Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(8): 1990-2019, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33811810

RESUMEN

The population is aging at a rate never seen before in human history. As the number of elderly adults grows, it is imperative we expand our understanding of the underpinnings of aging biology. Human lungs are composed of a unique panoply of cell types that face ongoing chemical, mechanical, biological, immunological, and xenobiotic stress over a lifetime. Yet, we do not fully appreciate the mechanistic drivers of lung aging and why age increases the risk of parenchymal lung disease, fatal respiratory infection, and primary lung cancer. Here, we review the molecular and cellular aspects of lung aging, local stress response pathways, and how the aging process predisposes to the pathogenesis of pulmonary disease. We place these insights into context of the COVID-19 pandemic and discuss how innate and adaptive immunity within the lung is altered with age.


Asunto(s)
Envejecimiento , Senescencia Celular , Enfermedades Pulmonares , Pulmón , Inmunidad Adaptativa , Anciano , Envejecimiento/inmunología , Envejecimiento/patología , COVID-19/inmunología , COVID-19/patología , Humanos , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Estrés Oxidativo
2.
Cell ; 183(7): 1848-1866.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301708

RESUMEN

Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.


Asunto(s)
Inmunidad , Neoplasias/inmunología , Neoplasias/metabolismo , Obesidad/metabolismo , Microambiente Tumoral , Adiposidad , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Células HEK293 , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Cinética , Linfocitos Infiltrantes de Tumor , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Análisis de Componente Principal , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteómica
3.
Cell ; 175(1): 101-116.e25, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30220459

RESUMEN

IDH1 mutations are common in low-grade gliomas and secondary glioblastomas and cause overproduction of (R)-2HG. (R)-2HG modulates the activity of many enzymes, including some that are linked to transformation and some that are probably bystanders. Although prior work on (R)-2HG targets focused on 2OG-dependent dioxygenases, we found that (R)-2HG potently inhibits the 2OG-dependent transaminases BCAT1 and BCAT2, likely as a bystander effect, thereby decreasing glutamate levels and increasing dependence on glutaminase for the biosynthesis of glutamate and one of its products, glutathione. Inhibiting glutaminase specifically sensitized IDH mutant glioma cells to oxidative stress in vitro and to radiation in vitro and in vivo. These findings highlight the complementary roles for BCATs and glutaminase in glutamate biosynthesis, explain the sensitivity of IDH mutant cells to glutaminase inhibitors, and suggest a strategy for maximizing the effectiveness of such inhibitors against IDH mutant gliomas.


Asunto(s)
Glioma/metabolismo , Ácido Glutámico/biosíntesis , Transaminasas/fisiología , Línea Celular Tumoral , Glioma/fisiopatología , Ácido Glutámico/efectos de los fármacos , Glutaratos/metabolismo , Glutaratos/farmacología , Homeostasis/efectos de los fármacos , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/fisiología , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/fisiología , Mutación , Oxidación-Reducción/efectos de los fármacos , Proteínas Gestacionales/genética , Proteínas Gestacionales/fisiología , Transaminasas/antagonistas & inhibidores , Transaminasas/genética
4.
Cell ; 166(3): 555-566, 2016 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-27471965

RESUMEN

Mitochondria are bioenergetic, biosynthetic, and signaling organelles that are integral in stress sensing to allow for cellular adaptation to the environment. Therefore, it is not surprising that mitochondria are important mediators of tumorigenesis, as this process requires flexibility to adapt to cellular and environmental alterations in addition to cancer treatments. Multiple aspects of mitochondrial biology beyond bioenergetics support transformation, including mitochondrial biogenesis and turnover, fission and fusion dynamics, cell death susceptibility, oxidative stress regulation, metabolism, and signaling. Thus, understanding mechanisms of mitochondrial function during tumorigenesis will be critical for the next generation of cancer therapeutics.


Asunto(s)
Carcinogénesis , Mitocondrias/fisiología , Animales , Humanos
6.
Cell ; 167(4): 985-1000.e21, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27881304

RESUMEN

Mitochondrial sirtuins, SIRT3-5, are NAD+-dependent deacylases and ADP-ribosyltransferases that are critical for stress responses. However, a comprehensive understanding of sirtuin targets, regulation of sirtuin activity, and the relationships between sirtuins remains a key challenge in mitochondrial physiology. Here, we employ systematic interaction proteomics to elucidate the mitochondrial sirtuin protein interaction landscape. This work reveals sirtuin interactions with numerous functional modules within mitochondria, identifies candidate sirtuin substrates, and uncovers a fundamental role for sequestration of SIRT3 by ATP synthase in mitochondrial homeostasis. In healthy mitochondria, a pool of SIRT3 binds ATP synthase, but upon matrix pH reduction with concomitant loss of mitochondrial membrane potential, SIRT3 dissociates. This release correlates with rapid deacetylation of matrix proteins, and SIRT3 is required for recovery of membrane potential. In vitro reconstitution experiments, as well as analysis of CRISPR/Cas9-engineered cells, indicate that pH-dependent SIRT3 release requires H135 in the ATP5O subunit of ATP synthase. Our SIRT3-5 interaction network provides a framework for discovering novel biological functions regulated by mitochondrial sirtuins.


Asunto(s)
Mitocondrias/metabolismo , Mapas de Interacción de Proteínas , Sirtuina 3/metabolismo , Acetilación , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales , Sirtuinas/clasificación , Sirtuinas/metabolismo
7.
Mol Cell ; 83(8): 1340-1349.e7, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37084714

RESUMEN

The glycerol-3-phosphate shuttle (G3PS) is a major NADH shuttle that regenerates reducing equivalents in the cytosol and produces energy in the mitochondria. Here, we demonstrate that G3PS is uncoupled in kidney cancer cells where the cytosolic reaction is ∼4.5 times faster than the mitochondrial reaction. The high flux through cytosolic glycerol-3-phosphate dehydrogenase (GPD) is required to maintain redox balance and support lipid synthesis. Interestingly, inhibition of G3PS by knocking down mitochondrial GPD (GPD2) has no effect on mitochondrial respiration. Instead, loss of GPD2 upregulates cytosolic GPD on a transcriptional level and promotes cancer cell proliferation by increasing glycerol-3-phosphate supply. The proliferative advantage of GPD2 knockdown tumor can be abolished by pharmacologic inhibition of lipid synthesis. Taken together, our results suggest that G3PS is not required to run as an intact NADH shuttle but is instead truncated to support complex lipid synthesis in kidney cancer.


Asunto(s)
Glicerol-3-Fosfato Deshidrogenasa (NAD+) , Neoplasias Renales , Lípidos , Humanos , Glicerol/metabolismo , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/genética , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/metabolismo , Glicerolfosfato Deshidrogenasa/genética , Glicerolfosfato Deshidrogenasa/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Lípidos/biosíntesis , NAD/metabolismo , Oxidación-Reducción , Fosfatos/metabolismo
8.
Mol Cell ; 81(18): 3708-3730, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34547235

RESUMEN

Lipids play crucial roles in signal transduction, contribute to the structural integrity of cellular membranes, and regulate energy metabolism. Questions remain as to which lipid species maintain metabolic homeostasis and which disrupt essential cellular functions, leading to metabolic disorders. Here, we discuss recent advances in understanding lipid metabolism with a focus on catabolism, synthesis, and signaling. Technical advances, including functional genomics, metabolomics, lipidomics, lipid-protein interaction maps, and advances in mass spectrometry, have uncovered new ways to prioritize molecular mechanisms mediating lipid function. By reviewing what is known about the distinct effects of specific lipid species in physiological pathways, we provide a framework for understanding newly identified targets regulating lipid homeostasis with implications for ameliorating metabolic diseases.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Enfermedades Metabólicas/metabolismo , Transducción de Señal/fisiología , Animales , Cromatina/metabolismo , Enfermedad , Metabolismo Energético/fisiología , Salud , Homeostasis/fisiología , Humanos , Inmunidad/fisiología , Lipidómica/métodos , Lípidos/fisiología , Enfermedades Metabólicas/fisiopatología , Metabolómica/métodos , Microbiota/fisiología
9.
Immunity ; 51(6): 980-981, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851904

RESUMEN

In a recent issue of Nature, Zhang et al. (2019) describe an additional histone post-translational modification, named histone lactylation. Following increased lactate production as a consequence of M1 polarization, histone lactylation regulates the induction of an M2-like phenotype in late stages of M1 macrophage activation to promote wound healing.


Asunto(s)
Histonas , Activación de Macrófagos , Regulación de la Expresión Génica , Macrófagos , Azúcares
10.
Cell ; 153(4): 840-54, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23663782

RESUMEN

Proliferating mammalian cells use glutamine as a source of nitrogen and as a key anaplerotic source to provide metabolites to the tricarboxylic acid cycle (TCA) for biosynthesis. Recently, mammalian target of rapamycin complex 1 (mTORC1) activation has been correlated with increased nutrient uptake and metabolism, but no molecular connection to glutaminolysis has been reported. Here, we show that mTORC1 promotes glutamine anaplerosis by activating glutamate dehydrogenase (GDH). This regulation requires transcriptional repression of SIRT4, the mitochondrial-localized sirtuin that inhibits GDH. Mechanistically, mTORC1 represses SIRT4 by promoting the proteasome-mediated destabilization of cAMP-responsive element binding 2 (CREB2). Thus, a relationship between mTORC1, SIRT4, and cancer is suggested by our findings. Indeed, SIRT4 expression is reduced in human cancer, and its overexpression reduces cell proliferation, transformation, and tumor development. Finally, our data indicate that targeting nutrient metabolism in energy-addicted cancers with high mTORC1 signaling may be an effective therapeutic approach.


Asunto(s)
Glutamina/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias/metabolismo , Sirtuinas/metabolismo , Factores de Transcripción Activadores/metabolismo , Animales , Proliferación Celular , Embrión de Mamíferos/citología , Metabolismo Energético , Glutamato Deshidrogenasa/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos , Trasplante de Neoplasias , Neoplasias/patología , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética , Trasplante Heterólogo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación
11.
Cell ; 150(1): 179-93, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22770219

RESUMEN

Aberrant Skp2 signaling has been implicated as a driving event in tumorigenesis. Although the underlying molecular mechanisms remain elusive, cytoplasmic Skp2 correlates with more aggressive forms of breast and prostate cancers. Here, we report that Skp2 is acetylated by p300 at K68 and K71, which is a process that can be antagonized by the SIRT3 deacetylase. Inactivation of SIRT3 leads to elevated Skp2 acetylation, which leads to increased Skp2 stability through impairment of the Cdh1-mediated proteolysis pathway. As a result, Skp2 oncogenic function is increased, whereby cells expressing an acetylation-mimetic mutant display enhanced cellular proliferation and tumorigenesis in vivo. Moreover, acetylation of Skp2 in the nuclear localization signal (NLS) promotes its cytoplasmic retention, and cytoplasmic Skp2 enhances cellular migration through ubiquitination and destruction of E-cadherin. Thus, our study identifies an acetylation-dependent regulatory mechanism governing Skp2 oncogenic function and provides insight into how cytoplasmic Skp2 controls cellular migration.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Neoplasias de la Próstata/patología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Quinasa de la Caseína I/metabolismo , Línea Celular Tumoral , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Humanos , Lisina/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Neoplasias de la Próstata/metabolismo , Procesamiento Proteico-Postraduccional , Señales de Clasificación de Proteína , Proteínas Quinasas Asociadas a Fase-S/química , Proteínas Quinasas Asociadas a Fase-S/genética , Alineación de Secuencia , Ubiquitinación
12.
Cell ; 149(1): 49-62, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22401813

RESUMEN

Decremental loss of PTEN results in cancer susceptibility and tumor progression. PTEN elevation might therefore be an attractive option for cancer prevention and therapy. We have generated several transgenic mouse lines with PTEN expression elevated to varying levels by taking advantage of bacterial artificial chromosome (BAC)-mediated transgenesis. The "Super-PTEN" mutants are viable and show reduced body size due to decreased cell number, with no effect on cell size. Unexpectedly, PTEN elevation at the organism level results in healthy metabolism characterized by increased energy expenditure and reduced body fat accumulation. Cells derived from these mice show reduced glucose and glutamine uptake and increased mitochondrial oxidative phosphorylation and are resistant to oncogenic transformation. Mechanistically we find that PTEN elevation orchestrates this metabolic switch by regulating PI3K-dependent and -independent pathways and negatively impacting two of the most pronounced metabolic features of tumor cells: glutaminolysis and the Warburg effect.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Tamaño Corporal , Recuento de Células , Proliferación Celular , Respiración de la Célula , Metabolismo Energético , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
13.
Mol Cell ; 72(4): 610-624, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30444998

RESUMEN

Growing appreciation of the diversity of post-translational modifications (PTMs) in the mitochondria necessitates reevaluation of the roles these modifications play in both health and disease. Compared to the cytosol and nucleus, the mitochondrial proteome is highly acylated, and remodeling of the mitochondrial "acylome" is a key adaptive mechanism that regulates fundamental aspects of mitochondrial biology. It is clear that we need to understand the underlying chemistry that regulates mitochondrial acylation, as well as how chemical properties of the acyl chain impact biological functions. Here, we dissect the sources of PTMs in the mitochondria, review major mitochondrial pathways that control levels of PTMs, and highlight how sirtuin enzymes respond to the bioenergetic state of the cell via NAD+ availability to regulate mitochondrial biology. By providing a framework connecting the chemistry of these modifications, their biochemical consequences, and the pathways that regulate the levels of acyl PTMs, we will gain a deeper understanding of the physiological significance of mitochondrial acylation and its role in mitochondrial adaptation.


Asunto(s)
Acilación/fisiología , Mitocondrias/metabolismo , Mitocondrias/fisiología , Humanos , Lisina/metabolismo , Proteínas Mitocondriales , Procesamiento Proteico-Postraduccional , Sirtuinas/fisiología
14.
Mol Cell ; 69(5): 729-743.e7, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29499131

RESUMEN

MCL-1 is a BCL-2 family protein implicated in the development and chemoresistance of human cancer. Unlike its anti-apoptotic homologs, Mcl-1 deletion has profound physiologic consequences, indicative of a broader role in homeostasis. We report that the BCL-2 homology 3 (BH3) α helix of MCL-1 can directly engage very long-chain acyl-CoA dehydrogenase (VLCAD), a key enzyme of the mitochondrial fatty acid ß-oxidation (FAO) pathway. Proteomic analysis confirmed that the mitochondrial matrix isoform of MCL-1 (MCL-1Matrix) interacts with VLCAD. Mcl-1 deletion, or eliminating MCL-1Matrix alone, selectively deregulated long-chain FAO, causing increased flux through the pathway in response to nutrient deprivation. Transient elevation in MCL-1 upon serum withdrawal, a striking increase in MCL-1 BH3/VLCAD interaction upon palmitic acid titration, and direct modulation of enzymatic activity by the MCL-1 BH3 α helix are consistent with dynamic regulation. Thus, the MCL-1 BH3 interaction with VLCAD revealed a separable, gain-of-function role for MCL-1 in the regulation of lipid metabolism.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Metabolismo de los Lípidos/fisiología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ácido Palmítico/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Animales , Línea Celular , Ratones , Ratones Noqueados , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Oxidación-Reducción , Estructura Secundaria de Proteína
15.
Mol Cell Proteomics ; 23(7): 100801, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38880243

RESUMEN

T cell activation is a complex biological process of naive cells maturing into effector cells. Proteomic and phospho-proteomic approaches have provided critical insights into this process, yet it is not always clear how changes in individual proteins or phosphorylation sites have functional significance. Here, we developed the Phosphorylation Integrated Thermal Shift Assay (PITSA) that combines the measurement of protein or phosphorylation site abundance and thermal stability into a single tandem mass tags experiment and apply this method to study T cell activation. We quantified the abundance and thermal stability of over 7500 proteins and 5000 phosphorylation sites and identified significant differences in chromatin-related, TCR signaling, DNA repair, and proliferative phosphoproteins. PITSA may be applied to a wide range of biological contexts to generate hypotheses as to which proteins or phosphorylation sites are functionally regulated in a given system as well as the mechanisms by which this regulation may occur.


Asunto(s)
Activación de Linfocitos , Proteómica , Linfocitos T , Fosforilación , Linfocitos T/metabolismo , Proteómica/métodos , Fosfoproteínas/metabolismo , Animales , Humanos , Estabilidad Proteica , Transducción de Señal , Espectrometría de Masas en Tándem , Ratones
16.
J Allergy Clin Immunol ; 153(1): 28-41, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37778472

RESUMEN

Regulatory T cells expressing the transcription factor forkhead box protein 3 mediate peripheral immune tolerance both to self-antigens and to the commensal flora. Their defective function due to inborn errors of immunity or acquired insults is associated with a broad range of autoimmune and immune dysregulatory diseases. Although their function in suppressing autoimmunity and enforcing commensalism is established, a broader role for regulatory T cells in tissue repair and metabolic regulation has emerged, enabled by unique programs of tissue adaptability and specialization. In this review, we focus on the myriad roles played by regulatory T cells in immunologic tolerance and host homeostasis and the potential to harness these cells in novel therapeutic approaches to human diseases.


Asunto(s)
Enfermedades Autoinmunes , Enfermedades del Sistema Inmune , Humanos , Linfocitos T Reguladores , Tolerancia Inmunológica , Enfermedades del Sistema Inmune/metabolismo , Autoinmunidad , Factores de Transcripción Forkhead
17.
Cell ; 137(3): 560-70, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19410549

RESUMEN

Sirtuins are NAD-dependent protein deacetylases that connect metabolism and aging. In mammals, there are seven sirtuins (SIRT1-7), three of which are associated with mitochondria. Here, we show that SIRT5 localizes in the mitochondrial matrix and interacts with carbamoyl phosphate synthetase 1 (CPS1), an enzyme, catalyzing the initial step of the urea cycle for ammonia detoxification and disposal. SIRT5 deacetylates CPS1 and upregulates its activity. During fasting, NAD in liver mitochondria increases, thereby triggering SIRT5 deacetylation of CPS1 and adaptation to the increase in amino acid catabolism. Indeed, SIRT5 KO mice fail to upregulate CPS1 activity and show elevated blood ammonia during fasting. Similar effects occur during long-term calorie restriction or a high protein diet. These findings demonstrate SIRT5 plays a pivotal role in ammonia detoxification and disposal by activating CPS1.


Asunto(s)
Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/metabolismo , Sirtuinas/metabolismo , Amoníaco/metabolismo , Animales , Células Cultivadas , Activación Enzimática , Humanos , Ratones , Especificidad por Sustrato
18.
Nature ; 560(7716): 102-106, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30022159

RESUMEN

Thermogenesis by brown and beige adipose tissue, which requires activation by external stimuli, can counter metabolic disease1. Thermogenic respiration is initiated by adipocyte lipolysis through cyclic AMP-protein kinase A signalling; this pathway has been subject to longstanding clinical investigation2-4. Here we apply a comparative metabolomics approach and identify an independent metabolic pathway that controls acute activation of adipose tissue thermogenesis in vivo. We show that substantial and selective accumulation of the tricarboxylic acid cycle intermediate succinate is a metabolic signature of adipose tissue thermogenesis upon activation by exposure to cold. Succinate accumulation occurs independently of adrenergic signalling, and is sufficient to elevate thermogenic respiration in brown adipocytes. Selective accumulation of succinate may be driven by a capacity of brown adipocytes to sequester elevated circulating succinate. Furthermore, brown adipose tissue thermogenesis can be initiated by systemic administration of succinate in mice. Succinate from the extracellular milieu is rapidly metabolized by brown adipocytes, and its oxidation by succinate dehydrogenase is required for activation of thermogenesis. We identify a mechanism whereby succinate dehydrogenase-mediated oxidation of succinate initiates production of reactive oxygen species, and drives thermogenic respiration, whereas inhibition of succinate dehydrogenase supresses thermogenesis. Finally, we show that pharmacological elevation of circulating succinate drives UCP1-dependent thermogenesis by brown adipose tissue in vivo, which stimulates robust protection against diet-induced obesity and improves glucose tolerance. These findings reveal an unexpected mechanism for control of thermogenesis, using succinate as a systemically-derived thermogenic molecule.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Ácido Succínico/metabolismo , Termogénesis/fisiología , Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/enzimología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/metabolismo , Animales , Femenino , Masculino , Metabolómica , Ratones , Obesidad/metabolismo , Obesidad/prevención & control , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismo , Ácido Succínico/farmacología , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/metabolismo
19.
Mol Cell ; 63(6): 1006-20, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27635760

RESUMEN

While much research has examined the use of glucose and glutamine by tumor cells, many cancers instead prefer to metabolize fats. Despite the pervasiveness of this phenotype, knowledge of pathways that drive fatty acid oxidation (FAO) in cancer is limited. Prolyl hydroxylase domain proteins hydroxylate substrate proline residues and have been linked to fuel switching. Here, we reveal that PHD3 rapidly triggers repression of FAO in response to nutrient abundance via hydroxylation of acetyl-coA carboxylase 2 (ACC2). We find that PHD3 expression is strongly decreased in subsets of cancer including acute myeloid leukemia (AML) and is linked to a reliance on fat catabolism regardless of external nutrient cues. Overexpressing PHD3 limits FAO via regulation of ACC2 and consequently impedes leukemia cell proliferation. Thus, loss of PHD3 enables greater utilization of fatty acids but may also serve as a metabolic and therapeutic liability by indicating cancer cell susceptibility to FAO inhibition.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Ácidos Grasos/metabolismo , Regulación Neoplásica de la Expresión Génica , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Leucemia Mieloide Aguda/metabolismo , Prolina/metabolismo , Acetil-CoA Carboxilasa/antagonistas & inhibidores , Acetil-CoA Carboxilasa/química , Acetil-CoA Carboxilasa/genética , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/química , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Células K562 , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Masculino , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos NOD , Modelos Moleculares , Trasplante de Neoplasias , Oxidación-Reducción , Prolina/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Homología Estructural de Proteína , Análisis de Supervivencia
20.
Anal Chem ; 95(30): 11243-11253, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37469028

RESUMEN

Matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) is a powerful analytical technique that provides spatially preserved detection and quantification of analytes in tissue specimens. However, clinical translation still requires improved throughput, precision, and accuracy. To accomplish this, we created "Chemical QuantArray", a gelatin tissue microarray (TMA) mold filled with serial dilutions of isotopically labeled endogenous metabolite standards. The mold is then cryo-sectioned onto a tissue homogenate to produce calibration curves. To improve precision and accuracy, we automatically remove pixels outside of each TMA well and investigated several intensity normalizations, including the utilization of a second stable isotope internal standard (IS). Chemical QuantArray enables the quantification of several endogenous metabolites over a wide dynamic range and significantly improve over current approaches. The technique reduces the space needed on the MALDI slides for calibration standards by approximately 80%. Furthermore, removal of empty pixels and normalization to an internal standard or matrix peak provided precision (<20% RSD) and accuracy (<20% DEV). Finally, we demonstrate the applicability of Chemical QuantArray by quantifying multiple purine metabolites in 14 clinical tumor specimens using a single MALDI slide. Chemical QuantArray improves the analytical characteristics and practical feasibility of MALDI-MSI metabolite quantification in clinical and translational applications.


Asunto(s)
Diagnóstico por Imagen , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Calibración , Estándares de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA