Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33.246
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 25(6): 981-993, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38811816

RESUMEN

Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic ß cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.


Asunto(s)
Glucemia , Inmunidad Innata , Interferón gamma , Animales , Interferón gamma/metabolismo , Interferón gamma/inmunología , Ratones , Glucemia/metabolismo , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Insulina/metabolismo , Insulina/inmunología , Ratones Noqueados , Hiperglucemia/inmunología , Factor 3 Regulador del Interferón/metabolismo , FN-kappa B/metabolismo , Humanos , Hígado/inmunología , Hígado/virología , Hígado/metabolismo , Masculino
2.
Cell ; 182(2): 372-387.e14, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32610084

RESUMEN

Acute psychological stress has long been known to decrease host fitness to inflammation in a wide variety of diseases, but how this occurs is incompletely understood. Using mouse models, we show that interleukin-6 (IL-6) is the dominant cytokine inducible upon acute stress alone. Stress-inducible IL-6 is produced from brown adipocytes in a beta-3-adrenergic-receptor-dependent fashion. During stress, endocrine IL-6 is the required instructive signal for mediating hyperglycemia through hepatic gluconeogenesis, which is necessary for anticipating and fueling "fight or flight" responses. This adaptation comes at the cost of enhancing mortality to a subsequent inflammatory challenge. These findings provide a mechanistic understanding of the ontogeny and adaptive purpose of IL-6 as a bona fide stress hormone coordinating systemic immunometabolic reprogramming. This brain-brown fat-liver axis might provide new insights into brown adipose tissue as a stress-responsive endocrine organ and mechanistic insight into targeting this axis in the treatment of inflammatory and neuropsychiatric diseases.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Interleucina-6/metabolismo , Estrés Psicológico , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Encéfalo/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Gluconeogénesis , Hiperglucemia/metabolismo , Hiperglucemia/patología , Interleucina-6/sangre , Interleucina-6/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Adrenérgicos beta 3/metabolismo , Receptores de Interleucina-6/metabolismo , Proteína Desacopladora 1/deficiencia , Proteína Desacopladora 1/genética
3.
Cell ; 165(2): 264-5, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058657

RESUMEN

In this issue of Cell, Okin and Medzhitov report that sustained inflammation promotes hyperglycemia by targeting the mevalonate pathway. This represents an important step forward in understanding the mechanisms underlying the association between chronic low-grade inflammation and disruption of normal tissue functions in metabolic diseases.


Asunto(s)
Hiperglucemia/metabolismo , Insulina , Humanos , Inflamación/metabolismo , Enfermedades Metabólicas , Transducción de Señal
4.
Cell ; 165(2): 343-56, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26997483

RESUMEN

Control of plasma glucose level is essential to organismal survival. Sustained inflammation has been implicated in control of glucose homeostasis in cases of infection, obesity, and type 2 diabetes; however, the precise role of inflammation in these complex disease states remains poorly understood. Here, we find that sustained inflammation results in elevated plasma glucose due to increased hepatic glucose production. We find that sustained inflammation suppresses CYP7A1, leading to accumulation of intermediate metabolites at the branch point of the mevalonate pathway. This results in prenylation of RHOC, which is concomitantly induced by inflammatory cytokines. Subsequent activation of RHO-associated protein kinase results in elevated plasma glucose. These findings uncover an unexpected mechanism by which sustained inflammation alters glucose homeostasis.


Asunto(s)
Vías Biosintéticas , Hepatitis/metabolismo , Hiperglucemia/metabolismo , Ácido Mevalónico/metabolismo , Animales , Glucemia/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Colesterol 7-alfa-Hidroxilasa/metabolismo , Ayuno/sangre , Lipopolisacáridos , Ratones , Ratones Obesos , Prenilación de Proteína , Transcripción Genética , Triglicéridos/sangre , Proteínas ras/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína rhoC de Unión a GTP
5.
Cell ; 167(3): 843-857.e14, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27720451

RESUMEN

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.


Asunto(s)
Glucagón/uso terapéutico , Enfermedades Metabólicas/tratamiento farmacológico , Triyodotironina/efectos de los fármacos , Animales , Aterosclerosis/tratamiento farmacológico , Peso Corporal/efectos de los fármacos , Huesos/efectos de los fármacos , Ingeniería Química/métodos , Colesterol/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Combinación de Medicamentos , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Glucagón/efectos adversos , Glucagón/química , Glucagón/farmacología , Hiperglucemia/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Terapia Molecular Dirigida , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Triyodotironina/efectos adversos , Triyodotironina/química , Triyodotironina/farmacología
6.
Physiol Rev ; 103(1): 7-30, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35635320

RESUMEN

In this paper, we provide an overview of the evolution of the definition of hyperglycemia during the past century and the alterations in glucose dynamics that cause fasting and postprandial hyperglycemia. We discuss how extensive mechanistic, physiological research into the factors and pathways that regulate the appearance of glucose in the circulation and its uptake and metabolism by tissues and organs has contributed knowledge that has advanced our understanding of different types of hyperglycemia, namely prediabetes and diabetes and their subtypes (impaired fasting plasma glucose, impaired glucose tolerance, combined impaired fasting plasma glucose, impaired glucose tolerance, type 1 diabetes, type 2 diabetes, gestational diabetes mellitus), their relationships with medical complications, and how to prevent and treat hyperglycemia.


Asunto(s)
Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Hiperglucemia , Estado Prediabético , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Glucosa , Intolerancia a la Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Estado Prediabético/diagnóstico , Embarazo , Azúcares
7.
Cell ; 163(6): 1457-67, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26627735

RESUMEN

A variety of signals finely tune insulin secretion by pancreatic ß cells to prevent both hyper-and hypoglycemic states. Here, we show that post-translational regulation of the transcription factors ETV1, ETV4, and ETV5 by the ubiquitin ligase COP1 (also called RFWD2) in ß cells is critical for insulin secretion. Mice lacking COP1 in ß cells developed diabetes due to insulin granule docking defects that were fully rescued by genetic deletion of Etv1, Etv4, and Etv5. Genes regulated by ETV1, ETV4, or ETV5 in the absence of mouse COP1 were enriched in human diabetes-associated genes, suggesting that they also influence human ß-cell pathophysiology. In normal ß cells, ETV4 was stabilized upon membrane depolarization and limited insulin secretion under hyperglycemic conditions. Collectively, our data reveal that ETVs negatively regulate insulin secretion for the maintenance of normoglycemia.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus/metabolismo , Exocitosis , Eliminación de Gen , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Secreción de Insulina , Ratones , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética
8.
Cell ; 160(4): 745-758, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25662011

RESUMEN

Impaired insulin-mediated suppression of hepatic glucose production (HGP) plays a major role in the pathogenesis of type 2 diabetes (T2D), yet the molecular mechanism by which this occurs remains unknown. Using a novel in vivo metabolomics approach, we show that the major mechanism by which insulin suppresses HGP is through reductions in hepatic acetyl CoA by suppression of lipolysis in white adipose tissue (WAT) leading to reductions in pyruvate carboxylase flux. This mechanism was confirmed in mice and rats with genetic ablation of insulin signaling and mice lacking adipose triglyceride lipase. Insulin's ability to suppress hepatic acetyl CoA, PC activity, and lipolysis was lost in high-fat-fed rats, a phenomenon reversible by IL-6 neutralization and inducible by IL-6 infusion. Taken together, these data identify WAT-derived hepatic acetyl CoA as the main regulator of HGP by insulin and link it to inflammation-induced hepatic insulin resistance associated with obesity and T2D.


Asunto(s)
Acetilcoenzima A/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Paniculitis/metabolismo , Tejido Adiposo Blanco/química , Adolescente , Animales , Diabetes Mellitus Tipo 2 , Dieta Alta en Grasa , Glucosa/metabolismo , Humanos , Hiperglucemia , Interleucina-6/análisis , Lipólisis , Masculino , Ratones , Obesidad/metabolismo , Ratas Sprague-Dawley
9.
Nature ; 629(8014): 1133-1141, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38750368

RESUMEN

The N-methyl-D-aspartate (NMDA) receptor is a glutamate-activated cation channel that is critical to many processes in the brain. Genome-wide association studies suggest that glutamatergic neurotransmission and NMDA receptor-mediated synaptic plasticity are important for body weight homeostasis1. Here we report the engineering and preclinical development of a bimodal molecule that integrates NMDA receptor antagonism with glucagon-like peptide-1 (GLP-1) receptor agonism to effectively reverse obesity, hyperglycaemia and dyslipidaemia in rodent models of metabolic disease. GLP-1-directed delivery of the NMDA receptor antagonist MK-801 affects neuroplasticity in the hypothalamus and brainstem. Importantly, targeting of MK-801 to GLP-1 receptor-expressing brain regions circumvents adverse physiological and behavioural effects associated with MK-801 monotherapy. In summary, our approach demonstrates the feasibility of using peptide-mediated targeting to achieve cell-specific ionotropic receptor modulation and highlights the therapeutic potential of unimolecular mixed GLP-1 receptor agonism and NMDA receptor antagonism for safe and effective obesity treatment.


Asunto(s)
Maleato de Dizocilpina , Péptido 1 Similar al Glucagón , Receptor del Péptido 1 Similar al Glucagón , Obesidad , Receptores de N-Metil-D-Aspartato , Animales , Humanos , Masculino , Ratones , Ratas , Tronco Encefálico/metabolismo , Tronco Encefálico/efectos de los fármacos , Modelos Animales de Enfermedad , Maleato de Dizocilpina/efectos adversos , Maleato de Dizocilpina/farmacología , Maleato de Dizocilpina/uso terapéutico , Dislipidemias/tratamiento farmacológico , Dislipidemias/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ratas Sprague-Dawley , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
10.
Cell ; 156(3): 396-7, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485448

RESUMEN

Vogt et al. demonstrate that, in mice, maternal high-fat feeding during lactation is sufficient to program the offspring for impaired energy and glucose homeostasis throughout their lifetime. They reveal that the resulting abnormal insulin signaling in the offspring interferes with the formation of hypothalamic neural circuits that contribute to metabolic status.


Asunto(s)
Dieta Alta en Grasa , Hiperglucemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Lactancia , Obesidad/metabolismo , Animales , Femenino , Masculino , Embarazo
11.
Cell ; 156(3): 495-509, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24462248

RESUMEN

Maternal metabolic homeostasis exerts long-term effects on the offspring's health outcomes. Here, we demonstrate that maternal high-fat diet (HFD) feeding during lactation predisposes the offspring for obesity and impaired glucose homeostasis in mice, which is associated with an impairment of the hypothalamic melanocortin circuitry. Whereas the number and neuropeptide expression of anorexigenic proopiomelanocortin (POMC) and orexigenic agouti-related peptide (AgRP) neurons, electrophysiological properties of POMC neurons, and posttranslational processing of POMC remain unaffected in response to maternal HFD feeding during lactation, the formation of POMC and AgRP projections to hypothalamic target sites is severely impaired. Abrogating insulin action in POMC neurons of the offspring prevents altered POMC projections to the preautonomic paraventricular nucleus of the hypothalamus (PVH), pancreatic parasympathetic innervation, and impaired glucose-stimulated insulin secretion in response to maternal overnutrition. These experiments reveal a critical timing, when altered maternal metabolism disrupts metabolic homeostasis in the offspring via impairing neuronal projections, and show that abnormal insulin signaling contributes to this effect.


Asunto(s)
Dieta Alta en Grasa , Hiperglucemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Lactancia , Obesidad/metabolismo , Animales , Axones/metabolismo , Femenino , Masculino , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Embarazo , Proopiomelanocortina/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal
12.
Nature ; 624(7992): 645-652, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38093014

RESUMEN

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


Asunto(s)
Células Dendríticas , Complicaciones de la Diabetes , Diabetes Mellitus , Susceptibilidad a Enfermedades , Hiperglucemia , Pulmón , Virosis , Animales , Ratones , Acetilcoenzima A/metabolismo , Acetilación , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Complicaciones de la Diabetes/inmunología , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Histonas/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/inmunología , Hiperglucemia/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Linfocitos T/inmunología , Virosis/complicaciones , Virosis/inmunología , Virosis/mortalidad , Virus/inmunología , Modelos Animales de Enfermedad , Humanos
13.
Cell ; 153(2): 413-25, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582329

RESUMEN

Here, we demonstrate that the fractalkine (FKN)/CX3CR1 system represents a regulatory mechanism for pancreatic islet ß cell function and insulin secretion. CX3CR1 knockout (KO) mice exhibited a marked defect in glucose and GLP1-stimulated insulin secretion, and this defect was also observed in vitro in isolated islets from CX3CR1 KO mice. In vivo administration of FKN improved glucose tolerance with an increase in insulin secretion. In vitro treatment of islets with FKN increased intracellular Ca(2+) and potentiated insulin secretion in both mouse and human islets. The KO islets exhibited reduced expression of a set of genes necessary for the fully functional, differentiated ß cell state, whereas treatment of wild-type (WT) islets with FKN led to increased expression of these genes. Lastly, expression of FKN in islets was decreased by aging and high-fat diet/obesity, suggesting that decreased FKN/CX3CR1 signaling could be a mechanism underlying ß cell dysfunction in type 2 diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores de Quimiocina/metabolismo , Transducción de Señal , Adulto , Envejecimiento , Animales , Receptor 1 de Quimiocinas CX3C , Cadáver , Quimiocina CX3CL1/administración & dosificación , Quimiocina CX3CL1/metabolismo , Dieta Alta en Grasa , Expresión Génica , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de Quimiocina/genética
14.
Nature ; 605(7911): 761-766, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35585240

RESUMEN

Diabetes mellitus is prevalent among women of reproductive age, and many women are left undiagnosed or untreated1. Gestational diabetes has profound and enduring effects on the long-term health of the offspring2,3. However, the link between pregestational diabetes and disease risk into adulthood in the next generation has not been sufficiently investigated. Here we show that pregestational hyperglycaemia renders the offspring more vulnerable to glucose intolerance. The expression of TET3 dioxygenase, responsible for 5-methylcytosine oxidation and DNA demethylation in the zygote4, is reduced in oocytes from a mouse model of hyperglycaemia (HG mice) and humans with diabetes. Insufficient demethylation by oocyte TET3 contributes to hypermethylation at the paternal alleles of several insulin secretion genes, including the glucokinase gene (Gck), that persists from zygote to adult, promoting impaired glucose homeostasis largely owing to the defect in glucose-stimulated insulin secretion. Consistent with these findings, mouse progenies derived from the oocytes of maternal heterozygous and homozygous Tet3 deletion display glucose intolerance and epigenetic abnormalities similar to those from the oocytes of HG mice. Moreover, the expression of exogenous Tet3 mRNA in oocytes from HG mice ameliorates the maternal effect in offspring. Thus, our observations suggest an environment-sensitive window in oocyte development that confers predisposition to glucose intolerance in the next generation through TET3 insufficiency rather than through a direct perturbation of the oocyte epigenome. This finding suggests a potential benefit of pre-conception interventions in mothers to protect the health of offspring.


Asunto(s)
Dioxigenasas , Intolerancia a la Glucosa , Hiperglucemia , Oocitos , Adulto , Animales , Dioxigenasas/metabolismo , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/genética , Hiperglucemia/metabolismo , Herencia Materna , Ratones , Oocitos/metabolismo
15.
Mol Cell ; 79(1): 43-53.e4, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32464093

RESUMEN

The physiological role of immune cells in the regulation of postprandial glucose metabolism has not been fully elucidated. We have found that adipose tissue macrophages produce interleukin-10 (IL-10) upon feeding, which suppresses hepatic glucose production in cooperation with insulin. Both elevated insulin and gut-microbiome-derived lipopolysaccharide in response to feeding are required for IL-10 production via the Akt/mammalian target of rapamycin (mTOR) pathway. Indeed, myeloid-specific knockout of the insulin receptor or bone marrow transplantation of mutant TLR4 marrow cells results in increased expression of gluconeogenic genes and impaired glucose tolerance. Furthermore, myeloid-specific Akt1 and Akt2 knockout results in similar phenotypes that are rescued by additional knockout of TSC2, an inhibitor of mTOR. In obesity, IL-10 production is impaired due to insulin resistance in macrophages, whereas adenovirus-mediated expression of IL-10 ameliorates postprandial hyperglycemia. Thus, the orchestrated response of the endogenous hormone and gut environment to feeding is a key regulator of postprandial glycemia.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Hiperglucemia/patología , Insulina/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Tejido Adiposo/metabolismo , Animales , Glucemia/análisis , Gluconeogénesis/genética , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Interleucina-10/fisiología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Periodo Posprandial , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/fisiología
16.
Proc Natl Acad Sci U S A ; 121(17): e2320934121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38630726

RESUMEN

Cullin RING E3 ligases (CRL) have emerged as key regulators of disease-modifying pathways and therapeutic targets. Cullin3 (Cul3)-containing CRL (CRL3) has been implicated in regulating hepatic insulin and oxidative stress signaling. However, CRL3 function in liver pathophysiology is poorly defined. Here, we report that hepatocyte Cul3 knockout results in rapid resolution of steatosis in obese mice. However, the remarkable resistance of hepatocyte Cul3 knockout mice to developing steatosis does not lead to overall metabolic improvement but causes systemic metabolic disturbances. Liver transcriptomics analysis identifies that CRL3 inactivation causes persistent activation of the nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant defense pathway, which also reprograms the lipid transcriptional network to prevent TG storage. Furthermore, global metabolomics reveals that NRF2 activation induces numerous NAD+-consuming aldehyde dehydrogenases to increase the cellular NADH/NAD+ ratio, a redox imbalance termed NADH reductive stress that inhibits the glycolysis-citrate-lipogenesis axis in Cul3 knockout livers. As a result, this NRF2-induced cellular lipid storage defect promotes hepatic ceramide accumulation, elevates circulating fatty acids, and worsens systemic insulin resistance in a vicious cycle. Hepatic lipid accumulation is restored, and liver injury and hyperglycemia are attenuated when NRF2 activation and NADH reductive stress are abolished in hepatocyte Cul3/Nrf2 double-knockout mice. The resistance to hepatic steatosis, hyperglycemia, and NADH reductive stress are observed in hepatocyte Keap1 knockout mice with NRF2 activation. In summary, our study defines a critical role of CRL3 in hepatic metabolic regulation and demonstrates that the CRL3 downstream NRF2 overactivation causes hepatic metabolic maladaptation to obesity and insulin resistance.


Asunto(s)
Hígado Graso , Hiperglucemia , Resistencia a la Insulina , Animales , Ratones , Ubiquitina-Proteína Ligasas/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , NAD/metabolismo , Proteínas Cullin/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Ratones Noqueados , Lípidos
17.
PLoS Biol ; 21(9): e3002323, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37738230

RESUMEN

The subcommissural organ (SCO) is a secretory tissue located on the roof of the brain's third ventricle. A new study published in PLOS Biology finds that the SCO responds to glucose by secreting signaling molecules into the cerebrospinal fluid (CSF), thereby decreasing the local ependyma-driven CSF movement.


Asunto(s)
Hiperglucemia , Humanos , Glucosa , Transporte Biológico , Moléculas de Adhesión Celular Neuronal
18.
PLoS Biol ; 21(9): e3002308, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37733692

RESUMEN

Hyperglycemia increases glucose concentrations in the cerebrospinal fluid (CSF), activating glucose-sensing mechanisms and feeding behavior in the hypothalamus. Here, we discuss how hyperglycemia temporarily modifies ependymal cell ciliary beating to increase hypothalamic glucose sensing. A high level of glucose in the rat CSF stimulates glucose transporter 2 (GLUT2)-positive subcommissural organ (SCO) cells to release SCO-spondin into the dorsal third ventricle. Genetic inactivation of mice GLUT2 decreases hyperglycemia-induced SCO-spondin secretion. In addition, SCO cells secrete Wnt5a-positive vesicles; thus, Wnt5a and SCO-spondin are found at the apex of dorsal ependymal cilia to regulate ciliary beating. Frizzled-2 and ROR2 receptors, as well as specific proteoglycans, such as glypican/testican (essential for the interaction of Wnt5a with its receptors) and Cx43 coupling, were also analyzed in ependymal cells. Finally, we propose that the SCO-spondin/Wnt5a/Frizzled-2/Cx43 axis in ependymal cells regulates ciliary beating, a cyclic and adaptive signaling mechanism to control glucose sensing.


Asunto(s)
Conexina 43 , Hiperglucemia , Animales , Ratones , Ratas , Neuroglía , Glucosa , Proteína Wnt-5a/genética
19.
EMBO Rep ; 25(1): 128-143, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177907

RESUMEN

Collateral circulation is essential for blood resupply to the ischemic heart, which is dictated by the contractile phenotypic restoration of vascular smooth muscle cells (VSMC). Here we investigate whether S-nitrosylation of AMP-activated protein kinase (AMPK), a key regulator of the VSMC phenotype, impairs collateral circulation. In rats with collateral growth and development, nitroglycerin decreases coronary collateral blood flow (CCBF), inhibits vascular contractile phenotypic restoration, and increases myocardial infarct size, accompanied by reduced AMPK activity in the collateral zone. Nitric oxide (NO) S-nitrosylates human recombinant AMPKγ1 at cysteine 131 and decreases AMP sensitivity of AMPK. In VSMCs, exogenous expression of S-nitrosylation-resistant AMPKγ1 or deficient NO synthase (iNOS) prevents the disruption of VSMC reprogramming. Finally, hyperhomocysteinemia or hyperglycemia increases AMPKγ1 S-nitrosylation, prevents vascular contractile phenotypic restoration, reduces CCBF, and increases the infarct size of the heart in Apoe-/- mice, all of which is rescued in Apoe-/-/iNOSsm-/- mice or Apoe-/- mice with enforced expression of the AMPKγ1-C130A mutant following RI/MI. We conclude that nitrosative stress disrupts coronary collateral circulation during hyperhomocysteinemia or hyperglycemia through AMPK S-nitrosylation.


Asunto(s)
Hiperglucemia , Hiperhomocisteinemia , Ratas , Ratones , Humanos , Animales , Circulación Colateral , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Músculo Liso Vascular , Hiperhomocisteinemia/metabolismo , Apolipoproteínas E/metabolismo , Hiperglucemia/metabolismo
20.
EMBO Rep ; 25(4): 1752-1772, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38491313

RESUMEN

Emerging evidence indicates that parental diseases can impact the health of subsequent generations through epigenetic inheritance. Recently, it was shown that maternal diabetes alters the metaphase II oocyte transcriptome, causing metabolic dysfunction in offspring. However, type 1 diabetes (T1D) mouse models frequently utilized in previous studies may be subject to several confounding factors due to severe hyperglycemia. This limits clinical translatability given improvements in glycemic control for T1D subjects. Here, we optimize a T1D mouse model to investigate the effects of appropriately managed maternal glycemic levels on oocytes and intrauterine development. We show that diabetic mice with appropriate glycemic control exhibit better long-term health, including maintenance of the oocyte transcriptome and chromatin accessibility. We further show that human oocytes undergoing in vitro maturation challenged with mildly increased levels of glucose, reflecting appropriate glycemic management, also retain their transcriptome. However, fetal growth and placental function are affected in mice despite appropriate glycemic control, suggesting the uterine environment rather than the germline as a pathological factor in developmental programming in appropriately managed diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Hiperglucemia , Humanos , Femenino , Embarazo , Ratones , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Placenta , Hiperglucemia/genética , Hiperglucemia/metabolismo , Oocitos/metabolismo , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA