Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.214
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 183(5): 1202-1218.e25, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33142117

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) tumors have a nutrient-poor, desmoplastic, and highly innervated tumor microenvironment. Although neurons can release stimulatory factors to accelerate PDAC tumorigenesis, the metabolic contribution of peripheral axons has not been explored. We found that peripheral axons release serine (Ser) to support the growth of exogenous Ser (exSer)-dependent PDAC cells during Ser/Gly (glycine) deprivation. Ser deprivation resulted in ribosomal stalling on two of the six Ser codons, TCC and TCT, and allowed the selective translation and secretion of nerve growth factor (NGF) by PDAC cells to promote tumor innervation. Consistent with this, exSer-dependent PDAC tumors grew slower and displayed enhanced innervation in mice on a Ser/Gly-free diet. Blockade of compensatory neuronal innervation using LOXO-101, a Trk-NGF inhibitor, further decreased PDAC tumor growth. Our data indicate that axonal-cancer metabolic crosstalk is a critical adaptation to support PDAC growth in nutrient poor environments.


Asunto(s)
Neuronas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Biosíntesis de Proteínas , Serina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Anciano , Animales , Axones/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular , Codón/genética , Femenino , Glicina/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mitocondrias/metabolismo , Tejido Nervioso/patología , Consumo de Oxígeno , Neoplasias Pancreáticas/patología , Pirazoles , Pirimidinas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/genética , Ratas
2.
Cell ; 177(3): 572-586.e22, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955884

RESUMEN

Drug resistance and relapse remain key challenges in pancreatic cancer. Here, we have used RNA sequencing (RNA-seq), chromatin immunoprecipitation (ChIP)-seq, and genome-wide CRISPR analysis to map the molecular dependencies of pancreatic cancer stem cells, highly therapy-resistant cells that preferentially drive tumorigenesis and progression. This integrated genomic approach revealed an unexpected utilization of immuno-regulatory signals by pancreatic cancer epithelial cells. In particular, the nuclear hormone receptor retinoic-acid-receptor-related orphan receptor gamma (RORγ), known to drive inflammation and T cell differentiation, was upregulated during pancreatic cancer progression, and its genetic or pharmacologic inhibition led to a striking defect in pancreatic cancer growth and a marked improvement in survival. Further, a large-scale retrospective analysis in patients revealed that RORγ expression may predict pancreatic cancer aggressiveness, as it positively correlated with advanced disease and metastasis. Collectively, these data identify an orthogonal co-option of immuno-regulatory signals by pancreatic cancer stem cells, suggesting that autoimmune drugs should be evaluated as novel treatment strategies for pancreatic cancer patients.


Asunto(s)
Adenocarcinoma/patología , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Epigénesis Genética , Biblioteca de Genes , Humanos , Ratones , Ratones Noqueados , Ratones SCID , Células Madre Neoplásicas/citología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina-10/antagonistas & inhibidores , Receptores de Interleucina-10/genética , Receptores de Interleucina-10/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transcriptoma , Células Tumorales Cultivadas
3.
Cell ; 174(4): 856-869.e17, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30096312

RESUMEN

Recent sequencing analyses have shed light on heterogeneous patterns of genomic aberrations in human gastric cancers (GCs). To explore how individual genetic events translate into cancer phenotypes, we established a biological library consisting of genetically engineered gastric organoids carrying various GC mutations and 37 patient-derived organoid lines, including rare genomically stable GCs. Phenotype analyses of GC organoids revealed divergent genetic and epigenetic routes to gain Wnt and R-spondin niche independency. An unbiased phenotype-based genetic screening identified a significant association between CDH1/TP53 compound mutations and the R-spondin independency that was functionally validated by CRISPR-based knockout. Xenografting of GC organoids further established the feasibility of Wnt-targeting therapy for Wnt-dependent GCs. Our results collectively demonstrate that multifaceted genetic abnormalities render human GCs independent of the stem cell niche and highlight the validity of the genotype-phenotype screening strategy in gaining deeper understanding of human cancers.


Asunto(s)
Adenocarcinoma/patología , Organoides/patología , Neoplasias Gástricas/patología , Estómago/patología , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Antígenos CD/genética , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Cadherinas/genética , Carcinogénesis , Proliferación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Organoides/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Trombospondinas/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteínas Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Annu Rev Biochem ; 86: 333-356, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28654324

RESUMEN

Many biochemical systems are spatially heterogeneous and exhibit nonlinear behaviors, such as state switching in response to small changes in the local concentration of diffusible molecules. Systems as varied as blood clotting, intracellular calcium signaling, and tissue inflammation are all heavily influenced by the balance of rates of reaction and mass transport phenomena including flow and diffusion. Transport of signaling molecules is also affected by geometry and chemoselective confinement via matrix binding. In this review, we use a phenomenon referred to as patchy switching to illustrate the interplay of nonlinearities, transport phenomena, and spatial effects. Patchy switching describes a change in the state of a network when the local concentration of a diffusible molecule surpasses a critical threshold. Using patchy switching as an example, we describe conceptual tools from nonlinear dynamics and chemical engineering that make testable predictions and provide a unifying description of the myriad possible experimental observations. We describe experimental microfluidic and biochemical tools emerging to test conceptual predictions by controlling transport phenomena and spatial distribution of diffusible signals, and we highlight the unmet need for in vivo tools.


Asunto(s)
Adenocarcinoma/metabolismo , Redes Reguladoras de Genes , Neoplasias Pulmonares/metabolismo , Redes y Vías Metabólicas/genética , Esclerosis Múltiple/metabolismo , Dinámicas no Lineales , Osteoporosis/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Transporte Biológico , Difusión , Humanos , Dispositivos Laboratorio en un Chip , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Microfluídica/instrumentación , Microfluídica/métodos , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Osteoporosis/genética , Osteoporosis/patología , Transducción de Señal
5.
Cell ; 170(5): 875-888.e20, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28757253

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal human malignancies, owing in part to its propensity for metastasis. Here, we used an organoid culture system to investigate how transcription and the enhancer landscape become altered during discrete stages of disease progression in a PDA mouse model. This approach revealed that the metastatic transition is accompanied by massive and recurrent alterations in enhancer activity. We implicate the pioneer factor FOXA1 as a driver of enhancer activation in this system, a mechanism that renders PDA cells more invasive and less anchorage-dependent for growth in vitro, as well as more metastatic in vivo. In this context, FOXA1-dependent enhancer reprogramming activates a transcriptional program of embryonic foregut endoderm. Collectively, our study implicates enhancer reprogramming, FOXA1 upregulation, and a retrograde developmental transition in PDA metastasis.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epigenómica , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Organoides/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología
6.
Cell ; 171(6): 1301-1315.e14, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-29195074

RESUMEN

The two oncogenes KRas and Myc cooperate to drive tumorigenesis, but the mechanism underlying this remains unclear. In a mouse lung model of KRasG12D-driven adenomas, we find that co-activation of Myc drives the immediate transition to highly proliferative and invasive adenocarcinomas marked by highly inflammatory, angiogenic, and immune-suppressed stroma. We identify epithelial-derived signaling molecules CCL9 and IL-23 as the principal instructing signals for stromal reprogramming. CCL9 mediates recruitment of macrophages, angiogenesis, and PD-L1-dependent expulsion of T and B cells. IL-23 orchestrates exclusion of adaptive T and B cells and innate immune NK cells. Co-blockade of both CCL9 and IL-23 abrogates Myc-induced tumor progression. Subsequent deactivation of Myc in established adenocarcinomas triggers immediate reversal of all stromal changes and tumor regression, which are independent of CD4+CD8+ T cells but substantially dependent on returning NK cells. We show that Myc extensively programs an immune suppressive stroma that is obligatory for tumor progression.


Asunto(s)
Adenocarcinoma/inmunología , Adenoma/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenoma/genética , Adenoma/patología , Animales , Carcinogénesis , Quimiocinas CC/inmunología , Modelos Animales de Enfermedad , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-23/inmunología , Neoplasias Pulmonares/patología , Proteínas Inflamatorias de Macrófagos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Microambiente Tumoral
7.
Cell ; 167(7): 1803-1813.e12, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984728

RESUMEN

There is growing evidence that stress-coping mechanisms represent tumor cell vulnerabilities that may function as therapeutically beneficial targets. Recent work has delineated an integrated stress adaptation mechanism that is characterized by the formation of cytoplasmic mRNA and protein foci, termed stress granules (SGs). Here, we demonstrate that SGs are markedly elevated in mutant KRAS cells following exposure to stress-inducing stimuli. The upregulation of SGs by mutant KRAS is dependent on the production of the signaling lipid molecule 15-deoxy-delta 12,14 prostaglandin J2 (15-d-PGJ2) and confers cytoprotection against stress stimuli and chemotherapeutic agents. The secretion of 15-d-PGJ2 by mutant KRAS cells is sufficient to enhance SG formation and stress resistance in cancer cells that are wild-type for KRAS. Our findings identify a mutant KRAS-dependent cell non-autonomous mechanism that may afford the establishment of a stress-resistant niche that encompasses different tumor subclones. These results should inform the design of strategies to eradicate tumor cell communities.


Asunto(s)
Adenocarcinoma/patología , Neoplasias del Colon/metabolismo , Gránulos Citoplasmáticos/metabolismo , Neoplasias Pancreáticas/patología , Prostaglandina D2/análogos & derivados , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Resistencia a Antineoplásicos , Factor 4A Eucariótico de Iniciación/metabolismo , Femenino , Xenoinjertos , Humanos , Ratones , Mutación , Trasplante de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Prostaglandina D2/biosíntesis , Prostaglandina D2/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Regulación hacia Arriba
8.
Nat Immunol ; 19(12): 1319-1329, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30397348

RESUMEN

Many tumors evolve sophisticated strategies to evade the immune system, and these represent major obstacles for efficient antitumor immune responses. Here we explored a molecular mechanism of metabolic communication deployed by highly glycolytic tumors for immunoevasion. In contrast to colon adenocarcinomas, melanomas showed comparatively high glycolytic activity, which resulted in high acidification of the tumor microenvironment. This tumor acidosis induced Gprotein-coupled receptor-dependent expression of the transcriptional repressor ICER in tumor-associated macrophages that led to their functional polarization toward a non-inflammatory phenotype and promoted tumor growth. Collectively, our findings identify a molecular mechanism of metabolic communication between non-lymphoid tissue and the immune system that was exploited by high-glycolytic-rate tumors for evasion of the immune system.


Asunto(s)
Adenocarcinoma/inmunología , Macrófagos/inmunología , Melanoma/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Acidosis/inmunología , Adenocarcinoma/metabolismo , Animales , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Glucólisis/inmunología , Humanos , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Cell ; 162(1): 146-59, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26140595

RESUMEN

KRAS is one of the most frequently mutated oncogenes in human cancer. Despite substantial efforts, no clinically applicable strategy has yet been developed to effectively treat KRAS-mutant tumors. Here, we perform a cell-line-based screen and identify strong synergistic interactions between cell-cycle checkpoint-abrogating Chk1- and MK2 inhibitors, specifically in KRAS- and BRAF-driven cells. Mechanistically, we show that KRAS-mutant cancer displays intrinsic genotoxic stress, leading to tonic Chk1- and MK2 activity. We demonstrate that simultaneous Chk1- and MK2 inhibition leads to mitotic catastrophe in KRAS-mutant cells. This actionable synergistic interaction is validated using xenograft models, as well as distinct Kras- or Braf-driven autochthonous murine cancer models. Lastly, we show that combined checkpoint inhibition induces apoptotic cell death in KRAS- or BRAF-mutant tumor cells directly isolated from patients. These results strongly recommend simultaneous Chk1- and MK2 inhibition as a therapeutic strategy for the treatment of KRAS- or BRAF-driven cancers.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Animales , Puntos de Control del Ciclo Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Células Tumorales Cultivadas
10.
Cell ; 158(1): 11-2, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995973

RESUMEN

It is of great therapeutic importance to understand why tumors relapse after the failure of therapies targeting oncogenes to which cancer cells are addicted. In this issue, Kapoor et al. and Shao et al. identify the transcriptional coactivator YAP1 as a central driver of compensation for the loss of K-Ras signaling in K-Ras-dependent cancers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Supervivencia Celular , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/metabolismo , Animales , Proteínas de Ciclo Celular , Humanos , Factores de Transcripción , Proteínas Señalizadoras YAP
11.
Cell ; 158(1): 185-197, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24954535

RESUMEN

Activating mutations in KRAS are among the most frequent events in diverse human carcinomas and are particularly prominent in human pancreatic ductal adenocarcinoma (PDAC). An inducible Kras(G12D)-driven mouse model of PDAC has established a critical role for sustained Kras(G12D) expression in tumor maintenance, providing a model to determine the potential for and the underlying mechanisms of Kras(G12D)-independent PDAC recurrence. Here, we show that some tumors undergo spontaneous relapse and are devoid of Kras(G12D) expression and downstream canonical MAPK signaling and instead acquire amplification and overexpression of the transcriptional coactivator Yap1. Functional studies established the role of Yap1 and the transcriptional factor Tead2 in driving Kras(G12D)-independent tumor maintenance. The Yap1/Tead2 complex acts cooperatively with E2F transcription factors to activate a cell cycle and DNA replication program. Our studies, along with corroborating evidence from human PDAC models, portend a novel mechanism of escape from oncogenic Kras addiction in PDAC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/patología , Ciclo Celular , Proteínas de Ciclo Celular , Línea Celular Tumoral , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción E2F/metabolismo , Humanos , Ratones , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción de Dominio TEA , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP , Proteínas ras/metabolismo
12.
Cell ; 152(3): 599-611, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374352

RESUMEN

Tumor cells have high-energetic and anabolic needs and are known to adapt their metabolism to be able to survive and keep proliferating under conditions of nutrient stress. We show that PKCζ deficiency promotes the plasticity necessary for cancer cells to reprogram their metabolism to utilize glutamine through the serine biosynthetic pathway in the absence of glucose. PKCζ represses the expression of two key enzymes of the pathway, PHGDH and PSAT1, and phosphorylates PHGDH at key residues to inhibit its enzymatic activity. Interestingly, the loss of PKCζ in mice results in enhanced intestinal tumorigenesis and increased levels of these two metabolic enzymes, whereas patients with low levels of PKCζ have a poor prognosis. Furthermore, PKCζ and caspase-3 activities are correlated with PHGDH levels in human intestinal tumors. Taken together, this demonstrates that PKCζ is a critical metabolic tumor suppressor in mouse and human cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Adenoma/metabolismo , Neoplasias del Colon/metabolismo , Proteína Quinasa C/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Vías Biosintéticas , Transformación Celular Neoplásica , Glucosa/metabolismo , Humanos , Ratones , Serina/biosíntesis , Organismos Libres de Patógenos Específicos , Estrés Fisiológico
13.
Cell ; 153(1): 86-100, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23540692

RESUMEN

Glutamate and its receptor N-methyl-D-aspartate receptor (NMDAR) have been associated with cancer, although their functions are not fully understood. Herein, we implicate glutamate-driven NMDAR signaling in a mouse model of pancreatic neuroendocrine tumorigenesis (PNET) and in selected human cancers. NMDAR was upregulated at the periphery of PNET tumors, particularly invasive fronts. Moreover, elevated coexpression of NMDAR and glutamate exporters correlated with poor prognosis in cancer patients. Treatment of a tumor-derived cell line with NMDAR antagonists impaired cancer cell proliferation and invasion. Flow conditions mimicking interstitial fluid pressure induced autologous glutamate secretion, activating NMDAR and its downstream MEK-MAPK and CaMK effectors, thereby promoting invasiveness. Congruently, pharmacological inhibition of NMDAR in mice with PNET reduced tumor growth and invasiveness. Therefore, beyond its traditional role in neurons, NMDAR may be activated in human tumors by fluid flow consequent to higher interstitial pressure, inducing an autocrine glutamate signaling circuit with resultant stimulation of malignancy.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Microambiente Tumoral
14.
Cell ; 149(3): 656-70, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22541435

RESUMEN

Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. Here, an inducible Kras(G12D)-driven PDAC mouse model establishes that advanced PDAC remains strictly dependent on Kras(G12D) expression. Transcriptome and metabolomic analyses indicate that Kras(G12D) serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that Kras(G12D) drives glycolysis intermediates into the nonoxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC.


Asunto(s)
Adenocarcinoma/metabolismo , Modelos Animales de Enfermedad , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Transcripción Genética
15.
Nature ; 580(7801): 136-141, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32238925

RESUMEN

Cancer genomics studies have identified thousands of putative cancer driver genes1. Development of high-throughput and accurate models to define the functions of these genes is a major challenge. Here we devised a scalable cancer-spheroid model and performed genome-wide CRISPR screens in 2D monolayers and 3D lung-cancer spheroids. CRISPR phenotypes in 3D more accurately recapitulated those of in vivo tumours, and genes with differential sensitivities between 2D and 3D conditions were highly enriched for genes that are mutated in lung cancers. These analyses also revealed drivers that are essential for cancer growth in 3D and in vivo, but not in 2D. Notably, we found that carboxypeptidase D is responsible for removal of a C-terminal RKRR motif2 from the α-chain of the insulin-like growth factor 1 receptor that is critical for receptor activity. Carboxypeptidase D expression correlates with patient outcomes in patients with lung cancer, and loss of carboxypeptidase D reduced tumour growth. Our results reveal key differences between 2D and 3D cancer models, and establish a generalizable strategy for performing CRISPR screens in spheroids to reveal cancer vulnerabilities.


Asunto(s)
Sistemas CRISPR-Cas/genética , Técnicas de Cultivo de Célula/métodos , Proliferación Celular/genética , Genoma Humano/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Esferoides Celulares/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Secuencias de Aminoácidos , Animales , Carboxipeptidasas/antagonistas & inhibidores , Carboxipeptidasas/deficiencia , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Terapia Molecular Dirigida , Mutación , Fenotipo , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Esferoides Celulares/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Cell ; 69(4): 594-609.e8, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29452639

RESUMEN

Accumulating evidence indicates that the MDM2 oncoprotein promotes tumorigenesis beyond its canonical negative effects on the p53 tumor suppressor, but these p53-independent functions remain poorly understood. Here, we show that a fraction of endogenous MDM2 is actively imported in mitochondria to control respiration and mitochondrial dynamics independently of p53. Mitochondrial MDM2 represses the transcription of NADH-dehydrogenase 6 (MT-ND6) in vitro and in vivo, impinging on respiratory complex I activity and enhancing mitochondrial ROS production. Recruitment of MDM2 to mitochondria increases during oxidative stress and hypoxia. Accordingly, mice lacking MDM2 in skeletal muscles exhibit higher MT-ND6 levels, enhanced complex I activity, and increased muscular endurance in mild hypoxic conditions. Furthermore, increased mitochondrial MDM2 levels enhance the migratory and invasive properties of cancer cells. Collectively, these data uncover a previously unsuspected function of the MDM2 oncoprotein in mitochondria that play critical roles in skeletal muscle physiology and may contribute to tumor progression.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Transformación Celular Neoplásica/patología , Complejo I de Transporte de Electrón/metabolismo , Regulación Neoplásica de la Expresión Génica , Mitocondrias/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Complejo I de Transporte de Electrón/genética , Genoma Mitocondrial , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Invasividad Neoplásica , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-mdm2/genética , Transducción de Señal , Transcripción Genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Mol Cell Proteomics ; 23(6): 100764, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38604503

RESUMEN

Efforts to address the poor prognosis associated with esophageal adenocarcinoma (EAC) have been hampered by a lack of biomarkers to identify early disease and therapeutic targets. Despite extensive efforts to understand the somatic mutations associated with EAC over the past decade, a gap remains in understanding how the atlas of genomic aberrations in this cancer impacts the proteome and which somatic variants are of importance for the disease phenotype. We performed a quantitative proteomic analysis of 23 EACs and matched adjacent normal esophageal and gastric tissues. We explored the correlation of transcript and protein abundance using tissue-matched RNA-seq and proteomic data from seven patients and further integrated these data with a cohort of EAC RNA-seq data (n = 264 patients), EAC whole-genome sequencing (n = 454 patients), and external published datasets. We quantified protein expression from 5879 genes in EAC and patient-matched normal tissues. Several biomarker candidates with EAC-selective expression were identified, including the transmembrane protein GPA33. We further verified the EAC-enriched expression of GPA33 in an external cohort of 115 patients and confirm this as an attractive diagnostic and therapeutic target. To further extend the insights gained from our proteomic data, an integrated analysis of protein and RNA expression in EAC and normal tissues revealed several genes with poorly correlated protein and RNA abundance, suggesting posttranscriptional regulation of protein expression. These outlier genes, including SLC25A30, TAOK2, and AGMAT, only rarely demonstrated somatic mutation, suggesting post-transcriptional drivers for this EAC-specific phenotype. AGMAT was demonstrated to be overexpressed at the protein level in EAC compared to adjacent normal tissues with an EAC-selective, post-transcriptional mechanism of regulation of protein abundance proposed. Integrated analysis of proteome, transcriptome, and genome in EAC has revealed several genes with tumor-selective, posttranscriptional regulation of protein expression, which may be an exploitable vulnerability.


Asunto(s)
Adenocarcinoma , Biomarcadores de Tumor , Neoplasias Esofágicas , Regulación Neoplásica de la Expresión Génica , Proteómica , Humanos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Proteómica/métodos , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Masculino , Femenino , Procesamiento Postranscripcional del ARN , Proteoma/metabolismo , Multiómica
18.
Mol Cell Proteomics ; 23(5): 100766, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38608841

RESUMEN

The diagnosis of primary lung adenocarcinomas with intestinal or mucinous differentiation (PAIM) remains challenging due to the overlapping histomorphological, immunohistochemical (IHC), and genetic characteristics with lung metastatic colorectal cancer (lmCRC). This study aimed to explore the protein biomarkers that could distinguish between PAIM and lmCRC. To uncover differences between the two diseases, we used tandem mass tagging-based shotgun proteomics to characterize proteomes of formalin-fixed, paraffin-embedded tumor samples of PAIM (n = 22) and lmCRC (n = 17).Then three machine learning algorithms, namely support vector machine (SVM), random forest, and the Least Absolute Shrinkage and Selection Operator, were utilized to select protein features with diagnostic significance. These candidate proteins were further validated in an independent cohort (PAIM, n = 11; lmCRC, n = 19) by IHC to confirm their diagnostic performance. In total, 105 proteins out of 7871 proteins were significantly dysregulated between PAIM and lmCRC samples and well-separated two groups by Uniform Manifold Approximation and Projection. The upregulated proteins in PAIM were involved in actin cytoskeleton organization, platelet degranulation, and regulation of leukocyte chemotaxis, while downregulated ones were involved in mitochondrial transmembrane transport, vasculature development, and stem cell proliferation. A set of ten candidate proteins (high-level expression in lmCRC: CDH17, ATP1B3, GLB1, OXNAD1, LYST, FABP1; high-level expression in PAIM: CK7 (an established marker), NARR, MLPH, S100A14) was ultimately selected to distinguish PAIM from lmCRC by machine learning algorithms. We further confirmed using IHC that the five protein biomarkers including CDH17, CK7, MLPH, FABP1 and NARR were effective biomarkers for distinguishing PAIM from lmCRC. Our study depicts PAIM-specific proteomic characteristics and demonstrates the potential utility of new protein biomarkers for the differential diagnosis of PAIM and lmCRC. These findings may contribute to improving the diagnostic accuracy and guide appropriate treatments for these patients.


Asunto(s)
Adenocarcinoma del Pulmón , Biomarcadores de Tumor , Neoplasias Colorrectales , Neoplasias Pulmonares , Proteómica , Humanos , Biomarcadores de Tumor/metabolismo , Proteómica/métodos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Masculino , Femenino , Diagnóstico Diferencial , Diferenciación Celular , Persona de Mediana Edad , Anciano , Adenocarcinoma/metabolismo , Adenocarcinoma/patología
19.
Gastroenterology ; 166(5): 772-786.e14, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38272100

RESUMEN

BACKGROUND & AIMS: Gastric carcinogenesis develops within a sequential carcinogenic cascade from precancerous metaplasia to dysplasia and adenocarcinoma, and oncogenic gene activation can drive the process. Metabolic reprogramming is considered a key mechanism for cancer cell growth and proliferation. However, how metabolic changes contribute to the progression of metaplasia to dysplasia remains unclear. We have examined metabolic dynamics during gastric carcinogenesis using a novel mouse model that induces Kras activation in zymogen-secreting chief cells. METHODS: We generated a Gif-rtTA;TetO-Cre;KrasG12D (GCK) mouse model that continuously induces active Kras expression in chief cells after doxycycline treatment. Histologic examination and imaging mass spectrometry were performed in the GCK mouse stomachs at 2 to 14 weeks after doxycycline treatment. Mouse and human gastric organoids were used for metabolic enzyme inhibitor treatment. The GCK mice were treated with a stearoyl- coenzyme A desaturase (SCD) inhibitor to inhibit the fatty acid desaturation. Tissue microarrays were used to assess the SCD expression in human gastrointestinal cancers. RESULTS: The GCK mice developed metaplasia and high-grade dysplasia within 4 months. Metabolic reprogramming from glycolysis to fatty acid metabolism occurred during metaplasia progression to dysplasia. Altered fatty acid desaturation through SCD produces a novel eicosenoic acid, which fuels dysplastic cell hyperproliferation and survival. The SCD inhibitor killed both mouse and human dysplastic organoids and selectively targeted dysplastic cells in vivo. SCD was up-regulated during carcinogenesis in human gastrointestinal cancers. CONCLUSIONS: Active Kras expression only in gastric chief cells drives the full spectrum of gastric carcinogenesis. Also, oncogenic metabolic rewiring is an essential adaptation for high-energy demand in dysplastic cells.


Asunto(s)
Metabolismo Energético , Ácidos Grasos , Metaplasia , Organoides , Proteínas Proto-Oncogénicas p21(ras) , Neoplasias Gástricas , Animales , Neoplasias Gástricas/patología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/genética , Humanos , Ácidos Grasos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Organoides/metabolismo , Ratones , Modelos Animales de Enfermedad , Carcinogénesis/metabolismo , Carcinogénesis/genética , Carcinogénesis/patología , Células Principales Gástricas/metabolismo , Células Principales Gástricas/patología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/genética , Ratones Transgénicos , Glucólisis , Adenocarcinoma/patología , Adenocarcinoma/metabolismo , Adenocarcinoma/genética , Progresión de la Enfermedad , Lesiones Precancerosas/patología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/genética
20.
Am J Pathol ; 194(6): 1137-1153, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38749609

RESUMEN

Preclinical models that display spontaneous metastasis are necessary to improve the therapeutic options for hormone receptor-positive breast cancers. Within this study, detailed cellular and molecular characterization was conducted on MCa-P1362, a newly established mouse model of metastatic breast cancer that is syngeneic in BALB/c mice. MCa-P1362 cancer cells express estrogen receptor, progesterone receptor, and the human epidermal growth factor receptor 2. MCa-P1362 cancer cells proliferate in vitro and in vivo in response to estrogen, yet do not depend on steroid hormones for growth and tumor progression. Analysis of MCa-P1362 tumor explants revealed the tumors contained a mixture of cancer cells and mesenchymal stromal cells. Through transcriptomic and functional analyses of both cancer and stromal cells, stem cells were detected within both populations. Functional studies demonstrated that MCa-P1362 cancer stem cells drove tumor initiation, whereas stromal cells from these tumors contributed to drug resistance. MCa-P1362 may serve as a useful preclinical model to investigate the cellular and molecular basis of breast tumor progression and therapeutic resistance.


Asunto(s)
Adenocarcinoma , Células Madre Mesenquimatosas , Ratones Endogámicos BALB C , Receptor ErbB-2 , Receptores de Estrógenos , Animales , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Femenino , Humanos , Receptor ErbB-2/metabolismo , Ratones , Receptores de Estrógenos/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Línea Celular Tumoral , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA