Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 158(1): 69-83, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995979

RESUMEN

Brown fat can reduce obesity through the dissipation of calories as heat. Control of thermogenic gene expression occurs via the induction of various coactivators, most notably PGC-1α. In contrast, the transcription factor partner(s) of these cofactors are poorly described. Here, we identify interferon regulatory factor 4 (IRF4) as a dominant transcriptional effector of thermogenesis. IRF4 is induced by cold and cAMP in adipocytes and is sufficient to promote increased thermogenic gene expression, energy expenditure, and cold tolerance. Conversely, knockout of IRF4 in UCP1(+) cells causes reduced thermogenic gene expression and energy expenditure, obesity, and cold intolerance. IRF4 also induces the expression of PGC-1α and PRDM16 and interacts with PGC-1α, driving Ucp1 expression. Finally, cold, ß-agonists, or forced expression of PGC-1α are unable to cause thermogenic gene expression in the absence of IRF4. These studies establish IRF4 as a transcriptional driver of a program of thermogenic gene expression and energy expenditure.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Factores Reguladores del Interferón/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Activación Transcripcional , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Frío , AMP Cíclico/metabolismo , Metabolismo Energético , Humanos , Canales Iónicos/genética , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Delgadez/metabolismo , Activación Transcripcional/efectos de los fármacos , Proteína Desacopladora 1
2.
Cell ; 152(3): 612-9, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374353

RESUMEN

Melanocortin 4 receptors (MC4Rs) in the central nervous system are key regulators of energy and glucose homeostasis. Notably, obese patients with MC4R mutations are hyperinsulinemic and resistant to obesity-induced hypertension. Although these effects are probably dependent upon the activity of the autonomic nervous system, the cellular effects of MC4Rs on parasympathetic and sympathetic neurons remain undefined. Here, we show that MC4R agonists inhibit parasympathetic preganglionic neurons in the brainstem. In contrast, MC4R agonists activate sympathetic preganglionic neurons in the spinal cord. Deletion of MC4Rs in cholinergic neurons resulted in elevated levels of insulin. Furthermore, re-expression of MC4Rs specifically in cholinergic neurons (including sympathetic preganglionic neurons) restores obesity-associated hypertension in MC4R null mice. These findings provide a cellular correlate of the autonomic side effects associated with MC4R agonists and demonstrate a role for MC4Rs expressed in cholinergic neurons in the regulation of insulin levels and in the development of obesity-induced hypertension.


Asunto(s)
Tronco Encefálico/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Presión Sanguínea , Tronco Encefálico/citología , Neuronas Colinérgicas/metabolismo , AMP Cíclico/metabolismo , Fenómenos Electrofisiológicos , Humanos , Canales KATP/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Obesidad/fisiopatología , Sistema Nervioso Parasimpático/metabolismo , Receptor de Melanocortina Tipo 4/genética , Médula Espinal/metabolismo , Sistema Nervioso Simpático/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(13): e2213857120, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36947517

RESUMEN

Poly(ADP-ribose) polymerase-1 (PARP1) has been reported to play an important role in longevity. Here, we showed that the knockdown of the PARP1 extended the lifespan of Drosophila, with particular emphasis on the skeletal muscle. The muscle-specific mutant Drosophila exhibited resistance to starvation and oxidative stress, as well as an increased ability to climb, with enhanced mitochondrial biogenesis and activity at an older age. Mechanistically, the inhibition of PARP1 increases the activity of AMP-activated protein kinase alpha (AMPKα) and mitochondrial turnover. PARP1 could interact with AMPKα and then regulate it via poly(ADP ribosyl)ation (PARylation) at residues E155 and E195. Double knockdown of PARP1 and AMPKα, specifically in muscle, could counteract the effects of PARP1 inhibition in Drosophila. Finally, we showed that increasing lifespan via maintaining mitochondrial network homeostasis required intact PTEN induced kinase 1 (PINK1). Taken together, these data indicate that the interplay between PARP1 and AMPKα can manipulate mitochondrial turnover, and be targeted to promote longevity.


Asunto(s)
Proteínas de Drosophila , Poli(ADP-Ribosa) Polimerasa-1 , Poli ADP Ribosilación , Animales , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Longevidad/genética , Músculos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
4.
J Biol Chem ; 300(2): 105601, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38159860

RESUMEN

Hepatocyte plays a principal role in preserving integrity of the liver homeostasis. Our recent study demonstrated that Kindlin-2, a focal adhesion protein that activates integrins and regulates cell-extracellular matrix interactions, plays an important role in regulation of liver homeostasis by inhibiting inflammation pathway; however, the molecular mechanism of how Kindlin-2 KO activates inflammation is unknown. Here, we show that Kindlin-2 loss largely downregulates the antioxidant glutathione-S-transferase P1 in hepatocytes by promoting its ubiquitination and degradation via a mechanism involving protein-protein interaction. This causes overproduction of intracellular reactive oxygen species and excessive oxidative stress in hepatocytes. Kindlin-2 loss upregulates osteopontin in hepatocytes partially because of upregulation of reactive oxygen species and consequently stimulates overproduction of inflammatory cytokines and infiltration in liver. The molecular and histological deteriorations caused by Kindlin-2 deficiency are markedly reversed by systemic administration of an antioxidant N-acetylcysteine in mice. Taken together, Kindlin-2 plays a pivotal role in preserving integrity of liver function.


Asunto(s)
Proteínas del Citoesqueleto , Inflamación , Proteínas de la Membrana , Estrés Oxidativo , Animales , Ratones , Antioxidantes/metabolismo , Homeostasis , Inflamación/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas del Citoesqueleto/metabolismo
5.
Mol Psychiatry ; 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39122778

RESUMEN

Astrocytes are multi-functional glial cells in the central nervous system that play critical roles in modulation of metabolism, extracellular ion and neurotransmitter levels, and synaptic plasticity. Astrocyte-derived signaling molecules mediate many of these modulatory functions of astrocytes, including vesicular release of ATP. In the present study, we used a unique genetic mouse model to investigate the functional significance of astrocytic exocytosis of ATP. Using primary cultured astrocytes, we show that loss of vesicular nucleotide transporter (Vnut), a primary transporter responsible for loading cytosolic ATP into the secretory vesicles, dramatically reduces ATP loading into secretory lysosomes and ATP release, without any change in the molecular machinery of exocytosis or total intracellular ATP content. Deletion of astrocytic Vnut in adult mice leads to increased anxiety, depressive-like behaviors, and decreased motivation for reward, especially in females, without significant impact on food intake, systemic glucose metabolism, cognition, or sociability. These behavioral alterations are associated with significant decreases in the basal extracellular dopamine levels in the nucleus accumbens. Likewise, ex vivo brain slices from these mice show a strong trend toward a reduction in evoked dopamine release in the nucleus accumbens. Mechanistically, the reduced dopamine signaling we observed is likely due to an increased expression of monoamine oxidases. Together, these data demonstrate a key modulatory role of astrocytic exocytosis of ATP in anxiety, depressive-like behavior, and motivation for reward, by regulating the mesolimbic dopamine circuitry.

6.
Liver Int ; 44(9): 2396-2408, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38847599

RESUMEN

BACKGROUND AND AIMS: Metabolic dysfunction-associated steatotic liver disease (MASLD) represents the foremost cause of chronic liver disease, yet its underlying mechanisms remain elusive. Our group previously discovered a novel long non-coding RNA (lncRNA) in rats, termed lncHC and its human counterpart, LNCHC. This study aimed to explore the role of LNCHC in the progression of MASLD. METHODS: RNA-binding proteins bound to LNCHC were searched by mass spectrometry. The target genes of LNCHC and Y-Box binding protein 1 (YBX1) were identified by RNA-seq. MASLD animal models were utilised to examine the roles of LNCHC, YBX1 and patatin-like phospholipase domain containing 3 (PNPLA3) in MASLD progression. RESULTS: Here, we identified LNCHC as a native restrainer during MASLD development. Notably, LNCHC directly binds YBX1 and prevents protein ubiquitination. Up-regulation of YBX1 then stabilises PNPLA3 mRNA to alleviate lipid accumulation in hepatocytes. Furthermore, both cell and animal studies demonstrate that LNCHC, YBX1 and PNPLA3 function to improve hepatocyte lipid accumulation and exacerbate metabolic dysfunction-associated steatohepatitis development. CONCLUSIONS: In summary, our findings unveil a novel LNCHC functionality in regulating YBX1 and PNPLA3 mRNA stability during MASLD development, providing new avenues in MASLD treatment.


Asunto(s)
Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteína 1 de Unión a la Caja Y , Proteína 1 de Unión a la Caja Y/metabolismo , Proteína 1 de Unión a la Caja Y/genética , Animales , Humanos , Ratas , Masculino , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Hepatocitos/metabolismo , Hígado Graso/metabolismo , Ubiquitinación , Aciltransferasas , Fosfolipasas A2 Calcio-Independiente
7.
J Cell Physiol ; 237(11): 4169-4179, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35998296

RESUMEN

Poly(ADP-ribose) polymerase 1 (PARP1) plays a key role in genome stability by modulating DNA-damage responses. Activated by DNA interruptions through ultraviolet (UV) exposure, PARylation is synthesized by PARP1 and serves as a survival mechanism for cancer and metabolic diseases. Several strategies including ROS and antimicrobial peptides (AMPs) function in host defenses, while the targeted tissue and mechanism under DNA damage are unknown. Here, we show that DNA damage induces responses specifically in the gut tissue. The knockdown of PARP1 reduces the activation of PARylation. Parp1 knockdown under DNA damage results in over-accumulated ROS and secretion of AMPs through the regulation of Relish, a subunit of nuclear factor-κB (NF-κB). Double-knockdown of Parp1 and Relish specifically in the gut inhibits AMP secretion. In conclusion, the host defense is achieved through ROS accumulation rather than the AMPs under DNA damage. In contrast, the knockdown of PARP1 exacerbates ROS accumulation to a harmful level. Under this circumstance, NF-κb targeted AMP secretion is provoked for host defense. Microbiome and functional analysis provide evidence for the hazard of DNA damage and show variations in the metabolic pathways following Parp1 inhibition. Our findings suggest the notion that PARP1 inhibition contributes to ROS accumulation under DNA damage and its role in NF-κb activation for host defense.


Asunto(s)
Microbioma Gastrointestinal , FN-kappa B , ADN/metabolismo , Daño del ADN , FN-kappa B/genética , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Especies Reactivas de Oxígeno
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt A): 2477-2485, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28499988

RESUMEN

The burden of disability, premature death, escalating health care costs and lost economic productivity due to obesity and its associated complications including hypertension, stroke, cardiovascular disease and type 2 diabetes is staggering [1,2]. A better understanding of metabolic homeostatic pathways will provide us with insights into the biological mechanisms of obesity and how to fundamentally address this epidemic [3-6]. In mammals, energy balance is maintained via a homeostatic system involving both peripheral and central melanocortin systems; changes in body weight reflect an unbalance of the energetic state [7-9]. Although the primary cause of obesity is unknown, there is significant effort to understand the role of the central melanocortin pathway in the brain as it has been shown that deficiency of proopiomelanocortin (POMC) [10,11] and melanocortin 4 receptors (MC4R) [12-15] in both rodents and humans results in severe hyperphagia and obesity [16-23]. In this review, we will summarize how the central melanocortin pathway helps regulate body mass and adiposity within a 'healthy' range through the 'nutrient sensing' network [24-28]. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.


Asunto(s)
Melanocortinas/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Metabolismo Energético , Humanos , Hiperfagia/genética , Hiperfagia/metabolismo , Mutación , Obesidad/genética , Obesidad/metabolismo , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal
10.
Int J Clin Oncol ; 22(6): 1076-1080, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28664301

RESUMEN

BACKGROUND: Renal cell carcinoma (RCC) is the third most common genitourinary cancer. Activated platelets play a pivotal role in cancer development and progression. Altered mean platelet volume (MPV) has been reported in several malignancies. The aim of the present study was to investigate the association of MPV with RCC. STUDY DESIGN: The study consisted of 145 patients with RCC, 110 patients with benign renal tumor and 132 healthy control subjects between January 2015 and December 2015. All participants' clinical and laboratory characteristics at initial diagnosis were collected. The odds ratios (ORs) for RCC were calculated using multivariate logistic regression analysis after adjusting for confounding variables across MPV quartiles. RESULTS: The patients with RCC had decreased pre-operative MPV compared to the patients with benign renal tumor and healthy control subjects. Furthermore, pre-operative MPV was reduced in benign renal tumor compared with healthy control subjects. Surgical tumor resection resulted in a significant increase in MPV levels (8.7 fL vs. 9.0 fL; p = 0.011). After adjusting for other risk factors, the ORs (95% CIs) for RCC in each MPV quartile were 25.725 (7.556-87.585), 7.447 (2.701-20.537), 0.703 (0.245-2.019), and 1.000, respectively. CONCLUSIONS: RCC patients have remarkably reduced MPV compared to patients with benign renal tumor and healthy control subjects. Moreover, decreased MPV was independently associated with RCC. Our results suggest that detection of MPV may be useful to assess the risk of RCC.


Asunto(s)
Carcinoma de Células Renales/sangre , Neoplasias Renales/sangre , Volúmen Plaquetario Medio , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Renales/patología , Estudios de Casos y Controles , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Factores de Riesgo
11.
Med Sci Monit ; 21: 3847-3852, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26649647

RESUMEN

BACKGROUND Gallstones are associated with dyslipidemia, metabolic syndrome, hyperinsulinemia, and carotid intima-media thickness, which all are related to hyperviscosity. Therefore, this study aimed to examine the associations between blood viscosity and gallstones. MATERIAL AND METHODS We investigated the associations between blood viscosity and gallstones in a study conducted in middle-aged subjects. RESULTS Among 849 enrolled patients, 421 (49.6%) had gallstone disease. Compared with control subjects, whole-blood viscosity (WBV) levels were increased in patients with gallstones. The prevalence of gallstones increased as WBV (3 s-1) quartiles increased. Furthermore, logistic regression analysis showed that WBV (3 s-1) was associated with gallstone disease. CONCLUSIONS We found that whole-blood viscosity at low shear rate was independently associated with gallstones. Whether control of hyperviscosity would reduce the risk of developing gallstones deserves further investigation.

12.
Am J Physiol Endocrinol Metab ; 306(3): E332-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24326423

RESUMEN

Rho kinase (ROCK) isoforms regulate insulin signaling and glucose metabolism negatively or positively in cultured cell lines and skeletal muscle. However, the in vivo function of the ROCK1 isoform in adipose tissue has not been addressed. To determine the specific role of the adipose ROCK1 isoform in the development of insulin resistance and obesity, mice lacking ROCK1 in adipose tissue globally or selectively were studied. Here, we show that insulin's ability to activate IRS-1/PI3K/Akt signaling was greatly enhanced in adipose tissue of ROCK1(-/-) mice compared with wild-type mice. These effects resulted from the inhibitory effect of ROCK1 on insulin receptor action, as evidenced by the fact that IR tyrosine phosphorylation was abolished in ROCK1(-/-) MEF cells when ROCK1 was reexpressed. Consistently, adipose-specific disruption of ROCK1 increased IR tyrosine phosphorylation in adipose tissue and modestly improved sensitivity to insulin in obese mice induced by high-fat feeding. This effect is independent of any changes in adiposity, number or size of adipocytes, and metabolic parameters, including glucose, insulin, leptin, and triglyceride levels, demonstrating a minimal effect of adipose ROCK1 on whole body metabolism. Enzymatic activity of ROCK1 in adipose tissue remained ∼50%, which likely originated from the fraction of stromal vascular cells, suggesting involvement of these cells for adipose metabolic regulation. Moreover, ROCK isoform activities were increased in adipose tissue of diet-induced or genetically obese mice. These data suggest that adipose ROCK1 isoform plays an inhibtory role for the regulation of insulin sensitivity in diet-induced obesity in vivo.


Asunto(s)
Dieta/efectos adversos , Eliminación de Gen , Resistencia a la Insulina/genética , Quinasas Asociadas a rho/genética , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Femenino , Isoenzimas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Especificidad de Órganos/genética
13.
Metabolites ; 14(3)2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38535295

RESUMEN

Adipose tissue undergoes changes with aging, leading to increased adiposity, inflammatory cell infiltration, reduced angiogenesis, heightened oxidative stress, and alterations in its metabolic function. Regular exercise has been recognized as a powerful intervention that can positively influence adipose tissue health and mitigate the effects of aging. However, the molecular mechanisms underlying the benefits of regular exercise on aging adipose tissue function remain poorly understood. Adipokines released through regular exercise play a potential role in mitigating adipose tissue aging, enhancing the metabolism of glucose and lipids, reducing inflammation and fibrosis, and promoting fat browning and thermogenesis. This review comprehensively summarizes the benefits of regular exercise in addressing the age-related decline in adipose tissue function. Utilizing relevant examples of this approach, we address the possibility of designing therapeutic interventions based on these molecular mechanisms.

14.
Phenomics ; 4(2): 203-212, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38884055

RESUMEN

Energy plays a vital role in biological processes. To assess energy metabolism status in a large population cohort, the standard operating procedure for measuring energy expenditure measurement using a whole-room calorimeter was purposed in this study. This protocol illustrates the procedure and specific details for validating methanol burning and evaluating the metabolic status of volunteers. In metabolic status evaluation, the (1) O2 consumption, (2) CO2 production, (3) energy expenditure, and (4) respiratory exchange ratio were first measured at resting and provided as basic phenotype items in Human Phenotype Atlas. Besides, it includes the procedure and results for measuring exercise-related activity thermogenesis and evaluating the impact of environmental temperature on energy metabolism. These results demonstrate the broader utility of the whole-room calorimeter. The implementation of this protocol is expected to enhance the data comparability in Human Phenotype Atla and provide a valuable reference for metabolism-related studies.

15.
Sci Rep ; 14(1): 3175, 2024 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326642

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) has emerged as one of the major causes of liver-related morbidity and mortality globally. It ranges from simple steatosis to non-alcoholic steatohepatitis (NASH) characterized by ballooning and hepatic inflammation. In the past few years, pyroptosis has been shown as a type of programmed cell death that triggers inflammation and plays a role in the development of NASH. However, the roles of pyroptosis-related genes (PRGs) in NASH remained unclear. In this study, we studied the expression level of pyroptosis-related genes (PRGs) in NASH and healthy controls, developed a diagnostic model of NASH based on PRGs and explored the pathological mechanisms associated with pyroptosis. We further compared immune status between NASH and healthy controls, analyzed immune status in different subtypes of NASH. We identified altogether twenty PRGs that were differentially expressed between NASH and normal liver tissues. Then, a novel diagnostic model consisting of seven PRGs including CASP3, ELANE, GZMA, CASP4, CASP9, IL6 and TP63 for NASH was constructed with an area under the ROC curve (AUC) of 0.978 (CI 0.965-0.99). Obvious variations in immune status between healthy controls and NASH cases were detected. Subsequently, the consensus clustering method based on differentially expressed PRGs was constructed to divide all NASH cases into two distinct pyroptosis subtypes with different immune and biological characteristics. Pyroptosis-related genes may play an important role in NASH and can provide new insights into the diagnosis and underlying mechanisms of NASH.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Piroptosis/genética , Inflamación/patología
16.
Nat Protoc ; 19(5): 1311-1347, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38307980

RESUMEN

As a key glycolytic metabolite, lactate has a central role in diverse physiological and pathological processes. However, comprehensive multiscale analysis of lactate metabolic dynamics in vitro and in vivo has remained an unsolved problem until now owing to the lack of a high-performance tool. We recently developed a series of genetically encoded fluorescent sensors for lactate, named FiLa, which illuminate lactate metabolism in cells, subcellular organelles, animals, and human serum and urine. In this protocol, we first describe the FiLa sensor-based strategies for real-time subcellular bioenergetic flux analysis by profiling the lactate metabolic response to different nutritional and pharmacological conditions, which provides a systematic-level view of cellular metabolic function at the subcellular scale for the first time. We also report detailed procedures for imaging lactate dynamics in live mice through a cell microcapsule system or recombinant adeno-associated virus and for the rapid and simple assay of lactate in human body fluids. This comprehensive multiscale metabolic analysis strategy may also be applied to other metabolite biosensors using various analytic platforms, further expanding its usability. The protocol is suited for users with expertise in biochemistry, molecular biology and cell biology. Typically, the preparation of FiLa-expressing cells or mice takes 2 days to 4 weeks, and live-cell and in vivo imaging can be performed within 1-2 hours. For the FiLa-based assay of body fluids, the whole measuring procedure generally takes ~1 min for one sample in a manual assay or ~3 min for 96 samples in an automatic microplate assay.


Asunto(s)
Técnicas Biosensibles , Ácido Láctico , Animales , Humanos , Ratones , Técnicas Biosensibles/métodos , Ácido Láctico/metabolismo , Ácido Láctico/análisis
17.
J Clin Invest ; 134(16)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38889010

RESUMEN

Myostatin (MSTN) has long been recognized as a critical regulator of muscle mass. Recently, there has been increasing interest in its role in metabolism. In our study, we specifically knocked out MSTN in brown adipose tissue (BAT) from mice (MSTNΔUCP1) and found that the mice gained more weight than did controls when fed a high-fat diet, with progressive hepatosteatosis and impaired skeletal muscle activity. RNA-Seq analysis indicated signatures of mitochondrial dysfunction and inflammation in the MSTN-ablated BAT. Further studies demonstrated that Kruppel-like factor 4 (KLF4) was responsible for the metabolic phenotypes observed, whereas fibroblast growth factor 21 (FGF21) contributed to the microenvironment communication between adipocytes and macrophages induced by the loss of MSTN. Moreover, the MSTN/SMAD2/3-p38 signaling pathway mediated the expression of KLF4 and FGF21 in adipocytes. In summary, our findings suggest that brown adipocyte-derived MSTN regulated BAT thermogenesis via autocrine and paracrine effects on adipocytes or macrophages, ultimately regulating systemic energy homeostasis.


Asunto(s)
Comunicación Autocrina , Factores de Crecimiento de Fibroblastos , Homeostasis , Factor 4 Similar a Kruppel , Macrófagos , Ratones Noqueados , Miostatina , Comunicación Paracrina , Termogénesis , Animales , Masculino , Ratones , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Microambiente Celular , Metabolismo Energético , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factor 4 Similar a Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Macrófagos/metabolismo , Miostatina/metabolismo , Miostatina/genética
18.
Nat Commun ; 15(1): 7603, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39217143

RESUMEN

Resistance to stress is a key determinant for mammalian functioning. While many studies have revealed neural circuits and substrates responsible for initiating and mediating stress responses, little is known about how the brain resists to stress and prevents overreactions. Here, we identified a previously uncharacterized neuropeptide Y (NPY) neuronal population in the dorsal raphe nucleus and ventrolateral periaqueductal gray region (DRN/vlPAG) with anxiolytic effects in male mice. NPYDRN/vlPAG neurons are rapidly activated by various stressful stimuli. Inhibiting these neurons exacerbated hypophagic and anxiety responses during stress, while activation significantly ameliorates acute stress-induced hypophagia and anxiety levels and transmits positive valence. Furthermore, NPYDRN/vlPAG neurons exert differential but synergic anxiolytic effects via inhibitory projections to the paraventricular thalamic nucleus (PVT) and the lateral hypothalamic area (LH). Together, our findings reveal a feedforward inhibition neural mechanism underlying stress resistance and suggest NPYDRN/vlPAG neurons as a potential therapeutic target for stress-related disorders.


Asunto(s)
Neuronas , Neuropéptido Y , Estrés Psicológico , Animales , Masculino , Neuropéptido Y/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Ratones , Estrés Psicológico/fisiopatología , Ratones Endogámicos C57BL , Ansiedad/fisiopatología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Sustancia Gris Periacueductal/fisiología , Tronco Encefálico/fisiología , Área Hipotalámica Lateral/fisiología , Estrés Fisiológico
19.
Cell Metab ; 36(2): 438-453.e6, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38325338

RESUMEN

The hypothalamus plays a crucial role in the progression of obesity and diabetes; however, its structural complexity and cellular heterogeneity impede targeted treatments. Here, we profiled the single-cell and spatial transcriptome of the hypothalamus in obese and sporadic type 2 diabetic macaques, revealing primate-specific distributions of clusters and genes as well as spatial region, cell-type-, and gene-feature-specific changes. The infundibular (INF) and paraventricular nuclei (PVN) are most susceptible to metabolic disruption, with the PVN being more sensitive to diabetes. In the INF, obesity results in reduced synaptic plasticity and energy sensing capability, whereas diabetes involves molecular reprogramming associated with impaired tanycytic barriers, activated microglia, and neuronal inflammatory response. In the PVN, cellular metabolism and neural activity are suppressed in diabetic macaques. Spatial transcriptomic data reveal microglia's preference for the parenchyma over the third ventricle in diabetes. Our findings provide a comprehensive view of molecular changes associated with obesity and diabetes.


Asunto(s)
Diabetes Mellitus , Núcleo Hipotalámico Paraventricular , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Transcriptoma/genética , Hipotálamo/metabolismo , Obesidad/metabolismo , Diabetes Mellitus/metabolismo , Perfilación de la Expresión Génica
20.
Cell Metab ; 36(5): 1000-1012.e6, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38582087

RESUMEN

The gut-brain axis is implicated in depression development, yet its underlying mechanism remains unclear. We observed depleted gut bacterial species, including Bifidobacterium longum and Roseburia intestinalis, and the neurotransmitter homovanillic acid (HVA) in individuals with depression and mouse depression models. Although R. intestinalis does not directly produce HVA, it enhances B. longum abundance, leading to HVA generation. This highlights a synergistic interaction among gut microbiota in regulating intestinal neurotransmitter production. Administering HVA, B. longum, or R. intestinalis to mouse models with chronic unpredictable mild stress (CUMS) and corticosterone (CORT)-induced depression significantly improved depressive symptoms. Mechanistically, HVA inhibited synaptic autophagic death by preventing excessive degradation of microtubule-associated protein 1 light chain 3 (LC3) and SQSTM1/p62 proteins, protecting hippocampal neurons' presynaptic membrane. These findings underscore the role of the gut microbial metabolism in modulating synaptic integrity and provide insights into potential novel treatment strategies for depression.


Asunto(s)
Depresión , Microbioma Gastrointestinal , Ácido Homovanílico , Ratones Endogámicos C57BL , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Depresión/tratamiento farmacológico , Depresión/metabolismo , Masculino , Humanos , Ácido Homovanílico/metabolismo , Sinapsis/metabolismo , Sinapsis/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA