Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 173(2): 321-337.e10, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29625050

RESUMEN

Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFß signaling, p53 and ß-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy.


Asunto(s)
Bases de Datos Genéticas , Neoplasias/patología , Transducción de Señal/genética , Genes Relacionados con las Neoplasias , Humanos , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
2.
Mol Cell ; 77(3): 645-655.e7, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31983508

RESUMEN

The lysosome is an acidic multi-functional organelle with roles in macromolecular digestion, nutrient sensing, and signaling. However, why cells require acidic lysosomes to proliferate and which nutrients become limiting under lysosomal dysfunction are unclear. To address this, we performed CRISPR-Cas9-based genetic screens and identified cholesterol biosynthesis and iron uptake as essential metabolic pathways when lysosomal pH is altered. While cholesterol synthesis is only necessary, iron is both necessary and sufficient for cell proliferation under lysosomal dysfunction. Remarkably, iron supplementation restores cell proliferation under both pharmacologic and genetic-mediated lysosomal dysfunction. The rescue was independent of metabolic or signaling changes classically associated with increased lysosomal pH, uncoupling lysosomal function from cell proliferation. Finally, our experiments revealed that lysosomal dysfunction dramatically alters mitochondrial metabolism and hypoxia inducible factor (HIF) signaling due to iron depletion. Altogether, these findings identify iron homeostasis as the key function of lysosomal acidity for cell proliferation.


Asunto(s)
Proliferación Celular/fisiología , Hierro/metabolismo , Lisosomas/metabolismo , Colesterol/biosíntesis , Colesterol/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Células Jurkat , Lisosomas/fisiología , Mitocondrias/metabolismo , Transducción de Señal/genética
3.
Mol Cell ; 74(1): 45-58.e7, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30846317

RESUMEN

Cells require a constant supply of fatty acids to survive and proliferate. Fatty acids incorporate into membrane and storage glycerolipids through a series of endoplasmic reticulum (ER) enzymes, but how these enzymes are regulated is not well understood. Here, using a combination of CRISPR-based genetic screens and unbiased lipidomics, we identified calcineurin B homologous protein 1 (CHP1) as a major regulator of ER glycerolipid synthesis. Loss of CHP1 severely reduces fatty acid incorporation and storage in mammalian cells and invertebrates. Mechanistically, CHP1 binds and activates GPAT4, which catalyzes the initial rate-limiting step in glycerolipid synthesis. GPAT4 activity requires CHP1 to be N-myristoylated, forming a key molecular interface between the two proteins. Interestingly, upon CHP1 loss, the peroxisomal enzyme, GNPAT, partially compensates for the loss of ER lipid synthesis, enabling cell proliferation. Thus, our work identifies a conserved regulator of glycerolipid metabolism and reveals plasticity in lipid synthesis of proliferating cells.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Retículo Endoplásmico/enzimología , Glicéridos/biosíntesis , Glicerol-3-Fosfato O-Aciltransferasa/metabolismo , Lipogénesis , Células 3T3 , Aciltransferasas/genética , Aciltransferasas/metabolismo , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al Calcio/genética , Proliferación Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/patología , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Glicerol-3-Fosfato O-Aciltransferasa/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Células Jurkat , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Ratones , Ácido Palmítico/toxicidad , Unión Proteica
4.
Nature ; 567(7746): 118-122, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30760928

RESUMEN

Cholesterol is essential for cells to grow and proliferate. Normal mammalian cells meet their need for cholesterol through its uptake or de novo synthesis1, but the extent to which cancer cells rely on each of these pathways remains poorly understood. Here, using a competitive proliferation assay on a pooled collection of DNA-barcoded cell lines, we identify a subset of cancer cells that is auxotrophic for cholesterol and thus highly dependent on its uptake. Through metabolic gene expression analysis, we pinpoint the loss of squalene monooxygenase expression as a cause of cholesterol auxotrophy, particularly in ALK+ anaplastic large cell lymphoma (ALCL) cell lines and primary tumours. Squalene monooxygenase catalyses the oxidation of squalene to 2,3-oxidosqualene in the cholesterol synthesis pathway and its loss results in accumulation of the upstream metabolite squalene, which is normally undetectable. In ALK+ ALCLs, squalene alters the cellular lipid profile and protects cancer cells from ferroptotic cell death, providing a growth advantage under conditions of oxidative stress and in tumour xenografts. Finally, a CRISPR-based genetic screen identified cholesterol uptake by the low-density lipoprotein receptor as essential for the growth of ALCL cells in culture and as patient-derived xenografts. This work reveals that the cholesterol auxotrophy of ALCLs is a targetable liability and, more broadly, that systematic approaches can be used to identify nutrient dependencies unique to individual cancer types.


Asunto(s)
Apoptosis , Colesterol/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Linfoma Anaplásico de Células Grandes/patología , Estrés Oxidativo , Escualeno/metabolismo , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular , Colesterol/biosíntesis , Código de Barras del ADN Taxonómico , Farnesil Difosfato Farnesil Transferasa/genética , Farnesil Difosfato Farnesil Transferasa/metabolismo , Femenino , Humanos , Hierro/metabolismo , Linfoma Anaplásico de Células Grandes/enzimología , Masculino , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Receptores de LDL/genética , Receptores de LDL/metabolismo , Escualeno-Monooxigenasa/genética , Escualeno-Monooxigenasa/metabolismo , Adulto Joven
5.
Nat Chem Biol ; 16(12): 1351-1360, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32778843

RESUMEN

Cancer cells rewire their metabolism and rely on endogenous antioxidants to mitigate lethal oxidative damage to lipids. However, the metabolic processes that modulate the response to lipid peroxidation are poorly defined. Using genetic screens, we compared metabolic genes essential for proliferation upon inhibition of cystine uptake or glutathione peroxidase-4 (GPX4). Interestingly, very few genes were commonly required under both conditions, suggesting that cystine limitation and GPX4 inhibition may impair proliferation via distinct mechanisms. Our screens also identify tetrahydrobiopterin (BH4) biosynthesis as an essential metabolic pathway upon GPX4 inhibition. Mechanistically, BH4 is a potent radical-trapping antioxidant that protects lipid membranes from autoxidation, alone and in synergy with vitamin E. Dihydrofolate reductase catalyzes the regeneration of BH4, and its inhibition by methotrexate synergizes with GPX4 inhibition. Altogether, our work identifies the mechanism by which BH4 acts as an endogenous antioxidant and provides a compendium of metabolic modifiers of lipid peroxidation.


Asunto(s)
Cistina/metabolismo , Ferroptosis/genética , Regulación Neoplásica de la Expresión Génica , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Tetrahidrofolato Deshidrogenasa/genética , Antineoplásicos/farmacología , Antioxidantes/farmacología , Biopterinas/análogos & derivados , Biopterinas/farmacología , Carbolinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cistina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Ferroptosis/efectos de los fármacos , Antagonistas del Ácido Fólico/farmacología , Perfilación de la Expresión Génica , Humanos , Células Jurkat , Peroxidación de Lípido/efectos de los fármacos , Metotrexato/farmacología , Estrés Oxidativo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Piperazinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Tetrahidrofolato Deshidrogenasa/metabolismo , Vitamina E/farmacología
6.
Proc Natl Acad Sci U S A ; 114(25): E4951-E4960, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28584132

RESUMEN

Oncogenic RAS mutations are present in 15-30% of thyroid carcinomas. Endogenous expression of mutant Ras is insufficient to initiate thyroid tumorigenesis in murine models, indicating that additional genetic alterations are required. We used Sleeping Beauty (SB) transposon mutagenesis to identify events that cooperate with HrasG12V in thyroid tumor development. Random genomic integration of SB transposons primarily generated loss-of-function events that significantly increased thyroid tumor penetrance in Tpo-Cre/homozygous FR-HrasG12V mice. The thyroid tumors closely phenocopied the histological features of human RAS-driven, poorly differentiated thyroid cancers. Characterization of transposon insertion sites in the SB-induced tumors identified 45 recurrently mutated candidate cancer genes. These mutation profiles were remarkably concordant with mutated cancer genes identified in a large series of human poorly differentiated and anaplastic thyroid cancers screened by next-generation sequencing using the MSK-IMPACT panel of cancer genes, which we modified to include all SB candidates. The disrupted genes primarily clustered in chromatin remodeling functional nodes and in the PI3K pathway. ATXN7, a component of a multiprotein complex with histone acetylase activity, scored as a significant SB hit. It was recurrently mutated in advanced human cancers and significantly co-occurred with RAS or NF1 mutations. Expression of ATXN7 mutants cooperated with oncogenic RAS to induce thyroid cell proliferation, pointing to ATXN7 as a previously unrecognized cancer gene.


Asunto(s)
Ataxina-7/genética , Carcinogénesis/genética , Cromatina/genética , Elementos Transponibles de ADN/genética , Genes ras/genética , Mutagénesis/genética , Glándula Tiroides/patología , Animales , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Oncogenes/genética , Fosfatidilinositol 3-Quinasas/genética , Carcinoma Anaplásico de Tiroides/genética
7.
Bioinformatics ; 33(14): 2238-2240, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334343

RESUMEN

MOTIVATION: While existing network visualization tools enable the exploration of cancer genomics data, most biologists prefer simplified, curated pathway diagrams, such as those featured in many manuscripts from The Cancer Genome Atlas (TCGA). These pathway diagrams typically summarize how a pathway is altered in individual cancer types, including alteration frequencies for each gene. RESULTS: To address this need, we developed the web-based tool PathwayMapper, which runs in most common web browsers. It can be used for viewing pre-curated cancer pathways, or as a graphical editor for creating new pathways, with the ability to overlay genomic alteration data from cBioPortal. In addition, a collaborative mode is available that allows scientists to co-operate interactively on constructing pathways, with support for concurrent modifications and built-in conflict resolution. AVAILABILITY AND IMPLEMENTATION: The PathwayMapper tool is accessible at http://pathwaymapper.org and the code is available on Github ( https://github.com/iVis-at-Bilkent/pathway-mapper ). CONTACT: ivis@cs.bilkent.edu.tr. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genómica/métodos , Redes y Vías Metabólicas , Neoplasias/metabolismo , Transducción de Señal , Programas Informáticos , Humanos
8.
Dev Biol ; 409(1): 297-309, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26569556

RESUMEN

Hox genes play crucial roles in establishing regional identity along the anterior-posterior axis in bilaterian animals, and have been implicated in generating morphological diversity throughout evolution. Here we report the identification, expression, and initial genomic characterization of the complete set of Hox genes from the amphipod crustacean Parhyale hawaiensis. Parhyale is an emerging model system that is amenable to experimental manipulations and evolutionary comparisons among the arthropods. Our analyses indicate that the Parhyale genome contains a single copy of each canonical Hox gene with the exception of fushi tarazu, and preliminary mapping suggests that at least some of these genes are clustered together in the genome. With few exceptions, Parhyale Hox genes exhibit both temporal and spatial colinearity, and expression boundaries correlate with morphological differences between segments and their associated appendages. This work represents the most comprehensive analysis of Hox gene expression in a crustacean to date, and provides a foundation for functional studies aimed at elucidating the role of Hox genes in arthropod development and evolution.


Asunto(s)
Anfípodos/embriología , Anfípodos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Animales , Sistemas CRISPR-Cas/genética , Mapeo Cromosómico , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Genes Reporteros , Genoma , Proteínas Fluorescentes Verdes/metabolismo , Cabeza/embriología , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Modelos Biológicos , Especificidad de Órganos/genética , Tórax/embriología , Tórax/metabolismo
9.
bioRxiv ; 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38765991

RESUMEN

Lipids are essential for tumours because of their structural, energetic, and signaling roles. While many cancer cells upregulate lipid synthesis, growing evidence suggests that tumours simultaneously intensify the uptake of circulating lipids carried by lipoproteins. Which mechanisms promote the uptake of extracellular lipids, and how this pool of lipids contributes to cancer progression, are poorly understood. Here, using functional genetic screens, we find that lipoprotein uptake confers resistance to lipid peroxidation and ferroptotic cell death. Lipoprotein supplementation robustly inhibits ferroptosis across numerous cancer types. Mechanistically, cancer cells take up lipoproteins through a pathway dependent on sulfated glycosaminoglycans (GAGs) linked to cell-surface proteoglycans. Tumour GAGs are a major determinant of the uptake of both low and high density lipoproteins. Impairment of glycosaminoglycan synthesis or acute degradation of surface GAGs decreases the uptake of lipoproteins, sensitizes cells to ferroptosis and reduces tumour growth in mice. We also find that human clear cell renal cell carcinomas, a distinctively lipid-rich tumour type, display elevated levels of lipoprotein-derived antioxidants and the GAG chondroitin sulfate than non-malignant human kidney. Altogether, our work identifies lipoprotein uptake as an essential anti-ferroptotic mechanism for cancer cells to overcome lipid oxidative stress in vivo, and reveals GAG biosynthesis as an unexpected mediator of this process.

10.
Nat Metab ; 4(6): 724-738, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35726024

RESUMEN

Stress-adaptive mechanisms enable tumour cells to overcome metabolic constraints under nutrient and oxygen shortage. Aspartate is an endogenous metabolic limitation under hypoxic conditions, but the nature of the adaptive mechanisms that contribute to aspartate availability and hypoxic tumour growth are poorly understood. Here we identify GOT2-catalysed mitochondrial aspartate synthesis as an essential metabolic dependency for the proliferation of pancreatic tumour cells under hypoxic culture conditions. In contrast, GOT2-catalysed aspartate synthesis is dispensable for pancreatic tumour formation in vivo. The dependence of pancreatic tumour cells on aspartate synthesis is bypassed in part by a hypoxia-induced potentiation of extracellular protein scavenging via macropinocytosis. This effect is mutant KRAS dependent, and is mediated by hypoxia-inducible factor 1 (HIF1A) and its canonical target carbonic anhydrase-9 (CA9). Our findings reveal high plasticity of aspartate metabolism and define an adaptive regulatory role for macropinocytosis by which mutant KRAS tumours can overcome nutrient deprivation under hypoxic conditions.


Asunto(s)
Ácido Aspártico , Neoplasias Pancreáticas , Línea Celular Tumoral , Humanos , Hipoxia , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética
11.
Nat Metab ; 3(4): 558-570, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33833465

RESUMEN

Mitochondrial DNA (mtDNA) encodes protein subunits and translational machinery required for oxidative phosphorylation (OXPHOS). Using repurposed whole-exome sequencing data, in the present study we demonstrate that pathogenic mtDNA mutations arise in tumours at a rate comparable to those in the most common cancer driver genes. We identify OXPHOS complexes as critical determinants shaping somatic mtDNA mutation patterns across tumour lineages. Loss-of-function mutations accumulate at an elevated rate specifically in complex I and often arise at specific homopolymeric hotspots. In contrast, complex V is depleted of all non-synonymous mutations, suggesting that impairment of ATP synthesis and mitochondrial membrane potential dissipation are under negative selection. Common truncating mutations and rarer missense alleles are both associated with a pan-lineage transcriptional programme, even in cancer types where mtDNA mutations are comparatively rare. Pathogenic mutations of mtDNA are associated with substantial increases in overall survival of colorectal cancer patients, demonstrating a clear functional relationship between genotype and phenotype. The mitochondrial genome is therefore frequently and functionally disrupted across many cancers, with major implications for patient stratification, prognosis and therapeutic development.


Asunto(s)
Linaje de la Célula/genética , ADN Mitocondrial/genética , ADN de Neoplasias/genética , Fosforilación Oxidativa , Adenosina Trifosfato/biosíntesis , Neoplasias Colorrectales/genética , Exoma/genética , Genoma Humano/genética , Genoma Mitocondrial , Genotipo , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/genética , Mutación/genética , Fenotipo , ARN/genética
12.
Sci Adv ; 6(41)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33036978

RESUMEN

Tumor environment influences anticancer therapy response but which extracellular nutrients affect drug sensitivity is largely unknown. Using functional genomics, we determine modifiers of l-asparaginase (ASNase) response and identify thiamine pyrophosphate kinase 1 as a metabolic dependency under ASNase treatment. While thiamine is generally not limiting for cell proliferation, a DNA-barcode competition assay identifies leukemia cell lines that grow suboptimally under low thiamine and are characterized by low expression of solute carrier family 19 member 2 (SLC19A2), a thiamine transporter. SLC19A2 is necessary for optimal growth and ASNase resistance, when standard medium thiamine is lowered ~100-fold to human plasma concentrations. In addition, humanizing blood thiamine content of mice through diet sensitizes SLC19A2-low leukemia cells to ASNase in vivo. Together, our work reveals that thiamine utilization is a determinant of ASNase response for some cancer cells and that oversupplying vitamins may affect therapeutic response in leukemia.


Asunto(s)
Antineoplásicos , Leucemia , Animales , Antineoplásicos/uso terapéutico , Asparaginasa/metabolismo , Asparaginasa/farmacología , Asparaginasa/uso terapéutico , Dieta , Leucemia/tratamiento farmacológico , Proteínas de Transporte de Membrana , Ratones , Tiamina/farmacología
13.
Nat Metab ; 2(6): 487-498, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32694732

RESUMEN

Coessentiality mapping has been useful to systematically cluster genes into biological pathways and identify gene functions1-3. Here, using the debiased sparse partial correlation (DSPC) method3, we construct a functional coessentiality map for cellular metabolic processes across human cancer cell lines. This analysis reveals 35 modules associated with known metabolic pathways and further assigns metabolic functions to unknown genes. In particular, we identify C12orf49 as an essential regulator of cholesterol and fatty acid metabolism in mammalian cells. Mechanistically, C12orf49 localizes to the Golgi, binds membrane-bound transcription factor peptidase, site 1 (MBTPS1, site 1 protease) and is necessary for the cleavage of its substrates, including sterol regulatory element binding protein (SREBP) transcription factors. This function depends on the evolutionarily conserved uncharacterized domain (DUF2054) and promotes cell proliferation under cholesterol depletion. Notably, c12orf49 depletion in zebrafish blocks dietary lipid clearance in vivo, mimicking the phenotype of mbtps1 mutants. Finally, in an electronic health record (EHR)-linked DNA biobank, C12orf49 is associated with hyperlipidaemia through phenome analysis. Altogether, our findings reveal a conserved role for C12orf49 in cholesterol and lipid homeostasis and provide a platform to identify unknown components of other metabolic pathways.


Asunto(s)
Colesterol/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Línea Celular , Proliferación Celular , Regulación de la Expresión Génica , Aparato de Golgi/metabolismo , Humanos , Hiperlipidemias/genética , Metabolismo de los Lípidos/genética , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Pez Cebra
14.
Cell Metab ; 31(4): 852-861.e6, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268116

RESUMEN

Activating transcription factor 4 (ATF4) is a master transcriptional regulator of the integrated stress response (ISR) that enables cell survival under nutrient stress. The mechanisms by which ATF4 couples metabolic stresses to specific transcriptional outputs remain unknown. Using functional genomics, we identified transcription factors that regulate the responses to distinct amino acid deprivation conditions. While ATF4 is universally required under amino acid starvation, our screens yielded a transcription factor, Zinc Finger and BTB domain-containing protein 1 (ZBTB1), as uniquely essential under asparagine deprivation. ZBTB1 knockout cells are unable to synthesize asparagine due to reduced expression of asparagine synthetase (ASNS), the enzyme responsible for asparagine synthesis. Mechanistically, ZBTB1 binds to the ASNS promoter and promotes ASNS transcription. Finally, loss of ZBTB1 sensitizes therapy-resistant T cell leukemia cells to L-asparaginase, a chemotherapeutic that depletes serum asparagine. Our work reveals a critical regulator of the nutrient stress response that may be of therapeutic value.


Asunto(s)
Asparagina/biosíntesis , Aspartatoamoníaco Ligasa/metabolismo , Leucemia , Proteínas Represoras/fisiología , Animales , Asparagina/deficiencia , Línea Celular Tumoral , Proliferación Celular , Regulación de la Expresión Génica , Humanos , Leucemia/metabolismo , Leucemia/patología , Ratones Endogámicos NOD , Ratones SCID , Transcripción Genética
15.
Cell Syst ; 6(3): 301-313.e3, 2018 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-29396322

RESUMEN

Tumor metabolism is reorganized to support proliferation in the face of growth-related stress. Unlike the widespread profiling of changes to metabolic enzyme levels in cancer, comparatively less attention has been paid to the substrates/products of enzyme-catalyzed reactions, small-molecule metabolites. We developed an informatic pipeline to concurrently analyze metabolomics data from over 900 tissue samples spanning seven cancer types, revealing extensive heterogeneity in metabolic changes relative to normal tissue across cancers of different tissues of origin. Despite this heterogeneity, a number of metabolites were recurrently differentially abundant across many cancers, such as lactate and acyl-carnitine species. Through joint analysis of metabolomic data alongside clinical features of patient samples, we also identified a small number of metabolites, including several polyamines and kynurenine, which were associated with aggressive tumors across several tumor types. Our findings offer a glimpse onto common patterns of metabolic reprogramming across cancers, and the work serves as a large-scale resource accessible via a web application (http://www.sanderlab.org/pancanmet).


Asunto(s)
Biología Computacional/métodos , Metabolómica/métodos , Neoplasias/metabolismo , Algoritmos , Humanos , Programas Informáticos
16.
Nat Cell Biol ; 20(7): 775-781, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29941933

RESUMEN

As oxygen is essential for many metabolic pathways, tumour hypoxia may impair cancer cell proliferation1-4. However, the limiting metabolites for proliferation under hypoxia and in tumours are unknown. Here, we assessed proliferation of a collection of cancer cells following inhibition of the mitochondrial electron transport chain (ETC), a major metabolic pathway requiring molecular oxygen5. Sensitivity to ETC inhibition varied across cell lines, and subsequent metabolomic analysis uncovered aspartate availability as a major determinant of sensitivity. Cell lines least sensitive to ETC inhibition maintain aspartate levels by importing it through an aspartate/glutamate transporter, SLC1A3. Genetic or pharmacologic modulation of SLC1A3 activity markedly altered cancer cell sensitivity to ETC inhibitors. Interestingly, aspartate levels also decrease under low oxygen, and increasing aspartate import by SLC1A3 provides a competitive advantage to cancer cells at low oxygen levels and in tumour xenografts. Finally, aspartate levels in primary human tumours negatively correlate with the expression of hypoxia markers, suggesting that tumour hypoxia is sufficient to inhibit ETC and, consequently, aspartate synthesis in vivo. Therefore, aspartate may be a limiting metabolite for tumour growth, and aspartate availability could be targeted for cancer therapy.


Asunto(s)
Ácido Aspártico/metabolismo , Proliferación Celular , Metabolismo Energético , Neoplasias/metabolismo , Hipoxia Tumoral , Microambiente Tumoral , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Transporte Biológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético/efectos de los fármacos , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Humanos , Metabolómica/métodos , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Mitocondrias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Transducción de Señal , Factores de Tiempo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
17.
Nat Cell Biol ; 20(10): 1228, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30089842

RESUMEN

In the version of this Letter originally published, the competing interests statement was missing. The authors declare no competing interests; this statement has now been added in all online versions of the Letter.

18.
Cell Rep ; 23(1): 172-180.e3, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29617658

RESUMEN

Precision oncology uses genomic evidence to match patients with treatment but often fails to identify all patients who may respond. The transcriptome of these "hidden responders" may reveal responsive molecular states. We describe and evaluate a machine-learning approach to classify aberrant pathway activity in tumors, which may aid in hidden responder identification. The algorithm integrates RNA-seq, copy number, and mutations from 33 different cancer types across The Cancer Genome Atlas (TCGA) PanCanAtlas project to predict aberrant molecular states in tumors. Applied to the Ras pathway, the method detects Ras activation across cancer types and identifies phenocopying variants. The model, trained on human tumors, can predict response to MEK inhibitors in wild-type Ras cell lines. We also present data that suggest that multiple hits in the Ras pathway confer increased Ras activity. The transcriptome is underused in precision oncology and, combined with machine learning, can aid in the identification of hidden responders.


Asunto(s)
Aprendizaje Automático , Neoplasias/genética , Proteínas ras/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Humanos , Neoplasias/metabolismo , Transducción de Señal , Proteínas ras/metabolismo
19.
J Clin Oncol ; 36(7): 633-641, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337640

RESUMEN

Purpose Treatment of advanced non-small-cell lung cancer with immune checkpoint inhibitors (ICIs) is characterized by durable responses and improved survival in a subset of patients. Clinically available tools to optimize use of ICIs and understand the molecular determinants of response are needed. Targeted next-generation sequencing (NGS) is increasingly routine, but its role in identifying predictors of response to ICIs is not known. Methods Detailed clinical annotation and response data were collected for patients with advanced non-small-cell lung cancer treated with anti-programmed death-1 or anti-programmed death-ligand 1 [anti-programmed cell death (PD)-1] therapy and profiled by targeted NGS (MSK-IMPACT; n = 240). Efficacy was assessed by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1, and durable clinical benefit (DCB) was defined as partial response/stable disease that lasted > 6 months. Tumor mutation burden (TMB), fraction of copy number-altered genome, and gene alterations were compared among patients with DCB and no durable benefit (NDB). Whole-exome sequencing (WES) was performed for 49 patients to compare quantification of TMB by targeted NGS versus WES. Results Estimates of TMB by targeted NGS correlated well with WES (ρ = 0.86; P < .001). TMB was greater in patients with DCB than with NDB ( P = .006). DCB was more common, and progression-free survival was longer in patients at increasing thresholds above versus below the 50th percentile of TMB (38.6% v 25.1%; P < .001; hazard ratio, 1.38; P = .024). The fraction of copy number-altered genome was highest in those with NDB. Variants in EGFR and STK11 associated with a lack of benefit. TMB and PD-L1 expression were independent variables, and a composite of TMB plus PD-L1 further enriched for benefit to ICIs. Conclusion Targeted NGS accurately estimates TMB and elevated TMB further improved likelihood of benefit to ICIs. TMB did not correlate with PD-L1 expression; both variables had similar predictive capacity. The incorporation of both TMB and PD-L1 expression into multivariable predictive models should result in greater predictive power.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Humanos , Mutación , Carga Tumoral , Secuenciación del Exoma
20.
Elife ; 62017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28099114

RESUMEN

The fundamental metabolic decision of a cell, the balance between respiration and fermentation, rests in part on expression of the mitochondrial genome (mtDNA) and coordination with expression of the nuclear genome (nuDNA). Previously we described mtDNA copy number depletion across many solid tumor types (Reznik et al., 2016). Here, we use orthogonal RNA-sequencing data to quantify mtDNA expression (mtRNA), and report analogously lower expression of mtRNA in tumors (relative to normal tissue) across a majority of cancer types. Several cancers exhibit a trio of mutually consistent evidence suggesting a drop in respiratory activity: depletion of mtDNA copy number, decreases in mtRNA levels, and decreases in expression of nuDNA-encoded respiratory proteins. Intriguingly, a minority of cancer types exhibit a drop in mtDNA expression but an increase in nuDNA expression of respiratory proteins, with unknown implications for respiratory activity. Our results indicate suppression of respiratory gene expression across many cancer types.


Asunto(s)
Respiración de la Célula , ADN Mitocondrial/genética , Expresión Génica , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias/patología , Dosificación de Gen , Perfilación de la Expresión Génica , Humanos , Neoplasias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA