Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552609

RESUMEN

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Asunto(s)
Neoplasias , Humanos , Carcinogénesis , Microbiota , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Obesidad/complicaciones , Calidad de Vida
2.
Artículo en Inglés | MEDLINE | ID: mdl-37696657

RESUMEN

Catabolic pathways change in anabolic diseases such as cancer to maintain metabolic homeostasis. The liver urea cycle (UC) is the main catabolic pathway for disposing excess nitrogen. Outside the liver, the UC enzymes are differentially expressed based on each tissue's needs for UC intermediates. In tumors, there are changes in the expression of UC enzymes selected for promoting tumorigenesis by increasing the availability of essential UC substrates and products. Consequently, there are compensatory changes in the expression of UC enzymes in the cells that compose the tumor microenvironment. Moreover, extrahepatic tumors induce changes in the expression of the liver UC, which contribute to the systemic manifestations of cancer, such as weight loss. Here, we review the multilayer changes in the expression of UC enzymes throughout carcinogenesis. Understanding the changes in UC expression in the tumor and its micro and macro environment can help identify biomarkers for early cancer diagnosis and vulnerabilities that can be targeted for therapy.


Asunto(s)
Neoplasias , Humanos , Neoplasias/metabolismo , Hígado/metabolismo , Urea/metabolismo , Microambiente Tumoral
3.
Nature ; 624(7992): 645-652, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38093014

RESUMEN

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Asunto(s)
Células Dendríticas , Complicaciones de la Diabetes , Diabetes Mellitus , Susceptibilidad a Enfermedades , Hiperglucemia , Pulmón , Virosis , Animales , Ratones , Acetilcoenzima A/metabolismo , Acetilación , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Complicaciones de la Diabetes/inmunología , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Histonas/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/inmunología , Hiperglucemia/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Linfocitos T/inmunología , Virosis/complicaciones , Virosis/inmunología , Virosis/mortalidad , Virus/inmunología , Modelos Animales de Enfermedad , Humanos
4.
Cancer Discov ; 13(7): 1616-1635, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-36972357

RESUMEN

Multiple studies have identified metabolic changes within the tumor and its microenvironment during carcinogenesis. Yet, the mechanisms by which tumors affect the host metabolism are unclear. We find that systemic inflammation induced by cancer leads to liver infiltration of myeloid cells at early extrahepatic carcinogenesis. The infiltrating immune cells via IL6-pSTAT3 immune-hepatocyte cross-talk cause the depletion of a master metabolic regulator, HNF4α, consequently leading to systemic metabolic changes that promote breast and pancreatic cancer proliferation and a worse outcome. Preserving HNF4α levels maintains liver metabolism and restricts carcinogenesis. Standard liver biochemical tests can identify early metabolic changes and predict patients' outcomes and weight loss. Thus, the tumor induces early metabolic changes in its macroenvironment with diagnostic and potentially therapeutic implications for the host. SIGNIFICANCE: Cancer growth requires a permanent nutrient supply starting from early disease stages. We find that the tumor extends its effect to the host's liver to obtain nutrients and rewires the systemic and tissue-specific metabolism early during carcinogenesis. Preserving liver metabolism restricts tumor growth and improves cancer outcomes. This article is highlighted in the In This Issue feature, p. 1501.


Asunto(s)
Hígado , Neoplasias Pancreáticas , Humanos , Hígado/metabolismo , Carcinogénesis/patología , Hepatocitos , Neoplasias Pancreáticas/patología , Inmunidad Innata , Microambiente Tumoral
5.
Cancers (Basel) ; 14(23)2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36497367

RESUMEN

Cancer occurs more frequently in men while autoimmune diseases (AIDs) occur more frequently in women. To explore whether these sex biases have a common basis, we collected 167 AID incidence studies from many countries for tissues that have both a cancer type and an AID that arise from that tissue. Analyzing a total of 182 country-specific, tissue-matched cancer-AID incidence rate sex bias data pairs, we find that, indeed, the sex biases observed in the incidence of AIDs and cancers that occur in the same tissue are positively correlated across human tissues. The common key factor whose levels across human tissues are most strongly associated with these incidence rate sex biases is the sex bias in the expression of the 37 genes encoded in the mitochondrial genome.

6.
Nat Commun ; 13(1): 7830, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36539415

RESUMEN

Metabolic reprogramming is critical for tumor initiation and progression. However, the exact impact of specific metabolic changes on cancer progression is poorly understood. Here, we integrate multimodal analyses of primary and metastatic clonally-related clear cell renal cancer cells (ccRCC) grown in physiological media to identify key stage-specific metabolic vulnerabilities. We show that a VHL loss-dependent reprogramming of branched-chain amino acid catabolism sustains the de novo biosynthesis of aspartate and arginine enabling tumor cells with the flexibility of partitioning the nitrogen of the amino acids depending on their needs. Importantly, we identify the epigenetic reactivation of argininosuccinate synthase (ASS1), a urea cycle enzyme suppressed in primary ccRCC, as a crucial event for metastatic renal cancer cells to acquire the capability to generate arginine, invade in vitro and metastasize in vivo. Overall, our study uncovers a mechanism of metabolic flexibility occurring during ccRCC progression, paving the way for the development of novel stage-specific therapies.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/genética , Aminoácidos de Cadena Ramificada , Nitrógeno , Neoplasias Renales/genética , Arginina/metabolismo , Línea Celular Tumoral
7.
Clin Genet ; 101(4): 442-447, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34967012

RESUMEN

Polymerase proofreading-associated polyposis (PPAP) and Lynch syndrome, caused by mutated POLE and mismatch repair (MMR) genes, respectively, are associated with adult-onset cancer. PPAP and MMR-deficient tumors are both hypermutated, and each has a unique mutational signature. We describe a 4.5-year-old boy with multiple café au lait spots who presented with metastatic Sonic Hedgehog-activated medulloblastoma, with partial response to intensive chemotherapy and immunotherapy. The tumor showed microsatellite stability, loss of PMS2 nuclear expression, and an exceptionally high tumor mutational burden of 276 Mut/Mb. Germline molecular analysis revealed an inherited heterozygous pathogenic POLE variant and a de novo heterozygous PMS2 pathogenic variant. The tumor featured the MMR, POLE, and POLE+MMR mutational signatures. This is the first description of a di-genic condition, which we named "POL-LYNCH syndrome," manifested by an aggressive ultra-mutant pediatric medulloblastoma with a unique genomic signature.


Asunto(s)
Neoplasias Cerebelosas , Neoplasias Colorrectales Hereditarias sin Poliposis , ADN Polimerasa II/genética , Meduloblastoma , Proteínas de Unión a Poli-ADP-Ribosa/genética , Neoplasias Cerebelosas/complicaciones , Neoplasias Cerebelosas/genética , Preescolar , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Reparación de la Incompatibilidad de ADN/genética , Proteínas de Unión al ADN/genética , Mutación de Línea Germinal/genética , Proteínas Hedgehog/genética , Humanos , Masculino , Meduloblastoma/genética , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/genética
8.
Mol Cell ; 81(18): 3749-3759, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34469752

RESUMEN

The expression of the urea cycle (UC) proteins is dysregulated in multiple cancers, providing metabolic benefits to tumor survival, proliferation, and growth. Here, we review the main changes described in the expression of UC enzymes and metabolites in different cancers at various stages and suggest that these changes are dynamic and should hence be viewed in a context-specific manner. Understanding the evolvability in the activity of the UC pathway in cancer has implications for cancer-immune cell interactions and for cancer diagnosis and therapy.


Asunto(s)
Carcinogénesis/metabolismo , Transformación Celular Neoplásica/metabolismo , Urea/metabolismo , Amoníaco/metabolismo , Línea Celular Tumoral , Proliferación Celular , Expresión Génica/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/fisiopatología
9.
Ann N Y Acad Sci ; 1506(1): 55-73, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34414571

RESUMEN

There is an increasing appreciation for the role of metabolism in cell signaling and cell decision making. Precise metabolic control is essential in development, as evident by the disorders caused by mutations in metabolic enzymes. The metabolic profile of cells is often cell-type specific, changing as cells differentiate or during tumorigenesis. Recent evidence has shown that changes in metabolism are not merely a consequence of changes in cell state but that metabolites can serve to promote and/or inhibit these changes. Metabolites can link metabolic pathways with cell signaling pathways via several mechanisms, for example, by serving as substrates for protein post-translational modifications, by affecting enzyme activity via allosteric mechanisms, or by altering epigenetic markers. Unraveling the complex interactions governing metabolism, gene expression, and protein activity that ultimately govern a cell's fate will require new tools and interactions across disciplines. On March 24 and 25, 2021, experts in cell metabolism, developmental biology, and human disease met virtually for the Keystone eSymposium, "Metabolic Decisions in Development and Disease." The discussions explored how metabolites impact cellular and developmental decisions in a diverse range of model systems used to investigate normal development, developmental disorders, dietary effects, and cancer-mediated changes in metabolism.


Asunto(s)
Congresos como Asunto/tendencias , Desarrollo Humano/fisiología , Enfermedades Metabólicas/fisiopatología , Redes y Vías Metabólicas/fisiología , Neoplasias/fisiopatología , Informe de Investigación , Animales , Epigénesis Genética/fisiología , Humanos , Enfermedades Metabólicas/genética , Neoplasias/genética , Transducción de Señal/fisiología
10.
Hum Genet ; 140(10): 1471-1485, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34417872

RESUMEN

Argininosuccinate lyase (ASL) is essential for the NO-dependent regulation of tyrosine hydroxylase (TH) and thus for catecholamine production. Using a conditional mouse model with loss of ASL in catecholamine neurons, we demonstrate that ASL is expressed in dopaminergic neurons in the substantia nigra pars compacta, including the ALDH1A1 + subpopulation that is pivotal for the pathogenesis of Parkinson disease (PD). Neuronal loss of ASL results in catecholamine deficiency, in accumulation and formation of tyrosine aggregates, in elevation of α-synuclein, and phenotypically in motor and cognitive deficits. NO supplementation rescues the formation of aggregates as well as the motor deficiencies. Our data point to a potential metabolic link between accumulations of tyrosine and seeding of pathological aggregates in neurons as initiators for the pathological processes involved in neurodegeneration. Hence, interventions in tyrosine metabolism via regulation of NO levels may be therapeutic beneficial for the treatment of catecholamine-related neurodegenerative disorders.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/genética , Familia de Aldehído Deshidrogenasa 1/metabolismo , Argininosuccinatoliasa/genética , Argininosuccinatoliasa/metabolismo , Neuronas Dopaminérgicas/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Fenotipo , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo
11.
Sci Adv ; 7(21)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34138729

RESUMEN

Virus-infected cells and cancers share metabolic commonalities that stem from their insatiable need to replicate while evading the host immune system. These similarities include hijacking signaling mechanisms that induce metabolic rewiring in the host to up-regulate nucleotide metabolism and, in parallel, suppress the immune response. In both cancer and viral infections, the host immune cells and, specifically, lymphocytes augment nucleotide synthesis to support their own proliferation and effector functions. Consequently, established treatment modalities targeting nucleotide metabolism against cancers and virally infected cells may result in restricted immune response. Encouragingly, following the introduction of immunotherapy against cancers, multiple studies improved our understanding for improving antigen presentation to the immune system. We propose here that understanding the immune consequences of targeting nucleotide metabolism against cancers may be harnessed to optimize therapy against viral infections.


Asunto(s)
Neoplasias , Virosis , Presentación de Antígeno , Humanos , Inmunoterapia , Neoplasias/terapia , Nucleótidos
12.
Dev Cell ; 56(8): 1182-1194.e6, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33773101

RESUMEN

Pyruvate dehydrogenase kinases (PDK1-4) inhibit the TCA cycle by phosphorylating pyruvate dehydrogenase complex (PDC). Here, we show that PDK family is dispensable for murine embryonic development and that BCKDK serves as a compensatory mechanism by inactivating PDC. First, we knocked out all four Pdk genes one by one. Surprisingly, Pdk total KO embryos developed and were born in expected ratios but died by postnatal day 4 because of hypoglycemia or ketoacidosis. Moreover, PDC was phosphorylated in these embryos, suggesting that another kinase compensates for PDK family. Bioinformatic analysis implicated branched-chain ketoacid dehydrogenase kinase (Bckdk), a key regulator of branched-chain amino acids (BCAAs) catabolism. Indeed, knockout of Bckdk and Pdk family led to the loss of PDC phosphorylation, an increase in PDC activity and pyruvate entry into the TCA cycle, and embryonic lethality. These findings reveal a regulatory crosstalk hardwiring BCAA and glucose catabolic pathways, which feed the TCA cycle.


Asunto(s)
Ciclo del Ácido Cítrico , Desarrollo Embrionario , Proteínas Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Animales Recién Nacidos , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Eliminación de Gen , Hipoglucemia/complicaciones , Hipoglucemia/enzimología , Hipoglucemia/patología , Cetosis/complicaciones , Cetosis/enzimología , Cetosis/patología , Ratones Noqueados , Modelos Biológicos , Fosforilación , Ácido Pirúvico/metabolismo
14.
Cancer Discov ; 10(12): 1797-1807, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33139243

RESUMEN

Cancer cells continuously rewire their metabolism to fulfill their need for rapid growth and survival while subject to changes in environmental cues. Thus, a vital component of a cancer cell lies in its metabolic adaptability. The constant demand for metabolic alterations requires flexibility, that is, the ability to utilize different metabolic substrates; as well as plasticity, that is, the ability to process metabolic substrates in different ways. In this review, we discuss how dynamic changes in cancer metabolism affect tumor progression and the consequential implications for cancer therapy. SIGNIFICANCE: Recognizing cancer dynamic metabolic adaptability as an entity can lead to targeted therapy that is expected to decrease drug resistance.


Asunto(s)
Neoplasias/terapia , Microambiente Tumoral/genética , Humanos
15.
Nat Cancer ; 1(9): 894-908, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-35121952

RESUMEN

Argininosuccinate synthase (ASS1) downregulation in different tumors has been shown to support cell proliferation and yet, in several common cancer subsets ASS1 expression associates with poor patient prognosis. Here we demonstrate that ASS1 expression under glucose deprivation is induced by c-MYC, providing survival benefit by increasing nitric oxide synthesis and activating the gluconeogenic enzymes pyruvate carboxylase and phosphoenolpyruvate carboxykinase by S-nitrosylation. The resulting increased flux through gluconeogenesis enhances serine, glycine and subsequently purine synthesis. Notably, high ASS1-expressing breast cancer mice do not respond to immune checkpoint inhibitors and patients with breast cancer with high ASS1 have more metastases. We further find that inhibiting purine synthesis increases pyrimidine to purine ratio, elevates expression of the immunoproteasome and significantly enhances the response of autologous primary CD8+ T cells to anti-PD-1. These results suggest that treating patients with high-ASS1 cancers with purine synthesis inhibition is beneficial and may also sensitize them to immune checkpoint inhibition therapy.


Asunto(s)
Argininosuccinato Sintasa , Neoplasias de la Mama , Animales , Argininosuccinato Sintasa/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico , Ratones , Purinas
16.
Oncogene ; 39(1): 164-175, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31462712

RESUMEN

Citrin, encoded by SLC25A13 gene, is an inner mitochondrial transporter that is part of the malate-aspartate shuttle, which regulates the NAD+/NADH ratio between the cytosol and mitochondria. Citrullinemia type II (CTLN-II) is an inherited disorder caused by germline mutations in SLC25A13, manifesting clinically in growth failure that can be alleviated by dietary restriction of carbohydrates. The association of citrin with glycolysis and NAD+/NADH ratio led us to hypothesize that it may play a role in carcinogenesis. Indeed, we find that citrin is upregulated in multiple cancer types and is essential for supplementing NAD+ for glycolysis and NADH for oxidative phosphorylation. Consequently, citrin deficiency associates with autophagy, whereas its overexpression in cancer cells increases energy production and cancer invasion. Furthermore, based on the human deleterious mutations in citrin, we found a potential inhibitor of citrin that restricts cancerous phenotypes in cells. Collectively, our findings suggest that targeting citrin may be of benefit for cancer therapy.


Asunto(s)
Carcinogénesis/genética , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Neoplasias/genética , Carbohidratos/genética , Citrulinemia/genética , Citrulinemia/metabolismo , Citosol/metabolismo , Citosol/patología , Regulación Neoplásica de la Expresión Génica/genética , Mutación de Línea Germinal/genética , Glutamatos/farmacología , Ácido Glutámico/análogos & derivados , Ácido Glutámico/farmacología , Glucólisis/genética , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosforilación Oxidativa/efectos de los fármacos
17.
Cell Rep ; 29(8): 2144-2153.e7, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31747589

RESUMEN

Patients with germline mutations in the urea-cycle enzyme argininosuccinate lyase (ASL) are at risk for developing neurobehavioral and cognitive deficits. We find that ASL is prominently expressed in the nucleus locus coeruleus (LC), the central source of norepinephrine. Using natural history data, we show that individuals with ASL deficiency are at risk for developing attention deficits. By generating LC-ASL-conditional knockout (cKO) mice, we further demonstrate altered response to stressful stimuli with increased seizure reactivity in LC-ASL-cKO mice. Depletion of ASL in LC neurons leads to reduced amount and activity of tyrosine hydroxylase (TH) and to decreased catecholamines synthesis, due to decreased nitric oxide (NO) signaling. NO donors normalize catecholamine levels in the LC, seizure sensitivity, and the stress response in LC-ASL-cKO mice. Our data emphasize ASL importance for the metabolic regulation of LC function with translational relevance for ASL deficiency (ASLD) patients as well as for LC-related pathologies.


Asunto(s)
Argininosuccinatoliasa/metabolismo , Locus Coeruleus/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Trastornos Innatos del Ciclo de la Urea/metabolismo , Animales , Catecolaminas/metabolismo , Núcleo Celular/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo , Convulsiones/metabolismo
18.
J Inherit Metab Dis ; 42(6): 1147-1161, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30723942

RESUMEN

The first patients affected by argininosuccinic aciduria (ASA) were reported 60 years ago. The clinical presentation was initially described as similar to other urea cycle defects, but increasing evidence has shown overtime an atypical systemic phenotype with a paradoxical observation, that is, a higher rate of neurological complications contrasting with a lower rate of hyperammonaemic episodes. The disappointing long-term clinical outcomes of many of the patients have challenged the current standard of care and therapeutic strategy, which aims to normalize plasma ammonia and arginine levels. Interrogations have raised about the benefit of newborn screening or liver transplantation on the neurological phenotype. Over the last decade, novel discoveries enabled by the generation of new transgenic argininosuccinate lyase (ASL)-deficient mouse models have been achieved, such as, a better understanding of ASL and its close interaction with nitric oxide metabolism, ASL physiological role outside the liver, and the pathophysiological role of oxidative/nitrosative stress or excessive arginine treatment. Here, we present a collaborative review, which highlights these recent discoveries and novel emerging concepts about ASL role in human physiology, ASA clinical phenotype and geographic prevalence, limits of current standard of care and newborn screening, pathophysiology of the disease, and emerging novel therapies. We propose recommendations for monitoring of ASA patients. Ongoing research aims to better understand the underlying pathogenic mechanisms of the systemic disease to design novel therapies.


Asunto(s)
Aciduria Argininosuccínica , Animales , Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/diagnóstico , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Aciduria Argininosuccínica/terapia , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Hiperamonemia/terapia , Recién Nacido , Hígado/diagnóstico por imagen , Hígado/patología , Hígado/cirugía , Trasplante de Hígado , Ratones , Ratones Transgénicos , Tamizaje Neonatal/métodos , Tamizaje Neonatal/tendencias , Estrés Oxidativo/fisiología , Fenotipo
19.
Cell Death Differ ; 26(9): 1566-1581, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413783

RESUMEN

It is well accepted that malignant transformation is associated with unique metabolism. Malignant transformation involves a variety of cellular pathways that are associated with initiation and progression of the malignant process that remain to be deciphered still. Here we used a mouse model of mutant p53 that presents a stepwise progressive transformation of adult Mesenchymal Stem Cells (MSCs). While the established parental p53Mut-MSCs induce tumors, the parental p53WT-MSCs that were established in parallel, did not. Furthermore, tumor lines derived from the parental p53Mut-MSCs (p53Mut-MSC-TLs), exhibited yet a more aggressive transformed phenotype, suggesting exacerbation in tumorigenesis. Metabolic tracing of these various cell types, indicated that while malignant transformation is echoed by a direct augmentation in glycolysis, the more aggressive p53Mut-MSC-TLs demonstrate increased mitochondrial oxidation that correlates with morphological changes in mitochondria mass and function. Finally, we show that these changes are p53Mut-dependent. Computational transcriptional analysis identified a mitochondrial gene signature specifically downregulated upon knock/out of p53Mut in MSC-TLs. Our results suggest that stem cells exhibiting different state of malignancy are also associated with a different quantitative and qualitative metabolic profile in a p53Mut-dependent manner. This may provide important insights for cancer prognosis and the use of specific metabolic inhibitors in a personalized designed cancer therapy.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Glucólisis/genética , Humanos , Células Madre Mesenquimatosas/patología , Metaboloma/genética , Ratones , Mitocondrias/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo
20.
Cancer Res ; 79(3): 518-533, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30573518

RESUMEN

Downregulation of the urea cycle enzyme argininosuccinate synthase (ASS1) by either promoter methylation or by HIF1α is associated with increased metastasis and poor prognosis in multiple cancers. We have previously shown that in normoxic conditions, ASS1 downregulation facilitates cancer cell proliferation by increasing aspartate availability for pyrimidine synthesis by the enzyme complex CAD. Here we report that in hypoxia, ASS1 expression in cancerous cells is downregulated further by HIF1α-mediated induction of miR-224-5p, making the cells more invasive and dependent on upstream substrates of ASS1 for survival. ASS1 was downregulated under acidic conditions, and ASS1-depleted cancer cells maintained a higher intracellular pH (pHi), depended less on extracellular glutamine, and displayed higher glutathione levels. Depletion of substrates of urea cycle enzymes in ASS1-deficient cancers decreased cancer cell survival. Thus, ASS1 levels in cancer are differentially regulated in various environmental conditions to metabolically benefit cancer progression. Understanding these alterations may help uncover specific context-dependent cancer vulnerabilities that may be targeted for therapeutic purposes. SIGNIFICANCE: Cancer cells in an acidic or hypoxic environment downregulate the expression of the urea cycle enzyme ASS1, which provides them with a redox and pH advantage, resulting in better survival.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/3/518/F1.large.jpg.


Asunto(s)
Argininosuccinato Sintasa/metabolismo , Neoplasias/metabolismo , Adolescente , Adulto , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Niño , Regulación hacia Abajo , Perfilación de la Expresión Génica , Glutamina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias/enzimología , Neoplasias/patología , Osteosarcoma/metabolismo , Osteosarcoma/patología , Oxidación-Reducción , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA