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













Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38687612

RESUMEN

Fetuses with growth restriction (FGR) have an early activation of hepatic glucose production (HGP), a hallmark of type 2 diabetes (T2D). Here we used fetal hepatic catheterization to directly measure HGP and substrate flux in an FGR sheep model. We hypothesized that FGR fetuses would have increased hepatic lactate and amino acid uptake to support increased HGP. Indeed, FGR compared to normal (CON) fetuses had increased HGP and activation of gluconeogenic genes. Unexpectedly, hepatic pyruvate output was increased while hepatic lactate and gluconeogenic amino acid uptake rates were decreased in FGR fetal liver. Hepatic oxygen consumption and total substrate uptake rates were lower. In FGR liver tissue, metabolite abundance, 13C-metabolite labeling, enzyme activity, and gene expression support decreased pyruvate oxidation and increased lactate production. Isolated hepatocytes from FGR fetuses had greater intrinsic capacity for lactate-fueled glucose production. FGR livers also had lower energy (ATP) and redox state (NADH:NAD+). Thus, reduced hepatic oxidative metabolism may make carbons available for increased HGP but also produces nutrient and energetic stress in FGR fetal liver. Intrinsic programming of these pathways regulating HGP in the FGR fetus may underlie increased HGP and T2D risk postnatally.

2.
Cell Commun Signal ; 21(1): 331, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37985999

RESUMEN

INTRODUCTION: Inflammation plays a significant role in various cancers, including lung cancer, where the inflammatory cytokine IL-1ß is often elevated in the tumor microenvironment. Patients with lung adenocarcinoma show higher levels of serum IL-1ß compared to healthy individual. Moreover, IL-1ß blockade reduces the incidence and mortality of lung cancer. Our prior studies revealed that alveolar type-II cells, the precursors for lung adenocarcinoma, display an induction in the expression of the enzyme tryptophan 2,3-dioxygenase (TDO2) during normal lung development. This induction of TDO2 coincides with an increase in IL-1ß levels and is likely caused by IL-1ß. Given that cancer cells can co-opt developmentally regulated pathways, we hypothesized that IL-1ß may exert its pro-tumoral function by stimulating TDO2 and indoleamine 2, 3-dioxygenase-1 (IDO1), parallel enzymes involved in the conversion of tryptophan (Trp) into the immune-suppressive oncometabolite kynurenine (Kyn). Our goal was to determine whether IL-1ß is a common upstream regulator of immune checkpoint regulators. METHODS: To determine whether IL-1ß regulates IDO1, TDO2, PD-L1, and PD-L2, we measured mRNA and protein levels in lung adenocarcinoma cells lines (A549, H1792, H1838, H2347, H2228, HCC364 and HCC827) grown in 2D or 3D and in immortalized normal lung epithelial cells (HBEC3-KT and HSAEC1-KT). To determine the importance of the NFκB pathway in mediating IL-1ß -regulated cellular effects, we used siRNA to knockdown RelA/p65 in IL-1ß treated cells. The levels of Trp and Kyn in the IL-1ß-treated cells and media were measured by mass spectrometry. RESULTS: Upon IL-1ß stimulation, lung adenocarcinoma cells exhibited significant increases in IDO1 mRNA and protein levels, a response that depended on the NFκB pathway. Interestingly, this induction was more pronounced in 3D spheroid cultures compared to monolayer cultures and was not observed in normal immortalized lung epithelial cells. Furthermore, the conversion of Trp to Kyn increased in cells exposed to IL-1ß, aligning with the heightened IDO1 expression. Remarkably, IL-1ß also upregulated the expression of programmed death ligand-1 (PD-L1) and PD-L2 in multiple cell lines, indicating that IL-1ß triggers parallel immune-suppressive mechanisms in lung adenocarcinoma cells. CONCLUSIONS: Our studies demonstrate that lung adenocarcinoma cells, but not normal immortalized lung epithelial cells, respond to IL-1ß signaling by inducing the expression of parallel immune checkpoint proteins that have the potential to promote immune evasion. Video Abstract.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Adenocarcinoma del Pulmón/metabolismo , Antígeno B7-H1/metabolismo , Quinurenina/metabolismo , Neoplasias Pulmonares/patología , ARN Mensajero , Triptófano , Microambiente Tumoral
3.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37883062

RESUMEN

Metabolism is crucial for development through supporting cell growth, energy production, establishing cell identity, developmental signaling and pattern formation. In many model systems, development occurs alongside metabolic transitions as cells differentiate and specialize in metabolism that supports new functions. Some cells exhibit metabolic flexibility to circumvent mutations or aberrant signaling, whereas other cell types require specific nutrients for developmental progress. Metabolic gradients and protein modifications enable pattern formation and cell communication. On an organism level, inadequate nutrients or stress can limit germ cell maturation, implantation and maturity through diapause, which slows metabolic activities until embryonic activation under improved environmental conditions.


Asunto(s)
Diapausa , Animales , Diapausa/fisiología , Implantación del Embrión/genética , Transducción de Señal , Metabolismo Energético
4.
Biomed Pharmacother ; 162: 114614, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37068330

RESUMEN

The continuing heavy toll of the COVID-19 pandemic necessitates development of therapeutic options. We adopted structure-based drug repurposing to screen FDA-approved drugs for inhibitory effects against main protease enzyme (Mpro) substrate-binding pocket of SARS-CoV-2 for non-covalent and covalent binding. Top candidates were screened against infectious SARS-CoV-2 in a cell-based viral replication assay. Promising candidates included atovaquone, mebendazole, ouabain, dronedarone, and entacapone, although atovaquone and mebendazole were the only two candidates with IC50s that fall within their therapeutic plasma concentration. Additionally, we performed Mpro assays on the top hits, which demonstrated inhibition of Mpro by dronedarone (IC50 18 µM), mebendazole (IC50 19 µM) and entacapone (IC50 9 µM). Atovaquone showed only modest Mpro inhibition, and thus we explored other potential mechanisms. Although atovaquone is Dihydroorotate dehydrogenase (DHODH) inhibitor, we did not observe inhibition of DHODH at the respective SARS-CoV-2 IC50. Metabolomic profiling of atovaquone treated cells showed dysregulation of purine metabolism pathway metabolite, where ecto-5'-nucleotidase (NT5E) was downregulated by atovaquone at concentrations equivalent to its antiviral IC50. Atovaquone and mebendazole are promising candidates with SARS-CoV-2 antiviral activity. While mebendazole does appear to target Mpro, atovaquone may inhibit SARS-CoV-2 viral replication by targeting host purine metabolism.


Asunto(s)
Antivirales , COVID-19 , Humanos , Antivirales/farmacología , SARS-CoV-2 , Dihidroorotato Deshidrogenasa , Reposicionamiento de Medicamentos , Dronedarona/farmacología , Pandemias , Atovacuona/farmacología , Mebendazol/farmacología , Purinas/farmacología , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/farmacología , Simulación de Dinámica Molecular
5.
Elife ; 122023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36881526

RESUMEN

Malonyl-CoA-acyl carrier protein transacylase (MCAT) is an enzyme involved in mitochondrial fatty acid synthesis (mtFAS) and catalyzes the transfer of the malonyl moiety of malonyl-CoA to the mitochondrial acyl carrier protein (ACP). Previously, we showed that loss-of-function of mtFAS genes, including Mcat, is associated with severe loss of electron transport chain (ETC) complexes in mouse immortalized skeletal myoblasts (Nowinski et al., 2020). Here, we report a proband presenting with hypotonia, failure to thrive, nystagmus, and abnormal brain MRI findings. Using whole exome sequencing, we identified biallelic variants in MCAT. Protein levels for NDUFB8 and COXII, subunits of complex I and IV respectively, were markedly reduced in lymphoblasts and fibroblasts, as well as SDHB for complex II in fibroblasts. ETC enzyme activities were decreased in parallel. Re-expression of wild-type MCAT rescued the phenotype in patient fibroblasts. This is the first report of a patient with MCAT pathogenic variants and combined oxidative phosphorylation deficiency.


Asunto(s)
S-Maloniltransferasa de la Proteína Transportadora de Grupos Acilo , Enfermedades Mitocondriales , Animales , Ratones , Adipogénesis , Encéfalo , Mitocondrias , Enfermedades Mitocondriales/genética , S-Maloniltransferasa de la Proteína Transportadora de Grupos Acilo/genética
6.
Sci Adv ; 8(35): eabn9550, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36044570

RESUMEN

In mice and humans with cancer, intravenous 13C-glucose infusion results in 13C labeling of tumor tricarboxylic acid (TCA) cycle intermediates, indicating that pyruvate oxidation in the TCA cycle occurs in tumors. The TCA cycle is usually coupled to the electron transport chain (ETC) because NADH generated by the cycle is reoxidized to NAD+ by the ETC. However, 13C labeling does not directly report ETC activity, and other pathways can oxidize NADH, so the ETC's role in these labeling patterns is unverified. We examined the impact of the ETC complex I inhibitor IACS-010759 on tumor 13C labeling. IACS-010759 suppresses TCA cycle labeling from glucose or lactate and increases labeling from glutamine. Cancer cells expressing yeast NADH dehydrogenase-1, which recycles NADH to NAD+ independently of complex I, display normalized labeling when complex I is inhibited, indicating that cancer cell ETC activity regulates TCA cycle metabolism and 13C labeling from multiple nutrients.


Asunto(s)
Complejo I de Transporte de Electrón , Glucosa , Glutamina , Neoplasias , Animales , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Isótopos , Ratones , NAD/metabolismo , Neoplasias/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
7.
Nature ; 604(7905): 349-353, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35388219

RESUMEN

Mammalian embryogenesis requires rapid growth and proper metabolic regulation1. Midgestation features increasing oxygen and nutrient availability concomitant with fetal organ development2,3. Understanding how metabolism supports development requires approaches to observe metabolism directly in model organisms in utero. Here we used isotope tracing and metabolomics to identify evolving metabolic programmes in the placenta and embryo during midgestation in mice. These tissues differ metabolically throughout midgestation, but we pinpointed gestational days (GD) 10.5-11.5 as a transition period for both placenta and embryo. Isotope tracing revealed differences in carbohydrate metabolism between the tissues and rapid glucose-dependent purine synthesis, especially in the embryo. Glucose's contribution to the tricarboxylic acid (TCA) cycle rises throughout midgestation in the embryo but not in the placenta. By GD12.5, compartmentalized metabolic programmes are apparent within the embryo, including different nutrient contributions to the TCA cycle in different organs. To contextualize developmental anomalies associated with Mendelian metabolic defects, we analysed mice deficient in LIPT1, the enzyme that activates 2-ketoacid dehydrogenases related to the TCA cycle4,5. LIPT1 deficiency suppresses TCA cycle metabolism during the GD10.5-GD11.5 transition, perturbs brain, heart and erythrocyte development and leads to embryonic demise by GD11.5. These data document individualized metabolic programmes in developing organs in utero.


Asunto(s)
Ciclo del Ácido Cítrico , Desarrollo Fetal , Metabolómica , Placenta , Animales , Embrión de Mamíferos/metabolismo , Femenino , Glucosa/metabolismo , Mamíferos/metabolismo , Ratones , Placenta/metabolismo , Embarazo
8.
J Inherit Metab Dis ; 44(4): 949-960, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33855712

RESUMEN

Glutamyl-tRNA synthetase 2 (encoded by EARS2) is a mitochondrial aminoacyl-tRNA synthetase required to translate the 13 subunits of the electron transport chain encoded by the mitochondrial DNA. Pathogenic EARS2 variants cause combined oxidative phosphorylation deficiency, subtype 12 (COXPD12), an autosomal recessive disorder involving lactic acidosis, intellectual disability, and other features of mitochondrial compromise. Patients with EARS2 deficiency present with variable phenotypes ranging from neonatal lethality to a mitigated disease with clinical improvement in early childhood. Here, we report a neonate homozygous for a rare pathogenic variant in EARS2 (c.949G>T; p.G317C). Metabolomics in primary fibroblasts from this patient revealed expected abnormalities in TCA cycle metabolites, as well as numerous changes in purine, pyrimidine, and fatty acid metabolism. To examine genotype-phenotype correlations in COXPD12, we compared the metabolic impact of reconstituting these fibroblasts with wild-type EARS2 versus four additional EARS2 variants from COXPD12 patients with varying clinical severity. Metabolomics identified a group of signature metabolites, mostly from the TCA cycle and amino acid metabolism, that discriminate between EARS2 variants causing relatively mild and severe COXPD12. Taken together, these findings indicate that metabolomics in patient-derived fibroblasts may help establish genotype-phenotype correlations in EARS2 deficiency and likely other mitochondrial disorders.


Asunto(s)
Variación Genética/genética , Glutamato-ARNt Ligasa/genética , Leucoencefalopatías/genética , Errores Innatos del Metabolismo/genética , Acidosis Láctica/etiología , Aminoacil-ARNt Sintetasas/genética , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Glutamato-ARNt Ligasa/metabolismo , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/etiología , Leucoencefalopatías/metabolismo , Masculino , Errores Innatos del Metabolismo/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación
9.
Nat Metab ; 3(4): 571-585, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33833463

RESUMEN

Nicotinamide adenine dinucleotide phosphate (NADP+) is vital to produce NADPH, a principal supplier of reducing power for biosynthesis of macromolecules and protection against oxidative stress. NADPH exists in separate pools, in both the cytosol and mitochondria; however, the cellular functions of mitochondrial NADPH are incompletely described. Here, we find that decreasing mitochondrial NADP(H) levels through depletion of NAD kinase 2 (NADK2), an enzyme responsible for production of mitochondrial NADP+, renders cells uniquely proline auxotrophic. Cells with NADK2 deletion fail to synthesize proline, due to mitochondrial NADPH deficiency. We uncover the requirement of mitochondrial NADPH and NADK2 activity for the generation of the pyrroline-5-carboxylate metabolite intermediate as the bottleneck step in the proline biosynthesis pathway. Notably, after NADK2 deletion, proline is required to support nucleotide and protein synthesis, making proline essential for the growth and proliferation of NADK2-deficient cells. Thus, we highlight proline auxotrophy in mammalian cells and discover that mitochondrial NADPH is essential to enable proline biosynthesis.


Asunto(s)
Proliferación Celular , Mitocondrias/metabolismo , NADP/metabolismo , Prolina/biosíntesis , Animales , Ciclo Celular/genética , Humanos , Ratones , Ratones Noqueados , Consumo de Oxígeno , Páncreas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Elife ; 92020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32804083

RESUMEN

Cells harbor two systems for fatty acid synthesis, one in the cytoplasm (catalyzed by fatty acid synthase, FASN) and one in the mitochondria (mtFAS). In contrast to FASN, mtFAS is poorly characterized, especially in higher eukaryotes, with the major product(s), metabolic roles, and cellular function(s) being essentially unknown. Here we show that hypomorphic mtFAS mutant mouse skeletal myoblast cell lines display a severe loss of electron transport chain (ETC) complexes and exhibit compensatory metabolic activities including reductive carboxylation. This effect on ETC complexes appears to be independent of protein lipoylation, the best characterized function of mtFAS, as mutants lacking lipoylation have an intact ETC. Finally, mtFAS impairment blocks the differentiation of skeletal myoblasts in vitro. Together, these data suggest that ETC activity in mammals is profoundly controlled by mtFAS function, thereby connecting anabolic fatty acid synthesis with the oxidation of carbon fuels.


In human, plant and other eukaryotic cells, fats are an important source of energy and also play many other roles including waterproofing, thermal insulation and energy storage. Eukaryotic cells have two systems that make the building blocks of fats (known as fatty acids) and one of these systems, called the mtFAS pathway, operates in small compartments known as mitochondria. This pathway only has one known product, a small fat molecule called lipoic acid, which mitochondria attach to several enzymes to allow them to work properly. The main role of mitochondria is to break down fats and other molecules to release chemical energy that powers many processes in cells. They achieve this using large groups of proteins known as ETC complexes. To build these complexes, families of proteins known as ETC assembly factors carefully coordinate the assembly of many proteins and small molecules into specific structures. However, it remains unclear precisely how this process works. Here, Nowinski et al. used a gene editing technique to mutate the genes encoding three enzymes in the mtFAS pathway in mammalian cells. The experiments found that the mutant cells had fewer ETC complexes and seemed to be less able to break down fats and other molecules than 'normal' cells. Furthermore, a family of ETC assembly factors were less stable in the mutant cells. These findings suggest that the mtFAS pathway controls how mitochondria assemble ETC complexes. Further experiments indicated that lipoic acid is not involved in the assembly of ETC complexes and that the mtFAS pathway produces another, as yet unidentified, product that regulates this process, instead. MEPAN syndrome is a rare neurological disorder that leads to progressive loss of control of movement, slurred speech and impaired vision in children. Patients with this syndrome have genetic mutations affecting components of the mtFAS pathway, therefore, a better understanding of how the pathway works may help researchers develop new treatments in the future. More broadly, these findings will have important ramifications for many other situations in which the activity of ETC complexes in mitochondria is modified.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Ácidos Grasos/biosíntesis , Mitocondrias/metabolismo , Mioblastos/fisiología , Animales , Diferenciación Celular , Línea Celular , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Células HEK293 , Humanos , Lipoilación/genética , Ratones , Oxidación-Reducción
11.
Science ; 368(6487)2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32273439

RESUMEN

Metabolic reprogramming is a hallmark of malignancy. As our understanding of the complexity of tumor biology increases, so does our appreciation of the complexity of tumor metabolism. Metabolic heterogeneity among human tumors poses a challenge to developing therapies that exploit metabolic vulnerabilities. Recent work also demonstrates that the metabolic properties and preferences of a tumor change during cancer progression. This produces distinct sets of vulnerabilities between primary tumors and metastatic cancer, even in the same patient or experimental model. We review emerging concepts about metabolic reprogramming in cancer, with particular attention on why metabolic properties evolve during cancer progression and how this information might be used to develop better therapeutic strategies.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Animales , Progresión de la Enfermedad , Humanos , Ratones , Terapia Molecular Dirigida , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/terapia , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología
12.
Nature ; 577(7788): 115-120, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31853067

RESUMEN

Metastasis requires cancer cells to undergo metabolic changes that are poorly understood1-3. Here we show that metabolic differences among melanoma cells confer differences in metastatic potential as a result of differences in the function of the MCT1 transporter. In vivo isotope tracing analysis in patient-derived xenografts revealed differences in nutrient handling between efficiently and inefficiently metastasizing melanomas, with circulating lactate being a more prominent source of tumour lactate in efficient metastasizers. Efficient metastasizers had higher levels of MCT1, and inhibition of MCT1 reduced lactate uptake. MCT1 inhibition had little effect on the growth of primary subcutaneous tumours, but resulted in depletion of circulating melanoma cells and reduced the metastatic disease burden in patient-derived xenografts and in mouse melanomas. In addition, inhibition of MCT1 suppressed the oxidative pentose phosphate pathway and increased levels of reactive oxygen species. Antioxidants blocked the effects of MCT1 inhibition on metastasis. MCT1high and MCT1-/low cells from the same melanomas had similar capacities to form subcutaneous tumours, but MCT1high cells formed more metastases after intravenous injection. Metabolic differences among cancer cells thus confer differences in metastatic potential as metastasizing cells depend on MCT1 to manage oxidative stress.


Asunto(s)
Melanoma/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Humanos , Melanoma/genética , Melanoma/secundario , Ratones , Transportadores de Ácidos Monocarboxílicos/genética , Estrés Oxidativo , Simportadores/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cell Rep ; 27(5): 1376-1386.e6, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31042466

RESUMEN

Inborn errors of metabolism (IEMs) link metabolic defects to human phenotypes. Modern genomics has accelerated IEM discovery, but assessing the impact of genomic variants is still challenging. Here, we integrate genomics and metabolomics to identify a cause of lactic acidosis and epilepsy. The proband is a compound heterozygote for variants in LIPT1, which encodes the lipoyltransferase required for 2-ketoacid dehydrogenase (2KDH) function. Metabolomics reveals abnormalities in lipids, amino acids, and 2-hydroxyglutarate consistent with loss of multiple 2KDHs. Homozygous knockin of a LIPT1 mutation reduces 2KDH lipoylation in utero and results in embryonic demise. In patient fibroblasts, defective 2KDH lipoylation and function are corrected by wild-type, but not mutant, LIPT1 alleles. Isotope tracing reveals that LIPT1 supports lipogenesis and balances oxidative and reductive glutamine metabolism. Altogether, the data extend the role of LIPT1 in metabolic regulation and demonstrate how integrating genomics and metabolomics can uncover broader aspects of IEM pathophysiology.


Asunto(s)
Acidosis Láctica/metabolismo , Aciltransferasas/genética , Mutación con Pérdida de Función , Acidosis Láctica/genética , Acidosis Láctica/patología , Aciltransferasas/metabolismo , Animales , Células Cultivadas , Niño , Ácidos Grasos/metabolismo , Femenino , Fibroblastos/metabolismo , Glutamina/metabolismo , Glutaratos/metabolismo , Humanos , Lipogénesis , Lipoilación , Masculino , Ratones , Oxígeno/metabolismo
14.
J Biol Chem ; 293(20): 7522-7530, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29191830

RESUMEN

Lipoic acid is an essential cofactor for mitochondrial metabolism and is synthesized de novo using intermediates from mitochondrial fatty-acid synthesis type II, S-adenosylmethionine and iron-sulfur clusters. This cofactor is required for catalysis by multiple mitochondrial 2-ketoacid dehydrogenase complexes, including pyruvate dehydrogenase, α-ketoglutarate dehydrogenase, and branched-chain ketoacid dehydrogenase. Lipoic acid also plays a critical role in stabilizing and regulating these multienzyme complexes. Many of these dehydrogenases are regulated by reactive oxygen species, mediated through the disulfide bond of the prosthetic lipoyl moiety. Collectively, its functions explain why lipoic acid is required for cell growth, mitochondrial activity, and coordination of fuel metabolism.


Asunto(s)
Metabolismo Energético , Metabolismo de los Lípidos , Mitocondrias/fisiología , Ácido Tióctico/metabolismo , Animales , Humanos , Oxidación-Reducción
15.
Sci Rep ; 7(1): 3455, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615704

RESUMEN

To maintain core body temperature in cold conditions, mammals activate a complex multi-organ metabolic response for heat production. White adipose tissue (WAT) primarily functions as an energy reservoir, while brown adipose tissue (BAT) is activated during cold exposure to generate heat from nutrients. Both BAT and WAT undergo specific metabolic changes during acute cold exposure. Here, we use an untargeted metabolomics approach to characterize the initial metabolic response to cold exposure in multiple adipose tissue depots in mice. Results demonstrate dramatically distinct metabolic responses during cold exposure in BAT and WAT. Amino acids, nucleotide pathways, and metabolites involved in redox regulation were greatly affected 4 hours post-exposure in BAT, while no polar metabolites were observed to significantly change in WAT depots up to 6 hours post exposure. Lipid metabolism was activated early (2 hours) in both BAT and the subcutaneous WAT depots, with the most striking change being observed in the modulation of diglyceride and monoglyceride levels in BAT. Overall, these data provide a timeline of global thermogenic metabolism in adipose depots during acute cold exposure. We have highlighted differences in visceral and subcutaneous WAT thermogenic metabolism and demonstrate the distinct metabolism of BAT during cold exposure.


Asunto(s)
Tejido Adiposo/metabolismo , Frío , Metaboloma , Metabolómica , Animales , Cromatografía Líquida de Alta Presión , Metabolismo Energético , Espectrometría de Masas , Metabolómica/métodos , Ratones , Oxidación-Reducción , Termogénesis
16.
J Physiol ; 594(24): 7455-7464, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27647490

RESUMEN

KEY POINTS: Both uncoupling protein 1 (UCP1) and UCP3 are important for mammalian thermoregulation. UCP1 and UCP3 in brown adipose tissue mediate early and late phases of sympathomimetic thermogenesis, respectively. Lipopolysaccharide thermogenesis requires skeletal muscle UCP3 but not UCP1. Acute noradrenaline-induced hyperthermia requires UCP1 but not UCP3. Loss of both UCP1 and UCP3 accelerate the loss of body temperature compared to UCP1KO alone during acute cold exposure. ABSTRACT: Uncoupling protein 1 (UCP1) is the established mediator of brown adipose tissue-dependent thermogenesis. In contrast, the role of UCP3, expressed in both skeletal muscle and brown adipose tissue, in thermoregulatory physiology is less well understood. Here, we show that mice lacking UCP3 (UCP3KO) have impaired sympathomimetic (methamphetamine) and completely abrogated lipopolysaccharide (LPS) thermogenesis, but a normal response to noradrenaline. By comparison, UCP1 knockout (UCP1KO) mice exhibit blunted methamphetamine and fully inhibited noradrenaline thermogenesis, but an increased febrile response to LPS. We further establish that mice lacking both UCP1 and 3 (UCPDK) fail to show methamphetamine-induced hyperthermia, and have a markedly accelerated loss of body temperature and survival after cold exposure compared to UCP1KO mice. Finally, we show that skeletal muscle-specific human UCP3 expression is able to significantly rescue LPS, but not sympathomimetic thermogenesis blunted in UCP3KO mice. These studies identify UCP3 as an important mediator of physiological thermogenesis and support a renewed focus on targeting UCP3 in metabolic physiology.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Proteína Desacopladora 1/fisiología , Proteína Desacopladora 3/fisiología , Animales , Frío , Hipertermia Inducida , Lipopolisacáridos/farmacología , Masculino , Metanfetamina/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Norepinefrina/farmacología , Proteína Desacopladora 1/genética , Proteína Desacopladora 3/genética
17.
Mol Cell Oncol ; 3(2): e1102795, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27308618

RESUMEN

The catabolic and energy-dissipating actions of mitochondrial uncoupling proteins (UCPs) conflict with many of the bioenergetic hallmarks of malignancy. We have recently demonstrated that overexpression of mitochondrial uncoupling protein 3 (Ucp3) in the basal epidermis impedes skin tumorigenesis through a novel pathway of thymoma viral proto-oncogene 1 (Akt1) inhibition via increased mitochondrial lipid catabolism.

18.
Nat Commun ; 6: 8137, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26310111

RESUMEN

To support growth, tumour cells reprogramme their metabolism to simultaneously upregulate macromolecular biosynthesis while maintaining energy production. Uncoupling proteins (UCPs) oppose this phenotype by inducing futile mitochondrial respiration that is uncoupled from ATP synthesis, resulting in nutrient wasting. Here using a UCP3 transgene targeted to the basal epidermis, we show that forced mitochondrial uncoupling inhibits skin carcinogenesis by blocking Akt activation. Similarly, Akt activation is markedly inhibited in UCP3 overexpressing primary human keratinocytes. Mechanistic studies reveal that uncoupling increases fatty acid oxidation and membrane phospholipid catabolism, and impairs recruitment of Akt to the plasma membrane. Overexpression of Akt overcomes metabolic regulation by UCP3, rescuing carcinogenesis. These findings demonstrate that mitochondrial uncoupling is an effective strategy to limit proliferation and tumorigenesis through inhibition of Akt, and illuminate a novel mechanism of crosstalk between mitochondrial metabolism and growth signalling.


Asunto(s)
Carcinogénesis/genética , Canales Iónicos/genética , Queratinocitos/metabolismo , Metabolismo de los Lípidos/genética , Proteínas Mitocondriales/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/genética , Animales , Carcinógenos/toxicidad , Proliferación Celular/genética , Citometría de Flujo , Ontología de Genes , Humanos , Immunoblotting , Canales Iónicos/metabolismo , Metaboloma , Metabolómica , Ratones , Ratones Transgénicos , Mitocondrias , Proteínas Mitocondriales/metabolismo , Neoplasias Experimentales , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/metabolismo , Acetato de Tetradecanoilforbol/toxicidad , Proteína Desacopladora 3
19.
Cell Cycle ; 11(14): 2681-90, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22751438

RESUMEN

Following acute hepatic injury, the metabolic capacity of the liver is altered during the process of compensatory hepatocyte proliferation by undefined mechanisms. In this study, we examined the regulation of de novo lipogenesis by cyclin D1, a key mediator of hepatocyte cell cycle progression. In primary hepatocytes, cyclin D1 significantly impaired lipogenesis in response to glucose stimulation. Cyclin D1 inhibited the glucose-mediated induction of key lipogenic genes, and similar effects were seen using a mutant (D1-KE) that does not activate cdk4 or induce cell cycle progression. Cyclin D1 (but not D1-KE) inhibited the activity of the carbohydrate response element-binding protein (ChREBP) by regulating the glucose-sensing motif of this transcription factor. Because changes in ChREBP activity could not fully explain the effect of cyclin D1, we examined hepatocyte nuclear factor 4α (HNF4α), which regulates numerous differentiated functions in the liver including lipid metabolism. We found that both cyclins D1 and D1-KE bound to HNF4α and significantly inhibited its recruitment to the promoter region of lipogenic genes in hepatocytes. Conversely, knockdown of cyclin D1 in the AML12 hepatocyte cell line promoted HNF4α activity and lipogenesis. In mouse liver, HNF4α bound to a central domain of cyclin D1 involved in transcriptional repression. Cyclin D1 inhibited lipogenic gene expression in the liver following carbohydrate feeding. Similar findings were observed in the setting of physiologic cyclin D1 expression in the regenerating liver. In conclusion, these studies demonstrate that cyclin D1 represses ChREBP and HNF4α function in hepatocytes via Cdk4-dependent and -independent mechanisms. These findings provide a direct link between the cell cycle machinery and the transcriptional control of metabolic function of the liver.


Asunto(s)
Ciclina D1/metabolismo , Glucosa/farmacología , Factor Nuclear 4 del Hepatocito/metabolismo , Lipogénesis/efectos de los fármacos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células Cultivadas , Ciclina D1/antagonistas & inhibidores , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Proteínas Nucleares/genética , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética
20.
J Biol Chem ; 286(7): 5166-74, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21163947

RESUMEN

Previous studies in our laboratory showed that isolated, intact adult rat liver mitochondria are able to oxidize the 3-carbon of serine and the N-methyl carbon of sarcosine to formate without the addition of any other cofactors or substrates. Conversion of these 1-carbon units to formate requires several folate-interconverting enzymes in mitochondria. The enzyme(s) responsible for conversion of 5,10-methylene-tetrahydrofolate (CH(2)-THF) to 10-formyl-THF in adult mammalian mitochondria are currently unknown. A new mitochondrial CH(2)-THF dehydrogenase isozyme, encoded by the MTHFD2L gene, has now been identified. The recombinant protein exhibits robust NADP(+)-dependent CH(2)-THF dehydrogenase activity when expressed in yeast. The enzyme is localized to mitochondria when expressed in CHO cells and behaves as a peripheral membrane protein, tightly associated with the matrix side of the mitochondrial inner membrane. The MTHFD2L gene is subject to alternative splicing and is expressed in adult tissues in humans and rodents. This CH(2)-THF dehydrogenase isozyme thus fills the remaining gap in the pathway from CH(2)-THF to formate in adult mammalian mitochondria.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Metilenotetrahidrofolato Deshidrogenasa (NADP)/biosíntesis , Mitocondrias/enzimología , Proteínas Mitocondriales/biosíntesis , Empalme Alternativo/fisiología , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Especificidad de Órganos/fisiología , Ratas , Ratas Sprague-Dawley , Tetrahidrofolatos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA