Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Blood Cell Ther ; 7(1): 14-24, 2024 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-38486826

RESUMEN

Patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT) procedures often experience high levels of uncertainty. In this study, we developed and implemented a nursing intervention program to help patients recognize and reduce pre-transplant uncertainty. This study used a pretest-posttest single-group design without a control group. Eighteen patients undergoing HSCT participated in the intervention program-which included informational support, confirmation that the patients understood the information provided, and emotional support. Outpatients received the intervention at their initial outpatient visits after their procedure dates were determined, while inpatients received it at discharge following their procedures. The Universal Uncertainty in Illness Scale (UUIS), which consists of 26 items and six subscales, was used as the primary outcome measure. The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) and the Hospital Anxiety and Depression Scale were used as secondary outcome measures. The sample included 18 individuals (13 male and five female participants; median age, 52 years). Most participants had acute lymphoblastic leukemia and had previously undergone bone marrow transplantations. Following our intervention, the total UUIS score significantly decreased, from 80.83 ± 18.42 before the intervention to 63.06 ± 23.53 afterward (t = 4.98, p < .001). Furthermore, significant post-intervention reductions were observed for all six subscales of the UUIS. There were no significant differences in the functional EORTC QLQ-C30 scale scores; however, the symptom scale showed a significant decrease in fatigue (pre = 35.19 ± 19.53, post = 25.93 ± 17.04, Z = -1.99, p < 0.046) and constipation (pre = 20.37 ± 20.26, post = 7.41 ± 14.26, Z = -2.11, p = 0.035). There were no significant differences in anxiety and depression levels pre- and post-intervention. Overall, the intervention effectively reduced both UUIS total and subscale scores related to pre-HSCT uncertainties. Assessing uncertainty prior to HSCT is vital to assisting patients in coping with the procedure. Nurses not only provide information but also tailor the information to the patients' cognitive abilities, thereby simplifying their understanding of the disease and its treatment.

2.
Am J Pathol ; 194(5): 693-707, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38309428

RESUMEN

Glucose lowering independently reduces liver fibrosis in human nonalcoholic fatty liver disease. This study investigated the impact of diabetes on steatohepatitis and established a novel mouse model for diabetic steatohepatitis. Male C57BL/6J mice were fed a 60% high-fat diet (HFD) and injected with carbon tetrachloride (CCl4) and streptozotocin (STZ) to induce diabetes. The HFD+CCl4+STZ group showed more severe liver steatosis, hepatocyte ballooning, and regenerative nodules compared with other groups. Diabetes up-regulated inflammatory cytokine-associated genes and increased the M1/M2 macrophage ratios in the liver. Single-cell RNA sequencing analysis of nonparenchymal cells in the liver showed that diabetes reduced Kupffer cells and increased bone marrow-derived recruited inflammatory macrophages, such as Ly6Chi-RM. Diabetes globally reduced liver sinusoidal endothelial cells (LSECs). Furthermore, genes related to the receptor for advanced glycation end products (RAGE)/Toll-like receptor 4 (TLR4) were up-regulated in Ly6Chi-RM and LSECs in mice with diabetes, suggesting a possible role of RAGE/TLR4 signaling in the interaction between inflammatory macrophages and LSECs. This study established a novel diabetic steatohepatitis model using a combination of HFD, CCl4, and STZ. Diabetes exacerbated steatosis, hepatocyte ballooning, fibrosis, regenerative nodule formation, and the macrophage M1/M2 ratios triggered by HFD and CCl4. Single-cell RNA sequencing analysis indicated that diabetes activated inflammatory macrophages and impairs LSECs through the RAGE/TLR4 signaling pathway. These findings open avenues for discovering novel therapeutic targets for diabetic steatohepatitis.


Asunto(s)
Diabetes Mellitus , Enfermedad del Hígado Graso no Alcohólico , Ratones , Masculino , Humanos , Animales , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Células Endoteliales/metabolismo , Transcriptoma , Ratones Endogámicos C57BL , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Cirrosis Hepática/patología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Dieta Alta en Grasa/efectos adversos
3.
Nat Commun ; 14(1): 167, 2023 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-36690638

RESUMEN

Hepatocellular death increases with hepatic steatosis aggravation, although its regulation remains unclear. Here we show that hepatic steatosis aggravation shifts the hepatocellular death mode from apoptosis to necroptosis, causing increased hepatocellular death. Our results reveal that the transcription factor ATF3 acts as a master regulator in this shift by inducing expression of RIPK3, a regulator of necroptosis. In severe hepatic steatosis, after partial hepatectomy, hepatic ATF3-deficient or -overexpressing mice display decreased or increased RIPK3 expression and necroptosis, respectively. In cultured hepatocytes, ATF3 changes TNFα-dependent cell death mode from apoptosis to necroptosis, as revealed by live-cell imaging. In non-alcoholic steatohepatitis (NASH) mice, hepatic ATF3 deficiency suppresses RIPK3 expression and hepatocellular death. In human NASH, hepatocellular damage is correlated with the frequency of hepatocytes expressing ATF3 or RIPK3, which overlap frequently. ATF3-dependent RIPK3 induction, causing a modal shift of hepatocellular death, can be a therapeutic target for steatosis-induced liver damage, including NASH.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Ratones , Masculino , Humanos , Animales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factores de Transcripción/metabolismo , Necroptosis , Apoptosis , Hepatocitos/metabolismo , Muerte Celular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Transcripción Activador 3/metabolismo
4.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628567

RESUMEN

Advanced glycation end-products (AGEs) and the receptor for AGEs (RAGE) are implicated in inflammatory reactions and vascular complications in diabetes. Signaling pathways downstream of RAGE are involved in NF-κB activation. In this study, we examined whether ethanol extracts of Saururus chinensis (Lour.) Baill. (SE) could affect RAGE signaling and vascular relaxation in streptozotocin (STZ)-induced diabetic rats. Treatment with SE inhibited AGEs-modified bovine serum albumin (AGEs-BSA)-elicited activation of NF-κB and could compete with AGEs-BSA binding to RAGE in a dose-dependent manner. Tumor necrosis factor-α (TNF-α) secretion induced by lipopolysaccharide (LPS)-a RAGE ligand-was also reduced by SE treatment in wild-type Ager+/+ mice as well as in cultured peritoneal macrophages from Ager+/+ mice but not in Ager-/- mice. SE administration significantly ameliorated diabetes-related dysregulation of acetylcholine-mediated vascular relaxation in STZ-induced diabetic rats. These results suggest that SE would inhibit RAGE signaling and would be useful for the improvement of vascular endothelial dysfunction in diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Saururaceae , Animales , Proteínas Portadoras , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Inflamación/tratamiento farmacológico , Ratones , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Ratas , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Saururaceae/metabolismo , Vasodilatación
5.
Int J Mol Sci ; 23(4)2022 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-35216202

RESUMEN

Non-enzymatic glycation is an unavoidable reaction that occurs across biological taxa. The final products of this irreversible reaction are called advanced glycation end-products (AGEs). The endogenously formed AGEs are known to be bioactive and detrimental to human health. Additionally, exogenous food-derived AGEs are debated to contribute to the development of aging and various diseases. Receptor for AGEs (RAGE) is widely known to elicit biological reactions. The binding of RAGE to other ligands (e.g., high mobility group box 1, S100 proteins, lipopolysaccharides, and amyloid-ß) can result in pathological processes via the activation of intracellular RAGE signaling pathways, including inflammation, diabetes, aging, cancer growth, and metastasis. RAGE is now recognized as a pattern-recognition receptor. All mammals have RAGE homologs; however, other vertebrates, such as birds, amphibians, fish, and reptiles, do not have RAGE at the genomic level. This evidence from an evolutionary perspective allows us to understand why mammals require RAGE. In this review, we provide an overview of the scientific knowledge about the role of RAGE in physiological and pathological processes. In particular, we focus on (1) RAGE biology, (2) the role of RAGE in physiological and pathophysiological processes, (3) RAGE isoforms, including full-length membrane-bound RAGE (mRAGE), and the soluble forms of RAGE (sRAGE), which comprise endogenous secretory RAGE (esRAGE) and an ectodomain-shed form of RAGE, and (4) oxytocin transporters in the brain and intestine, which are important for maternal bonding and social behaviors.


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Animales , Humanos , Relaciones Madre-Hijo , Oxitocina/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal/fisiología
6.
Biochem Biophys Res Commun ; 555: 74-80, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33813279

RESUMEN

The engagement of the receptor for advanced glycation end-products (receptor for AGEs, RAGE) with diverse ligands could elicit chronic vascular inflammation, such as atherosclerosis. Binding of cytoplasmic tail RAGE (ctRAGE) to diaphanous-related formin 1 (Diaph1) is known to yield RAGE intracellular signal transduction and subsequent cellular responses. However, the effectiveness of an inhibitor of the ctRAGE/Diaph1 interaction in attenuating the development of atherosclerosis is unclear. In this study, using macrophages from Ager+/+ and Ager-/- mice, we validated the effects of an inhibitor on AGEs-RAGE-induced foam cell formation. The inhibitor significantly suppressed AGEs-RAGE-evoked Rac1 activity, cell invasion, and uptake of oxidized low-density lipoprotein, as well as AGEs-induced NF-κB activation and upregulation of proinflammatory gene expression. Moreover, expression of Il-10, an anti-inflammatory gene, was restored by this antagonist. These findings suggest that the RAGE-Diaph1 inhibitor could be a potential therapeutic drug against RAGE-related diseases, such as chronic inflammation and atherosclerosis.


Asunto(s)
Células Espumosas/metabolismo , Macrófagos Peritoneales/patología , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Expresión Génica , Inflamación/genética , Inflamación/patología , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Neuropéptidos/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Receptor para Productos Finales de Glicación Avanzada/genética , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo
7.
J Diabetes Investig ; 12(1): 35-47, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32515547

RESUMEN

AIMS/INTRODUCTION: Sodium-glucose cotransporter 2 inhibitor (SGLT2i) lowers blood glucose and causes a whole-body energy deficit by boosting renal glucose excretion, thus affecting glucose and energy metabolism. This energy deficit not only decreases bodyweight, but also increases food intake. This food intake increase offsets the SGLT2i-induced bodyweight decrease, but the effect of the food intake increase on the SGLT2i regulation of glucose metabolism remains unclear. MATERIALS AND METHODS: We administered SGLT2i (luseogliflozin) for 4 weeks to hepatic gluconeogenic enzyme gene G6pc reporter mice with/without obesity, which were either fed freely or under a 3-hourly dietary regimen. The effect of feeding condition on the gluconeogenic response to SGLT2i was evaluated by plasma Gaussia luciferase activity, an index of the hepatic gluconeogenic response, in G6pc reporter mice. Energy expenditure was measured by indirect calorimetry. RESULTS: In the lean mice under controlled feeding, SGLT2i decreased bodyweight and plasma glucose, and increased the hepatic gluconeogenic response while decreasing blood insulin. SGLT2i also increased oxygen consumption under controlled feeding. However, free feeding negated all of these effects of SGLT2i. In the obese mice, SGLT2i decreased bodyweight, blood glucose and plasma insulin, ameliorated the upregulated hepatic gluconeogenic response, and increased oxygen consumption under controlled feeding. Under free feeding, although blood glucose was decreased and plasma insulin tended to decrease, the effects of SGLT2i - decreased bodyweight, alleviation of the hepatic gluconeogenic response and increased oxygen consumption - were absent. CONCLUSIONS: Food intake management is crucial for SGLT2i to affect glucose and energy metabolism during type 2 diabetes treatment.


Asunto(s)
Dieta , Metabolismo Energético , Gluconeogénesis , Glucosa/biosíntesis , Obesidad/tratamiento farmacológico , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Delgadez/tratamiento farmacológico , Animales , Diabetes Mellitus Tipo 2/prevención & control , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Obesidad/patología , Delgadez/metabolismo , Delgadez/patología
8.
Glycoconj J ; 38(3): 303-310, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33108607

RESUMEN

The receptor for advanced glycation end-products (receptor for AGEs, RAGE) is a pattern recognition receptor. The interaction of RAGE with its ligands, such as AGEs, S100 proteins, high mobility group box-1 (HMGB1), and lipopolysaccharides (LPS), is known to play a pivotal role in the propagation of immune responses and inflammatory reactions. The ligand-RAGE interaction elicits cellular responses, for example, in myeloid and lymphoid cells, through distinct pathways by activating NF-κB and Rac1/cdc42, which lead to cytokine production, cell migration, phagocytosis, maturation, and polarization. Recently, oxytocin, a peptide hormone and neuropeptide, was identified as a novel binding molecule for the RAGE; however, it cannot compete with the interaction of RAGE with other ligands or induce RAGE intracellular signaling. The RAGE transports oxytocin from the blood into the brain and regulates brain functions. In this review, we summarize the current understanding of glycation reaction, AGEs, and the RAGE-mediated biological responses as well as the physiological role of RAGE in immunity and social behaviors, particularly, maternal bonding.


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Oxitocina/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética
9.
Endocrinology ; 160(12): 2811-2824, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31517956

RESUMEN

Sodium-glucose cotransporter 2 inhibitor (SGLT2i) consistently reduces blood glucose levels in type 2 diabetes mellitus but increases hepatic gluconeogenic gene expression and glucose production, offsetting its glucose-lowering effect. This study aimed to elucidate the effect of SGLT2i on hepatic gluconeogenic response and its mechanism in both insulin-sensitive and insulin-resistant states. A hepatic mouse model was generated to show liver-specific expression of Gaussia luciferase (GLuc) driven by the gluconeogenic enzyme gene G6pc promoter. Hepatic gluconeogenic response was evaluated by measuring plasma GLuc activity. SGLT2i was given to lean and obese mice in single gavage administration or 4-week dietary administration with controlled feeding every 3 hours. In lean mice, single-dose SGLT2i increased plasma GLuc activity from 2 hours after administration, decreasing blood glucose and plasma insulin from 1 to 2 hours after administration. In obese mice, which had higher plasma GLuc activity than lean ones, SGLT2i did not further increase GLuc activity despite decreased blood glucose and plasma insulin. Hepatic Akt and GSK3ß phosphorylation was attenuated by single-dose SGLT2i in lean mice in accordance with the plasma insulin decrease, but not in obese mice. Long-term SGLT2i administration, which increased plasma GLuc activity in lean mice, decreased it in obese mice from 3 weeks after initiation, with increased hepatic Akt and GSK3ß phosphorylation. In conclusion, single SGLT2i administration increases hepatic gluconeogenic response in lean insulin-sensitive mice, but not in obese insulin-resistant mice. Long-term SGLT2i administration relieves obesity-induced upregulation of the hepatic gluconeogenic response by restoring impeded hepatic insulin signaling in obese insulin-resistant mice.


Asunto(s)
Gluconeogénesis/efectos de los fármacos , Resistencia a la Insulina , Obesidad/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/administración & dosificación , Animales , Dieta Alta en Grasa , Glucosa-6-Fosfatasa/genética , Insulina/sangre , Hígado/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico
10.
J Diabetes Investig ; 10(3): 659-666, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30369082

RESUMEN

AIMS/INTRODUCTION: Non-alcoholic steatohepatitis (NASH), which occurs in association with insulin resistance and hepatic fat accumulation, is characterized by chronic liver injury and fibrosis. NASH onset and progression is closely related to hepatic inflammation, which is partly regulated by the vagus nerve through the α7 nicotinic acetylcholine receptor (α7nAchR). Hepatic α7nAchR action is impeded in obesity and insulin resistance. In the present study, using α7nAchR knockout (α7KO) mice, we elucidated the effect of α7nAchR deficiency on NASH-related inflammation and fibrosis. MATERIALS AND METHODS: α7KO mice were fed an atherogenic high-fat diet (AD) for 32 weeks or methionine/choline-deficient diet (MCD) for 6 weeks, both of which induce NASH. Mice were then examined for the degree of NASH-related inflammation and fibrosis by hepatic gene expression analysis and Sirius red histological staining. RESULTS: Hepatic triglyceride accumulation and elevated plasma transaminase levels were observed in both AD and MCD mice, but the plasma transaminase level increase was higher in α7KO mice than in control mice. α7KO mice fed an AD showed significant upregulation of the Col1a1 gene encoding alpha-1 type I collagen, which is involved in liver fibrosis, and the Ccl2 gene encoding C-C motif chemokine ligand 2, a pro-inflammatory chemokine; α7KO mice fed an MCD had significant upregulation of the Col1a1 gene and the Tnf gene, an inflammatory cytokine. Histological analysis showed that AD and MCD exacerbated liver fibrosis in α7KO mice. CONCLUSIONS: The results of this study suggest that α7nAchR deficiency exacerbates hepatic inflammation and fibrosis in a diet-induced mouse model of NASH.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Inflamación/patología , Cirrosis Hepática/patología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Receptores Nicotínicos/fisiología , Animales , Deficiencia de Colina/complicaciones , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Citocinas/metabolismo , Inflamación/etiología , Cirrosis Hepática/etiología , Masculino , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología
11.
Nat Commun ; 9(1): 30, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29296001

RESUMEN

Impaired hepatic glucose uptake (HGU) causes postprandial hyperglycemia in type 2 diabetes. Here, we show that diminished hepatic Sirt2 activity impairs HGU in obese diabetic mice. Hepatic Sirt2 overexpression increases HGU in high-fat diet (HFD)-fed obese diabetic mice and mitigates their impaired glucose tolerance. Hepatic Sirt2 knockdown in non-diabetic mice reduces HGU and causes impaired glucose tolerance. Sirt2 promotes glucose-dependent HGU by deacetylating K126 of glucokinase regulatory protein (GKRP). Glucokinase and GKRP glucose-dependent dissociation is necessary for HGU but is inhibited in hepatocytes derived from obese diabetic mice, depleted of Sirt2 or transfected with GKRP acetylation-mimicking mutants. GKRP deacetylation-mimicking mutants dissociate from glucokinase in a glucose concentration-dependent manner in obese diabetic mouse-derived hepatocytes and increase HGU and glucose tolerance in HFD-induced or db/db obese diabetic mice. We demonstrate that Sirt2-dependent GKRP deacetylation improves impaired HGU and suggest that it may be a therapeutic target for type 2 diabetes.


Asunto(s)
Proteínas Portadoras/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica/fisiología , Glucosa/metabolismo , Hígado/enzimología , Sirtuina 2/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Transporte Biológico , Proteínas Portadoras/genética , Técnicas de Silenciamiento del Gen , Péptidos y Proteínas de Señalización Intracelular , Hígado/metabolismo , Ratones , Ratones Obesos , Mutación , Sirtuina 2/genética
12.
Diabetes ; 66(4): 1008-1021, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28122789

RESUMEN

Platelet-derived growth factor (PDGF) is a key factor in angiogenesis; however, its role in adult obesity remains unclear. In order to clarify its pathophysiological role, we investigated the significance of PDGF receptor ß (PDGFRß) in adipose tissue expansion and glucose metabolism. Mature vessels in the epididymal white adipose tissue (eWAT) were tightly wrapped with pericytes in normal mice. Pericyte desorption from vessels and the subsequent proliferation of endothelial cells were markedly increased in the eWAT of diet-induced obese mice. Analyses with flow cytometry and adipose tissue cultures indicated that PDGF-B caused the detachment of pericytes from vessels in a concentration-dependent manner. M1-macrophages were a major type of cells expressing PDGF-B in obese adipose tissue. In contrast, pericyte detachment was attenuated and vascularity within eWAT was reduced in tamoxifen-inducible conditional Pdgfrb-knockout mice with decreases in adipocyte size and chronic inflammation. Furthermore, Pdgfrb-knockout mice showed enhanced energy expenditure. Consequently, diet-induced obesity and the associated deterioration of glucose metabolism in wild-type mice were absent in Pdgfrb-knockout mice. Therefore, PDGF-B-PDGFRß signaling plays a significant role in the development of adipose tissue neovascularization and appears to be a fundamental target for the prevention of obesity and type 2 diabetes.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Proliferación Celular/genética , Células Endoteliales/citología , Glucosa/metabolismo , Neovascularización Patológica/genética , Obesidad/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Remodelación Vascular/genética , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/irrigación sanguínea , Animales , Western Blotting , Dieta Alta en Grasa , Citometría de Flujo , Técnica de Clampeo de la Glucosa , Macrófagos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Obesidad/metabolismo , Pericitos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
13.
J Nutr ; 147(1): 52-60, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27903831

RESUMEN

BACKGROUND: As the prevalence of nonalcoholic fatty liver disease (NAFLD), including steatosis and nonalcoholic steatohepatitis, is increasing, novel dietary approaches are required for the prevention and treatment of NAFLD. OBJECTIVE: We evaluated the potential of mung bean protein isolate (MuPI) to prevent NAFLD progression. METHODS: In Expts. 1 and 2, the hepatic triglyceride (TG) concentration was compared between 8-wk-old male mice fed a high-fat diet (61% of energy from fat) containing casein, MuPI, and soy protein isolate and an MuPI-constituent amino acid mixture as a source of amino acids (18% of energy) for 4 wk. In Expt. 3, hepatic fatty acid synthase (Fasn) expression was evaluated in 8-wk-old male Fasn-promoter-reporter mice fed a casein- or MuPI-containing high-fat diet for 20 wk. In Expt. 4, hepatic fibrosis was examined in 8-wk-old male mice fed an atherogenic diet (61% of energy from fat, containing 1.3 g cholesterol/100 g diet) containing casein or MuPI (18% of energy) as a protein source for 20 wk. RESULTS: In the high fat-diet mice, the hepatic TG concentration in the MuPI group decreased by 66% and 47% in Expt. 1 compared with the casein group (P < 0.001) and the soy protein isolate group (P = 0.001), respectively, and decreased by 56% in Expt. 2 compared with the casein group (P = 0.011). However, there was no difference between the MuPI-constituent amino acid mixture and casein groups in Expt. 2. In Expt. 3, Fasn-promoter-reporter activity and hepatic TG concentration were lower in the MuPI group than in those fed casein (P < 0.05). In Expt. 4, in mice fed an atherogenic diet, hepatic fibrosis was not induced in the MuPI group, whereas it developed overtly in the casein group. CONCLUSION: MuPI potently reduced hepatic lipid accumulation in mice and may be a potential foodstuff to prevent NAFLD onset and progression.


Asunto(s)
Proteínas en la Dieta/administración & dosificación , Hígado Graso/prevención & control , Inflamación/prevención & control , Cirrosis Hepática/prevención & control , Vigna/química , Animales , Grasas de la Dieta/toxicidad , Proteínas en la Dieta/análisis , Acido Graso Sintasa Tipo I/metabolismo , Hígado Graso/inducido químicamente , Regulación de la Expresión Génica , Inflamación/metabolismo , Cirrosis Hepática/metabolismo , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
14.
Cell Rep ; 14(10): 2362-74, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26947072

RESUMEN

Central insulin action activates hepatic IL-6/STAT3 signaling, which suppresses the gene expression of hepatic gluconeogenic enzymes. The vagus nerve plays an important role in this centrally mediated hepatic response; however, the precise mechanism underlying this brain-liver interaction is unclear. Here, we present our findings that the vagus nerve suppresses hepatic IL-6/STAT3 signaling via α7-nicotinic acetylcholine receptors (α7-nAchR) on Kupffer cells, and that central insulin action activates hepatic IL-6/STAT3 signaling by suppressing vagal activity. Indeed, central insulin-mediated hepatic IL-6/STAT3 activation and gluconeogenic gene suppression were impeded in mice with hepatic vagotomy, pharmacological cholinergic blockade, or α7-nAchR deficiency. In high-fat diet-induced obese and insulin-resistant mice, control of the vagus nerve by central insulin action was disturbed, inducing a persistent increase of inflammatory cytokines. These findings suggest that dysregulation of the α7-nAchR-mediated control of Kupffer cells by central insulin action may affect the pathogenesis of chronic hepatic inflammation in obesity.


Asunto(s)
Insulina/farmacología , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Nervio Vago/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Acetilcolina/metabolismo , Animales , Glucemia/análisis , Proteínas de Unión al Calcio , Células Cultivadas , Clorisondamina/farmacología , Dieta Alta en Grasa , Interleucina-6/sangre , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos del Hígado/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Nicotina/farmacología , Obesidad/metabolismo , Obesidad/patología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Nervio Vago/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/deficiencia , Receptor Nicotínico de Acetilcolina alfa 7/genética
15.
FASEB J ; 30(2): 849-62, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26514166

RESUMEN

ß-Klotho (ß-Kl), a transmembrane protein expressed in the liver, pancreas, adipose tissues, and brain, is essential for feedback suppression of hepatic bile acid synthesis. Because bile acid is a key regulator of lipid and energy metabolism, we hypothesized potential and tissue-specific roles of ß-Kl in regulating plasma lipid levels and body weight. By crossing ß-kl(-/-) mice with newly developed hepatocyte-specific ß-kl transgenic (Tg) mice, we generated mice expressing ß-kl solely in hepatocytes (ß-kl(-/-)/Tg). Gene expression, metabolomic, and in vivo flux analyses consistently revealed that plasma level of cholesterol, which is over-excreted into feces as bile acids in ß-kl(-/-), is maintained in ß-kl(-/-) mice by enhanced de novo cholesterogenesis. No compensatory increase in lipogenesis was observed, despite markedly decreased plasma triglyceride. Along with enhanced bile acid synthesis, these lipid dysregulations in ß-kl(-/-) were completely reversed in ß-kl(-/-)/Tg mice. In contrast, reduced body weight and resistance to diet-induced obesity in ß-kl(-/-) mice were not reversed by hepatocyte-specific restoration of ß-Kl expression. We conclude that ß-Kl in hepatocytes is necessary and sufficient for lipid homeostasis, whereas nonhepatic ß-Kl regulates energy metabolism. We further demonstrate that in a condition with excessive cholesterol disposal, a robust compensatory mechanism maintains cholesterol levels but not triglyceride levels in mice.


Asunto(s)
Peso Corporal/fisiología , Hepatocitos/metabolismo , Metabolismo de los Lípidos/fisiología , Proteínas de la Membrana/metabolismo , Animales , Colesterol/genética , Colesterol/metabolismo , Metabolismo Energético/fisiología , Hepatocitos/citología , Proteínas Klotho , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo
16.
Hepatology ; 61(4): 1343-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25420998

RESUMEN

UNLABELLED: The liver has robust regenerative potential in response to damage, but hepatic steatosis (HS) weakens this potential. We found that the enhanced integrated stress response (ISR) mediated by phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF2α) impairs regeneration in HS and that growth arrest and DNA damage-inducible 34 (Gadd34)-dependent suppression of ISR plays a crucial role in fatty liver regeneration. Although mice fed a high-fat diet for 2 weeks developed moderate fatty liver with no increase in eIF2α phosphorylation before 70% hepatectomy, they showed impaired liver regeneration as a result of reduced proliferation and increased death of hepatocytes with increased phosphorylation of eIF2α and ISR. An increased ISR through Gadd34 knockdown induced C/EBP homologous protein (CHOP)-dependent apoptosis and receptor-interacting protein kinase 3-dependent necrosis, resulting in increased hepatocyte death during fatty liver regeneration. Furthermore, Gadd34 knockdown and increased phosphorylation of eIF2α decreased cyclin D1 protein and reduced hepatocyte proliferation. In contrast, enhancement of Gadd34 suppressed phosphorylation of eIF2α and reduced CHOP expression and hepatocyte apoptosis without affecting hepatocyte proliferation, clearly improving fatty liver regeneration. In more severe fatty liver of leptin receptor-deficient db/db mice, forced expression of hepatic Gadd34 also promoted hepatic regeneration after hepatectomy. CONCLUSION: Gadd34-mediated regulation of ISR acts as a physiological defense mechanism against impaired liver regeneration resulting from steatosis and is thus a possible therapeutic target for impaired regeneration in HS.


Asunto(s)
Hígado Graso , Regeneración Hepática/fisiología , Proteína Fosfatasa 1/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
17.
Diabetes ; 64(2): 459-70, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25249578

RESUMEN

Circadian rhythm is crucial for preventing hepatic insulin resistance, although the mechanism remains uncovered. Here we report that the wake-active hypothalamic orexin system plays a key role in this regulation. Wild-type mice showed that a daily rhythm in blood glucose levels peaked at the awake period; however, the glucose rhythm disappeared in orexin knockout mice despite normal feeding rhythm. Central administration of orexin A during nighttime awake period acutely elevated blood glucose levels but subsequently lowered daytime glucose levels in normal and diabetic db/db mice. The glucose-elevating and -lowering effects of orexin A were suppressed by adrenergic antagonists and hepatic parasympathectomy, respectively. Moreover, the expression levels of hepatic gluconeogenic genes, including Pepck, were increased and decreased by orexin A at nanomolar and femtomolar doses, respectively. These results indicate that orexin can bidirectionally regulate hepatic gluconeogenesis via control of autonomic balance, leading to generation of the daily blood glucose oscillation. Furthermore, during aging, orexin deficiency enhanced endoplasmic reticulum (ER) stress in the liver and caused impairment of hepatic insulin signaling and abnormal gluconeogenic activity in pyruvate tolerance test. Collectively, the daily glucose rhythm under control of orexin appears to be important for maintaining ER homeostasis, thereby preventing insulin resistance in the liver.


Asunto(s)
Sistema Nervioso Autónomo/efectos de los fármacos , Ritmo Circadiano , Hipotálamo/metabolismo , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intracelular/farmacología , Hígado/efectos de los fármacos , Neuropéptidos/farmacología , Animales , Glucemia , Epinefrina/farmacología , Genes Transgénicos Suicidas , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/genética , Neuropéptidos/metabolismo , Neurotransmisores/farmacología , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
18.
Nat Commun ; 5: 4982, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25236782

RESUMEN

In obesity, a paracrine loop between adipocytes and macrophages augments chronic inflammation of adipose tissue, thereby inducing systemic insulin resistance and ectopic lipid accumulation. Obese adipose tissue contains a unique histological structure termed crown-like structure (CLS), where adipocyte-macrophage crosstalk is known to occur in close proximity. Here we show that Macrophage-inducible C-type lectin (Mincle), a pathogen sensor for Mycobacterium tuberculosis, is localized to macrophages in CLS, the number of which correlates with the extent of interstitial fibrosis. Mincle induces obesity-induced adipose tissue fibrosis, thereby leading to steatosis and insulin resistance in liver. We further show that Mincle in macrophages is crucial for CLS formation, expression of fibrosis-related genes and myofibroblast activation. This study indicates that Mincle, when activated by an endogenous ligand released from dying adipocytes, is involved in adipose tissue remodelling, thereby suggesting that sustained interactions between adipocytes and macrophages within CLS could be a therapeutic target for obesity-induced ectopic lipid accumulation.


Asunto(s)
Tejido Adiposo/fisiopatología , Lectinas Tipo C/metabolismo , Macrófagos/citología , Obesidad/metabolismo , Adipocitos/citología , Tejido Adiposo/metabolismo , Animales , Proteínas de Unión al Calcio , Fibrosis , Inflamación/metabolismo , Resistencia a la Insulina , Ligandos , Lípidos/química , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G
19.
Biochem Biophys Res Commun ; 443(1): 333-8, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24315873

RESUMEN

Endoplasmic reticulum (ER) stress suppresses osteoblast differentiation. Activating transcription factor (ATF) 3, a member of the ATF/cAMP response element-binding protein family of transcription factors, is induced by various stimuli including cytokines, hormones, DNA damage, and ER stress. However, the role of ATF3 in osteoblast differentiation has not been elucidated. Treatment with tunicamycin (TM), an ER stress inducer, increased ATF3 expression in the preosteoblast cell line, MC3T3-E1. Overexpression of ATF3 inhibited bone morphogenetic protein 2-stimulated expression and activation of alkaline phosphatase (ALP), an osteogenic marker. In addition, suppression of ALP expression by TM treatment was rescued by silencing of ATF3 using shRNA. Taken together, these data indicate that ATF3 is a novel negative regulator of osteoblast differentiation by specifically suppressing ALP gene expression in preosteoblasts.


Asunto(s)
Factor de Transcripción Activador 3/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/genética , Estrés del Retículo Endoplásmico , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Osteoblastos/citología , Osteogénesis/genética , Factor de Transcripción Activador 3/genética , Animales , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Línea Celular , Humanos , Ratones
20.
Nat Commun ; 4: 1829, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23652017

RESUMEN

The gut microbiota affects nutrient acquisition and energy regulation of the host, and can influence the development of obesity, insulin resistance, and diabetes. During feeding, gut microbes produce short-chain fatty acids, which are important energy sources for the host. Here we show that the short-chain fatty acid receptor GPR43 links the metabolic activity of the gut microbiota with host body energy homoeostasis. We demonstrate that GPR43-deficient mice are obese on a normal diet, whereas mice overexpressing GPR43 specifically in adipose tissue remain lean even when fed a high-fat diet. Raised under germ-free conditions or after treatment with antibiotics, both types of mice have a normal phenotype. We further show that short-chain fatty acid-mediated activation of GPR43 suppresses insulin signalling in adipocytes, which inhibits fat accumulation in adipose tissue and promotes the metabolism of unincorporated lipids and glucose in other tissues. These findings establish GPR43 as a sensor for excessive dietary energy, thereby controlling body energy utilization while maintaining metabolic homoeostasis.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Tracto Gastrointestinal/microbiología , Insulina/metabolismo , Metabolismo de los Lípidos , Microbiota , Receptores Acoplados a Proteínas G/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Metabolismo Energético , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Músculos/metabolismo , Músculos/patología , Obesidad/metabolismo , Obesidad/patología , Especificidad de Órganos , Fosfohidrolasa PTEN/metabolismo , Proteína Quinasa C/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Transducción de Señal , Delgadez/metabolismo , Delgadez/patología , Fosfolipasas de Tipo C/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...