Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nature ; 615(7952): 499-506, 2023 03.
Article in English | MEDLINE | ID: mdl-36890229

ABSTRACT

Mutations in fumarate hydratase (FH) cause hereditary leiomyomatosis and renal cell carcinoma1. Loss of FH in the kidney elicits several oncogenic signalling cascades through the accumulation of the oncometabolite fumarate2. However, although the long-term consequences of FH loss have been described, the acute response has not so far been investigated. Here we generated an inducible mouse model to study the chronology of FH loss in the kidney. We show that loss of FH leads to early alterations of mitochondrial morphology and the release of mitochondrial DNA (mtDNA) into the cytosol, where it triggers the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-TANK-binding kinase 1 (TBK1) pathway and stimulates an inflammatory response that is also partially dependent on retinoic-acid-inducible gene I (RIG-I). Mechanistically, we show that this phenotype is mediated by fumarate and occurs selectively through mitochondrial-derived vesicles in a manner that depends on sorting nexin 9 (SNX9). These results reveal that increased levels of intracellular fumarate induce a remodelling of the mitochondrial network and the generation of mitochondrial-derived vesicles, which allows the release of mtDNAin the cytosol and subsequent activation of the innate immune response.


Subject(s)
DNA, Mitochondrial , Fumarates , Immunity, Innate , Mitochondria , Animals , Mice , DNA, Mitochondrial/metabolism , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Fumarates/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Kidney/enzymology , Kidney/metabolism , Kidney/pathology , Cytosol/metabolism
2.
Nature ; 606(7916): 999-1006, 2022 06.
Article in English | MEDLINE | ID: mdl-35676472

ABSTRACT

Large-scale human genetic data1-3 have shown that cancer mutations display strong tissue-selectivity, but how this selectivity arises remains unclear. Here, using experimental models, functional genomics and analyses of patient samples, we demonstrate that the lineage transcription factor paired box 8 (PAX8) is required for oncogenic signalling by two common genetic alterations that cause clear cell renal cell carcinoma (ccRCC) in humans: the germline variant rs7948643 at 11q13.3 and somatic inactivation of the von Hippel-Lindau tumour suppressor (VHL)4-6. VHL loss, which is observed in about 90% of ccRCCs, can lead to hypoxia-inducible factor 2α (HIF2A) stabilization6,7. We show that HIF2A is preferentially recruited to PAX8-bound transcriptional enhancers, including a pro-tumorigenic cyclin D1 (CCND1) enhancer that is controlled by PAX8 and HIF2A. The ccRCC-protective allele C at rs7948643 inhibits PAX8 binding at this enhancer and downstream activation of CCND1 expression. Co-option of a PAX8-dependent physiological programme that supports the proliferation of normal renal epithelial cells is also required for MYC expression from the ccRCC metastasis-associated amplicons at 8q21.3-q24.3 (ref. 8). These results demonstrate that transcriptional lineage factors are essential for oncogenic signalling and that they mediate tissue-specific cancer risk associated with somatic and inherited genetic variants.


Subject(s)
Carcinogenesis , Kidney Neoplasms , PAX8 Transcription Factor , Signal Transduction , Alleles , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mutation , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Proto-Oncogene Proteins c-myc/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
3.
Genes Dev ; 31(13): 1339-1353, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28790158

ABSTRACT

Lung adenocarcinoma accounts for ∼40% of lung cancers, the leading cause of cancer-related death worldwide, and current therapies provide only limited survival benefit. Approximately half of lung adenocarcinomas harbor mutations in TP53 (p53), making these mutants appealing targets for lung cancer therapy. As mutant p53 remains untargetable, mutant p53-dependent phenotypes represent alternative targeting opportunities, but the prevalence and therapeutic relevance of such effects (gain of function and dominant-negative activity) in lung adenocarcinoma are unclear. Through transcriptional and functional analysis of murine KrasG12D -p53null , -p53R172H (conformational), and -p53R270H (contact) mutant lung tumors, we identified genotype-independent and genotype-dependent therapeutic sensitivities. Unexpectedly, we found that wild-type p53 exerts a dominant tumor-suppressive effect on mutant tumors, as all genotypes were similarly sensitive to its restoration in vivo. These data show that the potential of p53 targeted therapies is comparable across all p53-deficient genotypes and may explain the high incidence of p53 loss of heterozygosity in mutant tumors. In contrast, mutant p53 gain of function and their associated vulnerabilities can vary according to mutation type. Notably, we identified a p53R270H -specific sensitivity to simvastatin in lung tumors, and the transcriptional signature that underlies this sensitivity was also present in human lung tumors, indicating that this therapeutic approach may be clinically relevant.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Simvastatin/therapeutic use , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/genetics , Cell Death/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Genotype , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacology , Mice , Molecular Targeted Therapy , Mutation , Piperazines/pharmacology , Simvastatin/pharmacology
4.
Nat Commun ; 15(1): 5386, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918386

ABSTRACT

Aberrantly accumulated metabolites elicit intra- and inter-cellular pro-oncogenic cascades, yet current measurement methods require sample perturbation/disruption and lack spatio-temporal resolution, limiting our ability to fully characterize their function and distribution. Here, we show that Raman spectroscopy (RS) can directly detect fumarate in living cells in vivo and animal tissues ex vivo, and that RS can distinguish between Fumarate hydratase (Fh1)-deficient and Fh1-proficient cells based on fumarate concentration. Moreover, RS reveals the spatial compartmentalization of fumarate within cellular organelles in Fh1-deficient cells: consistent with disruptive methods, we observe the highest fumarate concentration (37 ± 19 mM) in mitochondria, where the TCA cycle operates, followed by the cytoplasm (24 ± 13 mM) and then the nucleus (9 ± 6 mM). Finally, we apply RS to tissues from an inducible mouse model of FH loss in the kidney, demonstrating RS can classify FH status. These results suggest RS could be adopted as a valuable tool for small molecule metabolic imaging, enabling in situ non-destructive evaluation of fumarate compartmentalization.


Subject(s)
Fumarate Hydratase , Fumarates , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Animals , Fumarates/metabolism , Mice , Fumarate Hydratase/metabolism , Fumarate Hydratase/genetics , Kidney/metabolism , Mitochondria/metabolism , Humans , Cell Nucleus/metabolism , Cytoplasm/metabolism
5.
Sci Adv ; 8(42): eabq8297, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36269833

ABSTRACT

Fumarate hydratase (FH) is a mitochondrial enzyme that catalyzes the reversible hydration of fumarate to malate in the tricarboxylic acid (TCA) cycle. Germline mutations of FH lead to hereditary leiomyomatosis and renal cell carcinoma (HLRCC), a cancer syndrome characterized by a highly aggressive form of renal cancer. Although HLRCC tumors metastasize rapidly, FH-deficient mice develop premalignant cysts in the kidneys, rather than carcinomas. How Fh1-deficient cells overcome these tumor-suppressive events during transformation is unknown. Here, we perform a genome-wide CRISPR-Cas9 screen to identify genes that, when ablated, enhance the proliferation of Fh1-deficient cells. We found that the depletion of the histone cell cycle regulator (HIRA) enhances proliferation and invasion of Fh1-deficient cells in vitro and in vivo. Mechanistically, Hira loss activates MYC and its target genes, increasing nucleotide metabolism specifically in Fh1-deficient cells, independent of its histone chaperone activity. These results are instrumental for understanding mechanisms of tumorigenesis in HLRCC and the development of targeted treatments for patients.

SELECTION OF CITATIONS
SEARCH DETAIL