALL and eradicates leukemia in mouse models of T-ALL, while sparing normal hematopoiesis. HSF1 drives a compact transcriptional program and among the direct HSF1 targets, specific chaperones and co-chaperones mediate its critical role in T-ALL. Notably, we demonstrate that the central T-ALL oncogene NOTCH1 hijacks the cellular stress response machinery by inducing the expression of HSF1 and its downstream effectors. The NOTCH1 signaling status controls the levels of chaperone/co-chaperone complexes and predicts the response of T-ALL patient samples to HSP90 inhibition. Our data demonstrate an integral crosstalk between mediators of oncogene and non-oncogene addiction and reveal critical nodes of the heat shock response pathway that can be targeted therapeutically.


Subject(s)
Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Stress, Physiological , Animals , Cell Line, Tumor , Gene Expression Regulation, Leukemic , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Heat-Shock Response , Hematopoiesis , Humans , Mice, Inbred C57BL , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Notch/metabolism , Signal Transduction
12.
Nat Commun ; 9(1): 542, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416042

ABSTRACT

The metazoan genome is compartmentalized in areas of highly interacting chromatin known as topologically associating domains (TADs). TADs are demarcated by boundaries mostly conserved across cell types and even across species. However, a genome-wide characterization of TAD boundary strength in mammals is still lacking. In this study, we first use fused two-dimensional lasso as a machine learning method to improve Hi-C contact matrix reproducibility, and, subsequently, we categorize TAD boundaries based on their insulation score. We demonstrate that higher TAD boundary insulation scores are associated with elevated CTCF levels and that they may differ across cell types. Intriguingly, we observe that super-enhancers are preferentially insulated by strong boundaries. Furthermore, we demonstrate that strong TAD boundaries and super-enhancer elements are frequently co-duplicated in cancer patients. Taken together, our findings suggest that super-enhancers insulated by strong TAD boundaries may be exploited, as a functional unit, by cancer cells to promote oncogenesis.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Insulator Elements/genetics , Neoplasms/genetics , Animals , CCCTC-Binding Factor , Chromatin , Epigenomics , Humans , Machine Learning
13.
BMC Genomics ; 18(1): 434, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28583068

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) have emerged as a class of factors that are important for regulating development and cancer. Computational prediction of lncRNAs from ultra-deep RNA sequencing has been successful in identifying candidate lncRNAs. However, the complexity of handling and integrating different types of genomics data poses significant challenges to experimental laboratories that lack extensive genomics expertise. RESULT: To address this issue, we have developed lncRNA-screen, a comprehensive pipeline for computationally screening putative lncRNA transcripts over large multimodal datasets. The main objective of this work is to facilitate the computational discovery of lncRNA candidates to be further examined by functional experiments. lncRNA-screen provides a fully automated easy-to-run pipeline which performs data download, RNA-seq alignment, assembly, quality assessment, transcript filtration, novel lncRNA identification, coding potential estimation, expression level quantification, histone mark enrichment profile integration, differential expression analysis, annotation with other type of segmented data (CNVs, SNPs, Hi-C, etc.) and visualization. Importantly, lncRNA-screen generates an interactive report summarizing all interesting lncRNA features including genome browser snapshots and lncRNA-mRNA interactions based on Hi-C data. CONCLUSION: lncRNA-screen provides a comprehensive solution for lncRNA discovery and an intuitive interactive report for identifying promising lncRNA candidates. lncRNA-screen is available as open-source software on GitHub.


Subject(s)
Databases, Genetic , Genomics/methods , RNA, Long Noncoding/genetics , User-Computer Interface , Data Mining , Epigenomics , Gene Expression Profiling , Histones/metabolism , Sequence Alignment , Software
14.
J Exp Med ; 212(11): 1833-50, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26438359

ABSTRACT

The cohesin complex (consisting of Rad21, Smc1a, Smc3, and Stag2 proteins) is critically important for proper sister chromatid separation during mitosis. Mutations in the cohesin complex were recently identified in a variety of human malignancies including acute myeloid leukemia (AML). To address the potential tumor-suppressive function of cohesin in vivo, we generated a series of shRNA mouse models in which endogenous cohesin can be silenced inducibly. Notably, silencing of cohesin complex members did not have a deleterious effect on cell viability. Furthermore, knockdown of cohesin led to gain of replating capacity of mouse hematopoietic progenitor cells. However, cohesin silencing in vivo rapidly altered stem cells homeostasis and myelopoiesis. Likewise, we found widespread changes in chromatin accessibility and expression of genes involved in myelomonocytic maturation and differentiation. Finally, aged cohesin knockdown mice developed a clinical picture closely resembling myeloproliferative disorders/neoplasms (MPNs), including varying degrees of extramedullary hematopoiesis (myeloid metaplasia) and splenomegaly. Our results represent the first successful demonstration of a tumor suppressor function for the cohesin complex, while also confirming that cohesin mutations occur as an early event in leukemogenesis, facilitating the potential development of a myeloid malignancy.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Proteins, Non-Histone/physiology , Hematopoietic Stem Cells/physiology , Homeostasis , Myeloproliferative Disorders/etiology , Tumor Suppressor Proteins/physiology , Adult , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Chromatin/physiology , Chromosomal Proteins, Non-Histone/genetics , Humans , Mice , Cohesins
15.
Cancer Cell ; 27(6): 755-68, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26058075

ABSTRACT

The role of the microenvironment in T cell acute lymphoblastic leukemia (T-ALL), or any acute leukemia, is poorly understood. Here we demonstrate that T-ALL cells are in direct, stable contact with CXCL12-producing bone marrow stroma. Cxcl12 deletion from vascular endothelial, but not perivascular, cells impeded tumor growth, suggesting a vascular niche for T-ALL. Moreover, genetic targeting of Cxcr4 in murine T-ALL after disease onset led to rapid, sustained disease remission, and CXCR4 antagonism suppressed human T-ALL in primary xenografts. Loss of CXCR4 targeted key T-ALL regulators, including the MYC pathway, and decreased leukemia initiating cell activity in vivo. Our data identify a T-ALL niche and suggest targeting CXCL12/CXCR4 signaling as a powerful therapeutic approach for T-ALL.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/biosynthesis , Endothelium, Vascular/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Pyridines/pharmacology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Chemokine CXCL12/genetics , Endothelium, Vascular/pathology , Female , Gene Deletion , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...