Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nat Metab ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937658
2.
J Microbiol ; 62(3): 137-152, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38587593

RESUMEN

In the evolving landscape of cancer research, the human microbiome emerges as a pivotal determinant reshaping our understanding of tumorigenesis and therapeutic responses. Advanced sequencing technologies have uncovered a vibrant microbial community not confined to the gut but thriving within tumor tissues. Comprising bacteria, viruses, and fungi, this diverse microbiota displays distinct signatures across various cancers, with most research primarily focusing on bacteria. The correlations between specific microbial taxa within different cancer types underscore their pivotal roles in driving tumorigenesis and influencing therapeutic responses, particularly in chemotherapy and immunotherapy. This review amalgamates recent discoveries, emphasizing the translocation of the oral microbiome to the gut as a potential marker for microbiome dysbiosis across diverse cancer types and delves into potential mechanisms contributing to cancer promotion. Furthermore, it highlights the adverse effects of the microbiome on cancer development while exploring its potential in fortifying strategies for cancer prevention and treatment.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Neoplasias , Humanos , Neoplasias/microbiología , Neoplasias/terapia , Disbiosis/microbiología , Microbiota , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Carcinogénesis , Inmunoterapia , Boca/microbiología
3.
Mol Metab ; 83: 101924, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521185

RESUMEN

OBJECTIVES: Gut microbiota increases energy availability through fermentation of dietary fibers to short-chain fatty acids in conventionally raised mice. Energy deficiency in germ-free (GF) mice increases glucagon-like peptide-1 (GLP-1) levels, which slows intestinal transit. To further analyze the role of GLP-1-mediated signaling in this model of energy deficiency, we re-derived mice lacking GLP-1 receptor (GLP-1R KO) as GF. METHODS: GLP-1R KO mice were rederived as GF through hysterectomy and monitored for 30 weeks. Mice were subjected to rescue experiments either through feeding an energy-rich diet or colonization with a normal cecal microbiota. Histology and intestinal function were assessed at different ages. Intestinal organoids were assessed to investigate stemness. RESULTS: Unexpectedly, 25% of GF GLP-1R KO mice died before 20 weeks of age, associated with enlarged ceca, increased cecal water content, increased colonic expression of apical ion transporters, reduced number of goblet cells and loss of colonic epithelial integrity. Colonocytes from GLP-1R KO mice were energy-deprived and exhibited increased ER-stress; mitochondrial fragmentation, increased oxygen levels and loss of stemness. Restoring colonic energy levels either by feeding a Western-style diet or colonization with a normal gut microbiota normalized gut phenotypes and prevented lethality. CONCLUSIONS: Our findings reveal a heretofore unrecognized role for GLP-1R signaling in the maintenance of colonic physiology and survival during energy deprivation.


Asunto(s)
Colon , Metabolismo Energético , Microbioma Gastrointestinal , Receptor del Péptido 1 Similar al Glucagón , Células Caliciformes , Ratones Noqueados , Transducción de Señal , Animales , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Microbioma Gastrointestinal/fisiología , Ratones , Células Caliciformes/metabolismo , Colon/metabolismo , Colon/microbiología , Ratones Endogámicos C57BL , Masculino , Femenino , Péptido 1 Similar al Glucagón/metabolismo
4.
BMB Rep ; 56(9): 469-481, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37605613

RESUMEN

The gut microbiome is widely recognized as a dynamic organ with a profound influence on human physiology and pathology. Extensive epidemiological and longitudinal cohort studies have provided compelling evidence that disruptions in the early-life microbiome can have long-lasting health implications. Various factors before, during, and after birth contribute to shaping the composition and function of the neonatal and infant microbiome. While these alterations can be partially restored over time, metabolic phenotypes may persist, necessitating research to identify the critical period for early intervention to achieve phenotypic recovery beyond microbiome composition. In this review, we provide current understanding of changes in the gut microbiota throughout life and the various factors affecting these changes. Specifically, we highlight the profound impact of early-life gut microbiota disruption on the development of diseases later in life and discuss perspectives on efforts to recover from such disruptions. [BMB Reports 2023; 56(9): 469-481].


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Recién Nacido , Humanos , Estudios Longitudinales , Cicatriz , Microbioma Gastrointestinal/fisiología , Fenotipo
6.
Int J Stem Cells ; 15(1): 70-84, 2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35220293

RESUMEN

The advent of human intestinal organoid systems has revolutionized the way we understand the interactions between the human gut and microorganisms given the host tropism of human microorganisms. The gut microorganisms have regionality (i.e., small versus large intestine) and the expression of various virulence factors in pathogens is influenced by the gut milieu. However, the culture conditions, optimized for human intestinal organoids, often do not fully support the proliferation and functionality of gut microorganisms. In addition, the regional identity of human intestinal organoids has not been considered to study specific microorganisms with regional preference. In this review we provide an overview of current efforts to understand the role of microorganisms in human intestinal organoids. Specifically, we will emphasize the importance of matching the regional preference of microorganisms in the gut and tailoring the appropriate luminal environmental conditions (i.e., oxygen, pH, and biochemical levels) for modeling real interactions between the gut and the microorganisms with human intestinal organoids.

7.
J Exp Med ; 219(2)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-34940790

RESUMEN

Phospholipase D (PLD)2 via its enzymatic activity regulates cell proliferation and migration and thus is implicated in cancer. However, the role of PLD2 in obesity and type 2 diabetes has not previously been investigated. Here, we show that during diet-induced thermogenesis and obesity, levels of PLD2 but not PLD1 in adipose tissue are inversely related with uncoupling protein 1, a key thermogenic protein. We demonstrate that the thermogenic program in adipose tissue is significantly augmented in mice with adipocyte-specific Pld2 deletion or treated with a PLD2-specific inhibitor and that these mice are resistant to high fat diet-induced obesity, glucose intolerance, and insulin resistance. Mechanistically, we show that Pld2 deletion in adipose tissue or PLD2 pharmacoinhibition acts via p62 to improve mitochondrial quality and quantity in adipocytes. Thus, PLD2 inhibition is an attractive therapeutic approach for obesity and type 2 diabetes by resolving defects in diet-induced thermogenesis.


Asunto(s)
Adipocitos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Fosfolipasa D/genética , Termogénesis/genética , Animales , Biomarcadores , Glucemia , Dieta Alta en Grasa , Metabolismo Energético , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Inmunohistoquímica , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Mitocondrias/ultraestructura , Obesidad/etiología , Obesidad/metabolismo , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
8.
Eur Heart J ; 42(43): 4481-4492, 2021 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-34297830

RESUMEN

AIMS: Cardiac injury and remodelling are associated with the rearrangement of cardiac lipids. Glycosphingolipids are membrane lipids that are important for cellular structure and function, and cardiac dysfunction is a characteristic of rare monogenic diseases with defects in glycosphingolipid synthesis and turnover. However, it is not known how cardiac glycosphingolipids regulate cellular processes in the heart. The aim of this study is to determine the role of cardiac glycosphingolipids in heart function. METHODS AND RESULTS: Using human myocardial biopsies, we showed that the glycosphingolipids glucosylceramide and lactosylceramide are present at very low levels in non-ischaemic human heart with normal function and are elevated during remodelling. Similar results were observed in mouse models of cardiac remodelling. We also generated mice with cardiomyocyte-specific deficiency in Ugcg, the gene encoding glucosylceramide synthase (hUgcg-/- mice). In 9- to 10-week-old hUgcg-/- mice, contractile capacity in response to dobutamine stress was reduced. Older hUgcg-/- mice developed severe heart failure and left ventricular dilatation even under baseline conditions and died prematurely. Using RNA-seq and cell culture models, we showed defective endolysosomal retrograde trafficking and autophagy in Ugcg-deficient cardiomyocytes. We also showed that responsiveness to ß-adrenergic stimulation was reduced in cardiomyocytes from hUgcg-/- mice and that Ugcg knockdown suppressed the internalization and trafficking of ß1-adrenergic receptors. CONCLUSIONS: Our findings suggest that cardiac glycosphingolipids are required to maintain ß-adrenergic signalling and contractile capacity in cardiomyocytes and to preserve normal heart function.


Asunto(s)
Glucosiltransferasas , Miocitos Cardíacos , Animales , Cardiomegalia , Glucosiltransferasas/genética , Ratones , Receptores Adrenérgicos
9.
Mamm Genome ; 32(4): 206-222, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33646347

RESUMEN

Accumulating evidence has revealed the link between the microbiota and various human diseases. Advances in high-throughput sequencing technologies have identified some consistent disease-associated microbial features, leading to the emerging concept of microbiome-based therapeutics. However, it is also becoming clear that there are considerable variations in the microbiota among patients with the same disease. Variations in the microbial composition and function contribute to substantial differences in metabolic status of the host via production of a myriad of biochemically and functionally different microbial metabolites. Indeed, compelling evidence indicates that individuality of the microbiome may result in individualized responses to microbiome-based therapeutics and other interventions. Mechanistic understanding of the role of the microbiota in diseases and drug metabolism would help us to identify causal relationships and thus guide the development of microbiome-based precision or personalized medicine. In this review, we provide an overview of current efforts to use microbiome-based interventions for the treatment of diseases such as cancer, neurological disorders, and diabetes to approach precision medicine.


Asunto(s)
Microbioma Gastrointestinal/genética , Genoma Humano/genética , Inactivación Metabólica/genética , Medicina de Precisión , Diabetes Mellitus/genética , Diabetes Mellitus/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Neoplasias/genética , Neoplasias/microbiología , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/microbiología
10.
Nat Commun ; 12(1): 1347, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33649331

RESUMEN

The human microbiome can produce metabolites that modulate insulin signaling. Type 2 diabetes patients have increased circulating concentrations of the microbially produced histidine metabolite, imidazole propionate (ImP) and administration of ImP in mice resulted in impaired glucose tolerance. Interestingly, the fecal microbiota of the patients had increased capacity to produce ImP, which is mediated by the bacterial enzyme urocanate reductase (UrdA). Here, we describe the X-ray structures of the ligand-binding domains of UrdA in four different states, representing the structural transitions along the catalytic reaction pathway of this unexplored enzyme linked to disease in humans. The structures in combination with functional data provide key insights into the mechanism of action of UrdA that open new possibilities for drug development strategies targeting type 2 diabetes.


Asunto(s)
Imidazoles/metabolismo , Oxidorreductasas/metabolismo , Shewanella/enzimología , Ácido Urocánico/metabolismo , Arginina/metabolismo , Dominio Catalítico , Flavina-Adenina Dinucleótido/metabolismo , Imidazoles/química , Cinética , Ligandos , Modelos Moleculares , Oxidorreductasas/química , Conformación Proteica , Dominios Proteicos , Especificidad por Sustrato , Termodinámica , Ácido Urocánico/química
11.
Cell Metab ; 32(4): 643-653.e4, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32783890

RESUMEN

Metformin is the first-line therapy for type 2 diabetes, but there are large inter-individual variations in responses to this drug. Its mechanism of action is not fully understood, but activation of AMP-activated protein kinase (AMPK) and changes in the gut microbiota appear to be important. The inhibitory role of microbial metabolites on metformin action has not previously been investigated. Here, we show that concentrations of the microbial metabolite imidazole propionate are higher in subjects with type 2 diabetes taking metformin who have high blood glucose. We also show that metformin-induced glucose lowering is not observed in mice pretreated with imidazole propionate. Furthermore, we demonstrate that imidazole propionate inhibits AMPK activity by inducing inhibitory AMPK phosphorylation, which is dependent on imidazole propionate-induced basal Akt activation. Finally, we identify imidazole propionate-activated p38γ as a novel kinase for Akt and demonstrate that p38γ kinase activity mediates the inhibitory action of imidazole propionate on metformin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Imidazoles/farmacología , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipoglucemiantes/farmacología , Imidazoles/administración & dosificación , Imidazoles/metabolismo , Inyecciones Intraperitoneales , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos
12.
Mol Metab ; 37: 100997, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32305515

RESUMEN

OBJECTIVE: Gut-derived inflammatory factors can impair glucose homeostasis, but the underlying mechanisms are not fully understood. In this study, we investigated how hepatic gene expression is regulated by gut colonization status through myeloid differentiation primary response 88 (MYD88) and how one of the regulated genes, lipopolysaccharide-binding protein (Lbp), affects insulin signaling and systemic glucose homeostasis. METHODS: Liver transcriptomics analysis was conducted on four groups of mice fed a chow diet: conventionally raised (CONV-R) wild-type, germ-free (GF) wild-type, CONV-R Myd88 KO, and GF Myd88 KO. Primary hepatocytes were exposed to combinations of lipopolysaccharide (LPS), LBP, and the LBP-blocking peptide LBPK95A, and the effect on insulin signaling was determined. To assess how LBP affects glucose metabolism in vivo, two mouse models were applied: treatment with LBPK95A and hepatic knockdown of Lbp using CRISPR-CAS9. RESULTS: We showed that the colonization status regulates gene expression in the liver and that a subset of these genes, including Lbp, is regulated through MYD88. Furthermore, we demonstrated that LBP impairs insulin signaling in hepatocytes in the presence of low levels of LPS and that the effect of LBP is abolished by LBPK95A. We showed that both systemic pharmacological blocking of LBP by LBPK95A and CRISPR-CAS9-mediated downregulation of hepatic Lbp improve glucose homeostasis. CONCLUSIONS: Our results demonstrate that the gut microbiota regulates hepatic expression of Lbp through MYD88-dependent signaling. LBP potentiates LPS inhibition of insulin signaling in vitro and impairs systemic glucose homeostasis in vivo.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Proteínas Portadoras/metabolismo , Glucosa/metabolismo , Glicoproteínas de Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas de Fase Aguda/genética , Animales , Metabolismo de los Hidratos de Carbono/fisiología , Proteínas Portadoras/genética , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Expresión Génica , Prueba de Tolerancia a la Glucosa , Hepatocitos/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/farmacología , Factor 88 de Diferenciación Mieloide/fisiología , Obesidad/metabolismo , Obesidad/fisiopatología , Transducción de Señal
13.
Mol Cell ; 78(4): 584-596, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32234490

RESUMEN

Many genomic studies have revealed associations between the gut microbiota composition and host metabolism. These observations led to the idea that a causal relationship could exist between the microbiota and metabolic diseases, a concept supported by studies showing compositional changes in the microbial community in metabolic diseases and transmissibility of host phenotype via microbiota transfer. Accumulating data suggest that the microbiota may affect host metabolic phenotypes through the production of metabolites. These bioactive microbial metabolites, sensitive fingerprints of microbial function, can act as inter-kingdom signaling messengers via penetration into host blood circulation and tissues. These fingerprints may be used for diagnostic purposes, and increased understanding of strain specificity in producing microbial metabolites can identify bacterial strains or specific metabolites that can be used for therapeutic purposes. Here, we will review data supporting the causal role of the gut microbiota in metabolism and discuss mechanisms and potential clinical implications.


Asunto(s)
Bacterias/metabolismo , Bacterias/patogenicidad , Microbioma Gastrointestinal , Enfermedades Metabólicas/etiología , Humanos , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología
14.
Mol Metab ; 23: 82-87, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30904385

RESUMEN

OBJECTIVES: The nuclear receptor superfamily is a potential target for the development of new treatments for obesity and metabolic diseases. Increasing evidence has pointed towards the retinoic acid-related orphan receptor-alpha (RORα) as an important nuclear receptor involved in several biological processes. RORα full body knockout mice display improved metabolic phenotypes on both chow and high fat (60% fat, 20% carbohydrate) diets, but also have severe behavioral abnormalities. Here we investigated the effect of hepatic RORα by generating mice with liver-specific RORα deletion to elucidate the role of this nuclear receptor on host metabolism. METHODS: 8 week-old mice with liver-specific RORα deletion and littermate controls were fed either chow or western-style diets (40% fat, 40% carbohydrate) for 12 weeks. Metabolic phenotyping was performed at the end of the dietary intervention. RESULTS: Here, we show that hepatic RORα deletion does not affect the metabolic susceptibility to either chow or western-style diet in terms of glucose metabolism and adiposity. CONCLUSIONS: Our data indicate that liver deletion of RORα does not have a pivotal role in the regulation of hepatic glucose and lipid metabolism on chow or western-style diet.


Asunto(s)
Dieta Occidental , Glucosa/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Tejido Adiposo Blanco/metabolismo , Adiposidad/genética , Animales , Dieta Vegetariana , Femenino , Técnicas de Inactivación de Genes , Hepatocitos/metabolismo , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo
15.
J Clin Pharmacol ; 59(4): 532-540, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30452773

RESUMEN

Amitriptyline is a tricyclic antidepressant that is metabolized mainly by CYP2C19 and CYP2D6 enzymes. Higher plasma levels of amitriptyline and its active metabolite, nortriptyline, are associated with an increased risk of adverse events including anticholinergic effects. The aim of this study was to evaluate the effects of CYP2C19 and CYP2D6 genetic polymorphisms on amitriptyline and nortriptyline pharmacokinetics. Twenty-four Korean healthy adult male volunteers were enrolled in the study after stratification by their CYP2C19 and CYP2D6 genotypes. Serial blood draws for pharmacokinetic analysis were made after a single oral 25-mg dose of amitriptyline was administered. Plasma amitriptyline and nortriptyline concentrations were measured by a validated liquid chromatography with tandem mass spectrometry. Population pharmacokinetic modeling analysis was conducted using NONMEM, which evaluated the effects of CYP2C19 and CYP2D6 genotypes on amitriptyline and nortriptyline pharmacokinetics. The biotransformation of amitriptyline into nortriptyline was significantly different between subjects with the CYP2C19*2/*2, *2/*3, and *3/*3 genotypes and those with the other genotypes, with an estimated metabolic clearance of 17 and 61.5 L/h, respectively. Clearance of amitriptyline through pathways other than biotransformation into nortriptyline was estimated as 18.8 and 30.6 L/h for subjects with the CYP2D6*10/*10 and *10/*5 genotypes and those with the other genotypes, respectively. This study demonstrated a quantitative effect of the CYP2C19 and CYP2D6 genotypes on amitriptyline and nortriptyline pharmacokinetics. Production of nortriptyline from amitriptyline was associated with CYP2C19 genotypes, and clearance of amitriptyline through pathways other than biotransformation into nortriptyline was associated with CYP2D6 genotypes. These observations may be useful in developing individualized, optimal therapy with amitriptyline.


Asunto(s)
Amitriptilina/farmacocinética , Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2D6/genética , Modelos Biológicos , Nortriptilina/farmacocinética , Adulto , Antidepresivos Tricíclicos/farmacocinética , Cromatografía Liquida , Genotipo , Humanos , Masculino , Polimorfismo Genético , Espectrometría de Masas en Tándem , Adulto Joven
16.
Cell ; 175(4): 947-961.e17, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401435

RESUMEN

Interactions between the gut microbiota, diet, and the host potentially contribute to the development of metabolic diseases. Here, we identify imidazole propionate as a microbially produced histidine-derived metabolite that is present at higher concentrations in subjects with versus without type 2 diabetes. We show that imidazole propionate is produced from histidine in a gut simulator at higher concentrations when using fecal microbiota from subjects with versus without type 2 diabetes and that it impairs glucose tolerance when administered to mice. We further show that imidazole propionate impairs insulin signaling at the level of insulin receptor substrate through the activation of p38γ MAPK, which promotes p62 phosphorylation and, subsequently, activation of mechanistic target of rapamycin complex 1 (mTORC1). We also demonstrate increased activation of p62 and mTORC1 in liver from subjects with type 2 diabetes. Our findings indicate that the microbial metabolite imidazole propionate may contribute to the pathogenesis of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Microbioma Gastrointestinal , Imidazoles/metabolismo , Insulina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/microbiología , Células HEK293 , Histidina/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Sequestosoma-1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Cell Signal ; 51: 130-138, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30092354

RESUMEN

Regulation of tyrosine phosphorylation on insulin receptor substrate-1 (IRS-1) is essential for insulin signaling. The protein tyrosine phosphatase (PTP) C1-Ten/Tensin2 has been implicated in the regulation of IRS-1, but the molecular basis of this dephosphorylation is not fully understood. Here, we demonstrate that the cellular phosphatase activity of C1-Ten/Tensin2 on IRS-1 is mediated by the binding of the C1-Ten/Tensin2 Src-homology 2 (SH2) domain to phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3). We show that the role of C1-Ten/Tensin2 is dependent on insulin-induced phosphoinositide 3-kinase activity. The C1-Ten/Tensin2 SH2 domain showed strong preference and high affinity for PtdIns(3,4,5)P3. Using site-directed mutagenesis, we identified three basic residues in the C1-Ten/Tensin2 SH2 domain that were critical for PtdIns(3,4,5)P3 binding but were not involved in phosphotyrosine binding and PTP activity. Using a PtdIns(3,4,5)P3 binding-deficient mutant, we showed that the specific binding of the C1-Ten/Tensin2 SH2 domain to PtdIns(3,4,5)P3 allowed C1-Ten/Tensin2 to function as a PTP in cells. Collectively, our findings suggest that the interaction between the C1-Ten/Tensin2 SH2 domain and PtdIns(3,4,5)P3 produces a negative feedback loop of insulin signaling through IRS-1.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Tensinas/química , Tensinas/metabolismo , Dominios Homologos src , Animales , Escherichia coli , Células HEK293 , Humanos , Células L , Ratones , Mutagénesis Sitio-Dirigida , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Tensinas/genética
18.
Sci Rep ; 7(1): 17777, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29259227

RESUMEN

Insulin resistance causes type 2 diabetes; therefore, increasing insulin sensitivity is a therapeutic approach against type 2 diabetes. Activating AMP-activated protein kinase (AMPK) is an effective approach for treating diabetes, and reduced insulin receptor substrate-1 (IRS-1) protein levels have been suggested as a molecular mechanism causing insulin resistance. Thus, dual targeting of AMPK and IRS-1 might provide an ideal way to treat diabetes. We found that 15,16-dihydrotanshinone I (DHTS), as a C1-Ten protein tyrosine phosphatase inhibitor, increased IRS-1 stability, improved glucose tolerance and reduced muscle atrophy. Identification of DHTS as a C1-Ten inhibitor revealed a new function of C1-Ten in AMPK inhibition, possibly through regulation of IRS-1. These findings suggest that C1-Ten inhibition by DHTS could provide a novel therapeutic strategy for insulin resistance-associated metabolic syndrome through dual targeting of IRS-1 and AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina/fisiología , Fenantrenos/farmacología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Furanos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Humanos , Hipoglucemiantes/farmacología , Insulina/metabolismo , Masculino , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/metabolismo , Quinonas
19.
Sci Rep ; 7(1): 12346, 2017 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-28955049

RESUMEN

Hypertrophy is a prominent feature of damaged podocytes in diabetic kidney disease (DKD). mTORC1 hyperactivation leads to podocyte hypertrophy, but the detailed mechanism of how mTORC1 activation occurs under pathological conditions is not completely known. Moreover, reduced nephrin tyrosine phosphorylation has been observed in podocytes under pathological conditions, but the molecular mechanism linking nephrin phosphorylation and pathology is unclear so far. In this study, we observed a significant increase in C1-Ten level in diabetic kidney and in high glucose-induced damaged podocytes. C1-Ten acts as a protein tyrosine phosphatase (PTPase) at the nephrin-PI3K binding site and renders PI3K for IRS-1, thereby activating mTORC1. Furthermore, C1-Ten causes podocyte hypertrophy and proteinuria by increasing mTORC1 activity in vitro and in vivo. These findings demonstrate the relationship between nephrin dephosphorylation and the mTORC1 pathway, mediated by C1-Ten PTPase activity. We suggest that C1-Ten contributes to the pathogenesis of DKD by inducing podocyte hypertrophy under high glucose conditions.


Asunto(s)
Nefropatías Diabéticas/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Membrana/metabolismo , Podocitos/patología , Proteínas Tirosina Fosfatasas/metabolismo , Tensinas/metabolismo , Animales , Glucosa/metabolismo , Células HEK293 , Humanos , Hipertrofia/patología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteinuria/etiología , Proteinuria/patología , Transducción de Señal
20.
Cell ; 165(6): 1332-1345, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259147

RESUMEN

A compelling set of links between the composition of the gut microbiota, the host diet, and host physiology has emerged. Do these links reflect cause-and-effect relationships, and what might be their mechanistic basis? A growing body of work implicates microbially produced metabolites as crucial executors of diet-based microbial influence on the host. Here, we will review data supporting the diverse functional roles carried out by a major class of bacterial metabolites, the short-chain fatty acids (SCFAs). SCFAs can directly activate G-coupled-receptors, inhibit histone deacetylases, and serve as energy substrates. They thus affect various physiological processes and may contribute to health and disease.


Asunto(s)
Fibras de la Dieta/metabolismo , Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal/fisiología , Animales , Histonas/metabolismo , Humanos , Intestinos/inmunología , Intestinos/microbiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...