Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
BMC Pediatr ; 24(1): 426, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961351

RESUMEN

BACKGROUND: Adipose tissue is significantly involved in inflammatory bowel disease (IBD). Vitamin D can affect both adipogenesis and inflammation. The aim of this study was to compare the production of selected adipokines, potentially involved in the pathogenesis of IBD - adiponectin, resistin, retinol binding protein 4 (RBP-4), adipocyte fatty acid binding protein and nesfatin-1 in children with IBD according to the presence of 25-hydroxyvitamin D (25(OH)D) deficiency. METHODS: The study was conducted as a case-control study in pediatric patients with IBD and healthy children of the same sex and age. In addition to adipokines and 25(OH)D, anthropometric parameters, markers of inflammation and disease activity were assessed in all participants. RESULTS: Children with IBD had significantly higher resistin levels regardless of 25(OH)D levels. IBD patients with 25(OH)D deficiency only had significantly lower RBP-4 compared to healthy controls and also compared to IBD patients without 25(OH)D deficiency. No other significant differences in adipokines were found in children with IBD with or without 25(OH)D deficiency. 25(OH)D levels in IBD patients corelated with RBP-4 only, and did not correlate with other adipokines. CONCLUSIONS: Whether the lower RBP-4 levels in the 25(OH)D-deficient group of IBD patients directly reflect vitamin D deficiency remains uncertain. The production of other adipokines does not appear to be directly related to vitamin D deficiency.


Asunto(s)
Adipoquinas , Deficiencia de Vitamina D , Vitamina D , Humanos , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/sangre , Masculino , Femenino , Niño , Estudios de Casos y Controles , Adipoquinas/sangre , Adolescente , Vitamina D/sangre , Vitamina D/análogos & derivados , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Proteínas Plasmáticas de Unión al Retinol/análisis , Resistina/sangre , Nucleobindinas/sangre , Adiponectina/sangre , Adiponectina/deficiencia , Proteínas de Unión al Calcio/sangre , Proteínas de Unión a Ácidos Grasos/sangre , Proteínas de Unión al ADN/sangre , Biomarcadores/sangre , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/complicaciones
2.
Circ Res ; 128(1): 136-149, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33411633

RESUMEN

The landmark discoveries of leptin and adiponectin firmly established adipose tissue as a sophisticated and highly active endocrine organ, opening a new era of investigating adipose-mediated tissue crosstalk. Both obesity-associated hyperleptinemia and hypoadiponectinemia are important biomarkers to predict cardiovascular outcomes, suggesting a crucial role for adiponectin and leptin in obesity-associated cardiovascular disorders. Normal physiological levels of adiponectin and leptin are indeed essential to maintain proper cardiovascular function. Insufficient adiponectin and leptin signaling results in cardiovascular dysfunction. However, a paradox of high levels of both leptin and adiponectin is emerging in the pathogenesis of cardiovascular disorders. Here, we (1) summarize the recent progress in the field of adiponectin and leptin and its association with cardiovascular disorders, (2) further discuss the underlying mechanisms for this new paradox of leptin and adiponectin action, and (3) explore the possible application of partial leptin reduction, in addition to increasing the adiponectin/leptin ratio as a means to prevent or reverse cardiovascular disorders.


Asunto(s)
Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Adiponectina/deficiencia , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Animales , Fármacos Antiobesidad/uso terapéutico , Cirugía Bariátrica , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/terapia , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/patología , Sistema Cardiovascular/fisiopatología , Humanos , Errores Innatos del Metabolismo/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Obesidad/terapia , Transducción de Señal
3.
J Pathol ; 257(2): 146-157, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35072951

RESUMEN

Obesity is one of the major risk factors for cancer. Clinical studies have demonstrated that circulating levels of adiponectin are inversely correlated, not only with the extent of adiposity, but also with the incidence of several types of cancer, particularly endometrial cancer (EC). However, thus far, adiponectin remains a correlative factor, without definitive evidence to show a causal effect in EC and the potential mechanism(s) involved. To address this issue, we introduced an Apn-null mutation into Pten haploid-deficient (Pten+/- ) mice. The Pten heterozygous mutation alone led to the development of EC in less than 30% of female mice; however, when combined with the Apn-null mutation, the incidence of endometrial lesions rose to at least two-thirds. Although Apn deficiency did not further potentiate the Akt activation caused by the Pten mutation, it elevated the phosphorylation of mitogen-activated protein kinase (MAPK) p42/44, indicating activation of the MAPK signaling pathway. Treatment of Apn-/- ;Pten+/- mice with a MEK inhibitor blocked the development of EC. Finally, xenografts of a PTEN-proficient human EC cell line grew faster in Apn-deficient mice, whereas an adiponectin receptor agonist reduced xenograft growth of a PTEN-deficient human EC cell line. Thus, reduction of adiponectin activity promotes EC development, at least in the context of Pten mutation, by activating MAPK. © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Endometriales , Errores Innatos del Metabolismo , Adiponectina/deficiencia , Adiponectina/genética , Adiponectina/farmacología , Animales , Neoplasias Endometriales/patología , Femenino , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
4.
Diabetologia ; 65(4): 733-747, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35091821

RESUMEN

AIMS/HYPOTHESIS: Obesity and hepatic steatosis are risk factors for gestational diabetes mellitus (GDM), a common complication of pregnancy. Adiponectin is a fat-derived hormone that improves hepatic steatosis and insulin sensitivity. Low levels of circulating adiponectin are associated with GDM development. We hypothesised that adiponectin deficiency causes fatty liver during pregnancy, contributing to the development of GDM. METHODS: To determine the role of adiponectin in fatty liver development during pregnancy, we compared pregnant (third week of pregnancy) adiponectin knockout (KO) mice (strain B6;129-Adipoqtm1Chan/J) with wild-type mice and assessed several variables of hepatic lipid metabolism and glucose homeostasis. The impact of adiponectin supplementation was measured by administering adenovirus-mediated full-length adiponectin at the end of the second week of pregnancy and comparing with green fluorescent protein control. RESULTS: In the third week of pregnancy, fasted pregnant adiponectin KO mice were hyperglycaemic on a low-fat diet (9.2 mmol/l vs 7.7 mmol/l in controls, p<0.05) and were glucose and pyruvate intolerant relative to wild-type mice. Pregnant adiponectin KO mice developed hepatic steatosis and a threefold elevation in hepatic triacylglycerols (p<0.05) relative to wild-type mice. Gestational weight gain and food consumption were similar in KO and wild-type mice. Adenoviral-mediated adiponectin supplementation to pregnant adiponectin KO mice improved glucose tolerance, prevented fasting hyperglycaemia and attenuated fatty liver development. CONCLUSIONS/INTERPRETATION: Adiponectin deficiency increased hepatic lipid accumulation during the period of pregnancy associated with increased fat utilisation. Consequently, adiponectin deficiency contributed to glucose intolerance, dysregulated gluconeogenesis and hyperglycaemia, all of which are characteristic of GDM. Increasing adiponectin in the last week of pregnancy alleviated hepatic steatosis and restored normal glucose homeostasis during pregnancy.


Asunto(s)
Diabetes Gestacional , Hígado Graso , Hiperglucemia , Resistencia a la Insulina , Adiponectina/deficiencia , Adiponectina/metabolismo , Animales , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Hígado Graso/metabolismo , Femenino , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Hígado/metabolismo , Errores Innatos del Metabolismo , Ratones , Ratones Noqueados , Embarazo
5.
J Nucl Cardiol ; 29(3): 1405-1414, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33501546

RESUMEN

BACKGROUND: The role of adipose tissue (AT) in arterial inflammation in familial dyslipidaemias is poorly studied. We investigated the relationship between AT and arterial inflammation in patients with heterozygous familial hypercholesterolemia (heFH) and familial combined hyperlipidemia (FCH). METHODS AND RESULTS: A total of 40 patients (20 heFH/20 FCH) and a subgroup of 20 of non-heFH/FCH patients were enrolled. Participants underwent blood sampling for serum adipokine measurements and Fluorine-18 fluorodeoxyglucose (18F-FDG) PET/CT imaging. Abdominal visceral (VAT) and subcutaneous (SAT) AT volumes and AT and abdominal aorta 18F-FDG uptake were quantified. FCH patients had increased VAT (pANOVA = 0.004) and SAT volumes (pANOVA = 0.003), lower VAT metabolic activity (pANOVA = 0.0047), and lower adiponectin levels (pANOVA = 0.007) compared to heFH or the control group. Log(Serum adiponectin) levels were correlated with aortic TBR (b = - 0.118, P = 0.038). In mediation analysis, VAT volume was the major determinant of circulating adiponectin, an effect partly mediated via VAT TBR. Clustering of the population of heFH/FCH by VAT volume/TBR and serum adiponectin identified two distinct patient clusters with significant differences in aortic TBR levels (2.11 ± 0.06 vs 1.89 ± 0.05, P= 0.012). CONCLUSIONS: VAT phenotype (increased VAT volume and/or high VAT TBR) and hypoadiponectinemia may account for the observed differences in arterial inflammation levels between heFH and FCH patients.


Asunto(s)
Arteritis , Dislipidemias , Adiponectina/deficiencia , Adiponectina/metabolismo , Tejido Adiposo , Dislipidemias/diagnóstico por imagen , Dislipidemias/metabolismo , Radioisótopos de Flúor , Fluorodesoxiglucosa F18/metabolismo , Humanos , Grasa Intraabdominal/diagnóstico por imagen , Grasa Intraabdominal/metabolismo , Errores Innatos del Metabolismo , Fenotipo , Tomografía Computarizada por Tomografía de Emisión de Positrones
6.
Vascular ; 30(2): 384-391, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33866881

RESUMEN

OBJECTIVES: Albuminuria and serum adiponectin levels are factors that have been associated with the development of cardiovascular disease in patients with diabetes mellitus. Here we investigated the relationship between serum adiponectin levels and aortic stiffness in nondialysis diabetic kidney disease patients with stage 3-5 chronic kidney disease. METHODS: Fasting blood samples were obtained from 80 nondialysis diabetic kidney disease patients with stage 3-5 chronic kidney disease. Carotid-femoral pulse wave velocity (cfPWV) was measured using applanation tonometry; cfPWV values of >10 m/s were defined as aortic stiffness. Serum adiponectin levels were determined by enzyme immunoassay. RESULTS: Forty-two patients (52.5%) with nondialysis diabetic kidney disease were diagnosed with aortic stiffness. The patients in this group were older (p = 0.011), had higher systolic blood pressure (p = 0.002) and urine albumin-to-creatinine ratios (p = 0.013), included fewer females (p = 0.024), and had lower serum adiponectin (p = 0.001) levels than those in the control group. Multivariable logistic regression analysis revealed that serum adiponectin was independently associated with aortic stiffness (odds ratio = 0.930, 95% confidence interval: 0.884-0.978, p = 0.005) and also positively correlated with cfPWV values by multivariable linear regression (ß = -0.309, p = 0.002) in nondialysis diabetic kidney disease patients. CONCLUSIONS: The results suggested that serum adiponectin levels could be used to predict aortic stiffness in nondialysis diabetic kidney disease patients with stage 3-5 chronic kidney disease.


Asunto(s)
Diabetes Mellitus , Fallo Renal Crónico , Rigidez Vascular , Adiponectina/deficiencia , Femenino , Humanos , Masculino , Errores Innatos del Metabolismo , Análisis de la Onda del Pulso
7.
Int J Mol Sci ; 23(9)2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35563332

RESUMEN

Adiponectin administration to pregnant mice decreases nutrient transport and fetal growth. An adiponectin deficiency, on the other hand, as seen in obese women during pregnancy, alters fetal growth; however, the mechanism is unclear. To determine the role of adiponectin on placenta function and fetal growth, we used adiponectin knockout, adiponectin heterozygote that displays reduced adiponectin levels, and wild-type mice on a control diet or high fat/high sucrose (HF/HS) diet. Triglycerides (TGs) in the serum, liver, and placenta were measured using colorimetric assays. Gene expression was measured using quantitative RT-PCR. Adiponectin levels did not affect fetal weight, but it reduced adiponectin levels, increased fetal serum and placenta TG content. Wildtype dams on a HF/HS diet protected the fetuses from fatty acid overload as judged by increased liver TGs in dams and normal serum and liver TG levels in fetuses, while low adiponectin was associated with increased fetal liver TGs. Low maternal adiponectin increased the expression of genes involved in fatty acid transport; Lpl and Cd36 in the placenta. Adiponectin deficiency does not affect fetal growth but induces placental dysfunction and increases fetal TG load, which is enhanced with obesity. This could lead to imprinting effects on the fetus and the development of metabolic dysfunction in the offspring.


Asunto(s)
Adiponectina , Placenta , Adiponectina/deficiencia , Adiponectina/metabolismo , Animales , Ácidos Grasos/metabolismo , Femenino , Desarrollo Fetal , Errores Innatos del Metabolismo , Ratones , Obesidad/metabolismo , Placenta/metabolismo , Embarazo
8.
FASEB J ; 33(12): 13617-13631, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31585050

RESUMEN

Adiponectin (APN), an adipocyte-derived adipokine, has been shown to limit lung injury originating from endothelial cell (EC) damage. Previously we reported that obese mice with low circulatory APN levels exhibited pulmonary vascular endothelial dysfunction. This study was designed to investigate the cellular and molecular mechanisms underlying the pulmonary endothelium-dependent protective effects of APN. Our results demonstrated that in APN-/- mice, there was an inherent state of endothelium mitochondrial dysfunction that could contribute to endothelial activation and increased susceptibility to LPS-induced acute lung injury (ALI). We noted that APN-/- mice showed decreased expression of mitochondrial biogenesis regulatory protein peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) and its downstream proteins nuclear respiratory factor 1, transcription factor A, mitochondrial, and Sirtuin (Sirt)3 and Sirt1 expression in whole lungs and in freshly isolated lung ECs from these mice at baseline and subjected to LPS-induced ALI. We further showed that treating APN-/- mice with PGC-1α activator pyrroloquinoline quinone enhances mitochondrial biogenesis and function in lung endothelium and attenuation of ALI. These results suggest that the pulmonary endothelium-protective properties of APN are mediated, at least in part, by an enhancement of mitochondrial biogenesis through a mechanism involving PGC-1α activation.-Shah, D., Torres, C., Bhandari, V. Adiponectin deficiency induces mitochondrial dysfunction and promotes endothelial activation and pulmonary vascular injury.


Asunto(s)
Adiponectina/deficiencia , Endotelio Vascular/patología , Inflamasomas , Lesión Pulmonar/etiología , Errores Innatos del Metabolismo/complicaciones , Mitocondrias/patología , Lesiones del Sistema Vascular/etiología , Adiponectina/fisiología , Animales , Endotelio Vascular/metabolismo , Lipopolisacáridos/toxicidad , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Biogénesis de Organelos , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
9.
Epilepsy Behav ; 106: 107043, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32217418

RESUMEN

Epilepsy, a common neurologic condition, is associated with a greater prevalence of type 2 diabetes mellitus (T2DM). We examined potential drivers for the comorbidity of epilepsy and T2DM in an attempt to elucidate possible biological mechanisms underlying the development of processes in individuals. We searched PubMed and Medline up to December 2019. Our search yielded 3361 articles, of which 82 were included in the scoping review. We reviewed articles focusing on the association of epilepsy and T2DM, drivers, and biological mechanisms. We found that epilepsy is associated with obesity and obesity is associated with T2DM. Treatment with valproate (either sodium or acid) is associated with weight increase and hyperinsulinemia, while topiramate causes weight loss. People with epilepsy are less likely to exercise, which is protective against obesity. Mitochondrial dysfunction and adiponectin deficiency are common to epilepsy and T2DM. One possible mechanism for the comorbidity is mitochondrial dysfunction and adiponectin deficiency, which promotes epilepsy, obesity, and T2DM. Another possible mechanism is that people with epilepsy are more likely to be obese because of the lack of exercise and the effects of some antiseizure medications (ASMs), which makes them susceptible to T2DM because of the development of mitochondrial dysfunction and adiponectin deficiency. A third mechanism is that people with epilepsy have greater mitochondrial dysfunction and lower adiponectin levels than people without epilepsy at baseline, which may exacerbate after treatment with ASMs. Future research involving a combined genetic and molecular pathway approach will likely yield valuable insight regarding the comorbidity of epilepsy and T2DM.


Asunto(s)
Anticonvulsivantes/efectos adversos , Diabetes Mellitus Tipo 2/epidemiología , Epilepsia/epidemiología , Obesidad/epidemiología , Adiponectina/sangre , Adiponectina/deficiencia , Comorbilidad , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/inducido químicamente , Epilepsia/sangre , Epilepsia/tratamiento farmacológico , Femenino , Humanos , Masculino , Errores Innatos del Metabolismo/sangre , Errores Innatos del Metabolismo/epidemiología , Mitocondrias/metabolismo , Obesidad/sangre , Aumento de Peso/fisiología
10.
Cell Biochem Funct ; 38(1): 106-117, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31746004

RESUMEN

Preeclampsia is not fully understood; and few biomarkers, therapeutic targets, and therapeutic agents for its management have been identified. Original investigative findings suggest that abnormal placentation triggers preeclampsia and leads to hypertension, proteinuria, endothelial dysfunction, and inflammation, which are characteristics of the disease. Because of the regulatory roles that it plays in several metabolic processes, adiponectin has become a cytokine of interest in metabolic medicine. In this review, we have discussed the role of adiponectin in trophoblast proliferation, trophoblast differentiation, trophoblast invasion of the decidua, and decidual angiogenesis, which are the major phases of placentation. Also, we have highlighted the physiological profile of adiponectin in the course of normal pregnancy. Moreover, we have discussed the involvement of adiponectin in hypertension, endothelial dysfunction, inflammation, and proteinuria. Furthermore, we have summarized the reported relationship between the maternal serum adiponectin level and preeclampsia. The available evidence indicates that adiponectin level physiologically falls as pregnancy advances, regulates placentation, and exhibits protective effects against the symptoms of preeclampsia and that while hyperadiponectinemia is evident in normal-weight preeclamptic women, hypoadiponectinemia is evident in overweight and obese preeclamptic women. Therefore, the clinical use of adiponectin as a biomarker, therapeutic target, or therapeutic agent against the disease looks promising and should be considered.


Asunto(s)
Adiponectina/metabolismo , Placentación , Preeclampsia/metabolismo , Adiponectina/deficiencia , Femenino , Humanos , Errores Innatos del Metabolismo/metabolismo , Embarazo
11.
J Cell Physiol ; 234(5): 7062-7069, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30479003

RESUMEN

Central adiponectin (APN) in either the globular (gAPN) or full-length forms decreases sympathetic tone and increases trabecular bone mass in mice through the hypothalamus. It is known that cannabinoid type-1 (CB1) receptors are expressed in the hypothalamic ventromedial nucleus and participate in energy metabolism by controlling sympathetic activity. However, whether central APN could influence endocannabinoid signaling through CB1 receptor to regulate bone metabolism has not been characterized. Here we demonstrate that gAPN downregulated CB1 expression in embryonic mouse hypothalamus N1 cells in vitro. gAPN intracerebroventricular (icv) infusions also decreased hypothalamic CB1 expression and bone formation parameters in APN-knockout (APN-KO) and wild-type mice. Most importantly, mice pretreated with icv infusions with the CB1 receptor agonist arachidonyl-2'-chloroethylamine or antagonist rimonabant attenuated or enhanced respectively central APN induction of bone formation. We then investigated whether epigenetic signaling mechanisms were involved in the downregulation of hypothalamic CB1 expression by gAPN. We found gAPN enhanced expression levels of various histone deacetylases (HDACs), especially HDAC5. Furthermore, chromatin immunoprecipitation assays revealed HDAC5 bound to the transcriptional start site transcription start site 2 region of the CB1 promoter. In summary, our study identified a possible novel central APN-HDAC5-CB1 signaling mechanism that promotes peripheral bone formation through epigenetic regulation of hypothalamic CB1 expression.


Asunto(s)
Adiponectina/administración & dosificación , Adiponectina/metabolismo , Remodelación Ósea/efectos de los fármacos , Hueso Esponjoso/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Fémur/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Adiponectina/deficiencia , Adiponectina/genética , Animales , Sitios de Unión , Hueso Esponjoso/metabolismo , Células Cultivadas , Regulación hacia Abajo , Fémur/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Hipotálamo/metabolismo , Infusiones Intraventriculares , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Receptor Cannabinoide CB1/genética
12.
Diabetologia ; 61(4): 932-941, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29224189

RESUMEN

AIM/HYPOTHESIS: Adiponectin (APN), a circulating hormone secreted by mature adipocytes, has been extensively studied because it has beneficial metabolic effects. While many studies have focused on the congenital loss of APN and its effects on systemic body glucose and lipid metabolism, little is known about the effects triggered by acute loss of APN in the adult mouse. We anticipated that genetically induced acute depletion of APN in adult mice would have a more profound effect on systemic metabolic health than congenital deletion of Adipoq, the gene encoding APN, with its associated potential for adaptive responses that may mask the phenotypes. METHODS: Mice carrying loxP-flanked regions of Adipoq were generated and bred to the Adipoq (APN) promoter-driven reverse tetracycline-controlled transactivator (rtTA) (APN-rtTA) gene and a tet-responsive Cre line (TRE-Cre) to achieve acute depletion of APN. Upon acute removal of APN in adult mice, systemic glucose and lipid homeostasis were assessed under basal and insulinopenic conditions. RESULTS: The acute depletion of APN results in more severe systemic insulin resistance and hyperlipidaemia than in mice with congenital loss of APN. Furthermore, the acute depletion of APN in adult mice results in a much more dramatic reduction in survival rate, with 50% of inducible knockouts dying in the first 5 days under insulinopenic conditions compared with 0% of congenital Adipoq knockout mice under similar conditions. CONCLUSIONS/INTERPRETATION: Acute systemic removal of APN results in a much more negative metabolic phenotype compared with congenital knockout of Adipoq. Specifically, our data demonstrate that acute depletion of APN is especially detrimental to lipid homeostasis, both under basal and insulinopenic conditions. This suggests that compensatory mechanisms exist in congenital knockout mice that offset some of the metabolic actions covered by APN.


Asunto(s)
Adiponectina/deficiencia , Tejido Adiposo/fisiopatología , Adipocitos/metabolismo , Adiponectina/genética , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Homeostasis , Hiperlipidemias/fisiopatología , Inflamación , Insulina/metabolismo , Resistencia a la Insulina , Lipasa/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Fenotipo , Pioglitazona/química , Regiones Promotoras Genéticas , Factores de Tiempo
13.
Circulation ; 136(22): 2162-2177, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-28978553

RESUMEN

BACKGROUND: Cell therapy remains the most promising approach against ischemic heart injury. However, the poor survival of engrafted stem cells in the ischemic environment limits their therapeutic efficacy for cardiac repair after myocardial infarction. CTRP9 (C1q/tumor necrosis factor-related protein-9) is a novel prosurvival cardiokine with significantly downregulated expression after myocardial infarction. Here we tested a hypothesis that CTRP9 might be a cardiokine required for a healthy microenvironment promoting implanted stem cell survival and cardioprotection. METHODS: Mice were subjected to myocardial infarction and treated with adipose-derived mesenchymal stem cells (ADSCs, intramyocardial transplantation), CTRP9, or their combination. Survival, cardiac remodeling and function, cardiomyocytes apoptosis, and ADSCs engraftment were evaluated. Whether CTRP9 directly regulates ADSCs function was determined in vitro. Discovery-drive approaches followed by cause-effect analysis were used to uncover the molecular mechanisms of CTRP9. RESULTS: Administration of ADSCs alone failed to exert significant cardioprotection. However, administration of ADSCs in addition to CTRP9 further enhanced the cardioprotective effect of CTRP9 (P<0.05 or P<0.01 versus CTRP9 alone), suggesting a synergistic effect. Administration of CTRP9 at a dose recovering physiological CTRP9 levels significantly prolonged ADSCs retention/survival after implantation. Conversely, the number of engrafted ADSCs was significantly reduced in the CTRP9 knockout heart. In vitro study demonstrated that CTRP9 promoted ADSCs proliferation and migration, and it protected ADSCs against hydrogen peroxide-induced cellular death. CTRP9 enhances ADSCs proliferation/migration by extracellular regulated protein kinases (ERK)1/2-matrix metallopeptidase 9 signaling and promotes antiapoptotic/cell survival via ERK-nuclear factor erythroid-derived 2-like 2/antioxidative protein expression. N-cadherin was identified as a novel CTRP9 receptor mediating ADSCs signaling. Blockade of either N-cadherin or ERK1/2 completely abolished the previously noted CTRP9 effects. Although CTRP9 failed to promote ADSCs cardiogenic differentiation, CTRP9 promotes superoxide dismutase 3 expression and secretion from ADSCs, protecting cardiomyocytes against oxidative stress-induced cell death. CONCLUSIONS: We provide the first evidence that CTRP9 promotes ADSCs proliferation/survival, stimulates ADSCs migration, and attenuates cardiomyocyte cell death by previously unrecognized signaling mechanisms. These include binding with N-cadherin, activation of ERK-matrix metallopeptidase 9 and ERK-nuclear factor erythroid-derived 2-like 2 signaling, and upregulation/secretion of antioxidative proteins. These results suggest that CTRP9 is a cardiokine critical in maintaining a healthy microenvironment facilitating stem cell engraftment in infarcted myocardial tissue, thereby enhancing stem cell therapeutic efficacy.


Asunto(s)
Adiponectina/metabolismo , Glicoproteínas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/metabolismo , Regeneración , Transducción de Señal , Adiponectina/administración & dosificación , Adiponectina/deficiencia , Adiponectina/genética , Tejido Adiposo/citología , Animales , Apoptosis , Cadherinas/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Glicoproteínas/administración & dosificación , Glicoproteínas/deficiencia , Glicoproteínas/genética , Proteínas Fluorescentes Verdes/genética , Peróxido de Hidrógeno/toxicidad , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Factor 2 Relacionado con NF-E2/metabolismo , Fenotipo , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre , Superóxido Dismutasa/metabolismo , Factores de Tiempo
14.
Am J Pathol ; 187(7): 1577-1585, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28463682

RESUMEN

Adiponectin (ApN) is a hormone that exhibits anti-inflammatory effects on skeletal muscle exposed to acute and chronic inflammation. We have previously tested the implication of ApN in Duchenne muscular dystrophy (DMD) using mdx mice, a model of DMD, and by generating transgenic mdx mice overexpressing ApN. We showed that ApN can act as a preventive agent and delay disease progression by reducing muscle inflammation/injury and improving force/myogenesis. Herein, we took an opposite approach and crossed mdx mice with ApN knockout mice, to obtain mdx mice with ApN depletion. The aims were to test whether ApN deficiency could worsen the mdx phenotype and whether ApN supplementation can reverse several muscle abnormalities once the disease is settled. mdx-knockout mice exhibited lower muscle force/endurance as well as increased muscle damage when compared to regular mdx mice. Local administration of the ApN gene significantly reduced the expression of several oxidative stress/inflammatory markers and increased the expression of myogenic markers in the skeletal muscle. Finally, the presence of ApN markedly reduced the activity of NF-κB, a key player in muscle inflammation and myogenesis. ApN proves to be a powerful protector of the skeletal muscle capable of reversing the disease progression, thus making it a potential therapeutic agent for DMD.


Asunto(s)
Adiponectina/deficiencia , Errores Innatos del Metabolismo/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Adiponectina/administración & dosificación , Adiponectina/genética , Adiponectina/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/prevención & control , Masculino , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/fisiopatología , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , FN-kappa B/inmunología
15.
Cardiovasc Diabetol ; 17(1): 133, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30301460

RESUMEN

BACKGROUND: Gestational diabetes (GDM) and milder gestational impaired glucose tolerance (GIGT) identify women at risk of developing type 2 diabetes and cardiovascular disease later in life. Accordingly, the postpartum years after gestational dysglycemia can provide insight into early events in the natural history of these disorders. We thus sought to prospectively evaluate the relationship between gestational glucose tolerance and emerging cardiometabolic biomarkers [adiponectin, chemerin, retinol-binding protein-4 (RBP-4), C-reactive protein (CRP), plasminogen activator inhibitor-1 (PAI-1)] at both 1- and 3-years postpartum in a cohort reflecting the full spectrum of gestational dysglycemia (from normal to GIGT to GDM). METHODS: Three-hundred-and-thirty-nine women completed a glucose challenge test (GCT) and oral glucose tolerance test (OGTT) in pregnancy, which identified 4 gestational glucose tolerance groups: GDM (n = 105); GIGT (n = 59); abnormal GCT with normal OGTT (n = 99); and normal GCT with normal OGTT (n = 76). At 1- and 3-years postpartum, the women underwent repeat OGTT with measurement of biomarkers (adiponectin/chemerin/RBP-4/CRP/PAI-1). RESULTS: Serum adiponectin was lower in women with GDM and GIGT at both 1-year and 3-years (both P ≤ 0.002), whereas chemerin, RBP-4, CRP and PAI-1 showed no differences across the 4 groups. Importantly, the change in PAI-1 between 1- and 3-years progressively increased from the normal GCT group to the abnormal GCT group to GIGT to GDM (P = 0.03). Indeed, both GDM (t = 2.98, P = 0.003) and GIGT (t = 2.14, P = 0.03) independently predicted an increase in PAI-1 from 1- to 3-years postpartum. CONCLUSIONS: Hypoadiponectinemia and rising PAI-1 over time are early features of the cardiometabolic biomarker profile of women with recent gestational dysglycemia.


Asunto(s)
Adiponectina/deficiencia , Glucemia/metabolismo , Diabetes Gestacional/sangre , Errores Innatos del Metabolismo/sangre , Inhibidor 1 de Activador Plasminogénico/sangre , Adiponectina/sangre , Adulto , Biomarcadores/sangre , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/etiología , Diabetes Gestacional/diagnóstico , Regulación hacia Abajo , Femenino , Humanos , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/etiología , Embarazo , Estudios Prospectivos , Factores de Riesgo , Factores de Tiempo , Regulación hacia Arriba
16.
Stem Cells ; 35(7): 1835-1848, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28480607

RESUMEN

Myelotoxic injury, such as chemotherapeutic agents and ionizing radiation, unlocks the vigorous power of hematopoietic stem cells (HSCs) to replenish the hematopoietic system, making quiescent HSCs enter the cell cycle. Considering that both HSC-intrinsic and -extrinsic mechanisms enforce quiescence of HSCs, the drastic change in bone marrow (BM) environment after injury, represented by massive expansion of BM adipocytes, might trigger HSC activation. BM adipocytes, the major cellular component in the ablated marrow, however, reportedly suppress proliferation of hematopoietic cells, which may indicate the BM adipocytogenesis is an irrational response of injured organism. Given that adipose tissue is an endocrine organ with pleiotropic functions, we hypothesized that adipocyte-derived factors, especially adiponectin, an anti-inflammatory adipokine involved in regulation of granulopoiesis, are implicated in HSC activation. Myeloablative intervention increased BM adiponectin by multiple mechanisms, including adipocyte expansion and increased diffusion from the blood. Adiponectin-null (Adipoq -/- ) mice showed delayed hematopoietic recovery after BM injury, with Adipoq-/- HSCs more quiescent and defective in mammalian target of rapamycin complex 1 (mTORC1) activation. Recombinant adiponectin promoted not only HSC activation in vivo but cytokine-induced activation in vitro, and shortened the time for exit from quiescence in an mTORC1-dependent manner. These data illustrate a scarcely-reported example of a cell-extrinsic factor, adiponectin, enhancing quiescence exit of HSCs, and subsequent hematopoietic recovery. Our findings also highlight adipocytes as a source of adiponectin to ensure the proliferative burst of hematopoietic cells in ablated marrow. Stem Cells 2017;35:1835-1848.


Asunto(s)
Adiponectina/genética , Médula Ósea/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Adiponectina/deficiencia , Animales , Compuestos de Bencidrilo/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Ciclofosfamida/farmacología , Citarabina/farmacología , Compuestos Epoxi/farmacología , Fluorouracilo/farmacología , Regulación de la Expresión Génica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Agonistas Mieloablativos/farmacología , Poli I-C/farmacología , Transducción de Señal , Sirolimus/farmacología , Irradiación Corporal Total
17.
Mol Psychiatry ; 22(7): 1044-1055, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27137743

RESUMEN

Post-traumatic stress disorder (PTSD) is characterized by exaggerated fear expression and impaired fear extinction. The underlying molecular and cellular mechanisms of PTSD are largely unknown. The current pharmacological and non-pharmacological treatments for PTSD are either ineffective or temporary with high relapse rates. Here we report that adiponectin-deficient mice exhibited normal contextual fear conditioning but displayed slower extinction learning. Infusions of adiponectin into the dentate gyrus (DG) of the hippocampus in fear-conditioned mice facilitated extinction of contextual fear. Whole-cell patch-clamp recordings in brain slices revealed that intrinsic excitability of DG granule neurons was enhanced by adiponectin deficiency and suppressed after treatment with the adiponectin mimetic AdipoRon, which were associated with increased input resistance and hyperpolarized resting membrane potential, respectively. Moreover, deletion of AdipoR2, but not AdipoR1 in the DG, resulted in augmented fear expression and reduced extinction, accompanied by intrinsic hyperexcitability of DG granule neurons. Adiponectin and AdipoRon failed to induce facilitation of fear extinction and elicit inhibition of intrinsic excitability of DG neurons in AdipoR2 knockout mice. These results indicated that adiponectin action via AdipoR2 was both necessary and sufficient for extinction of contextual fear and intrinsic excitability of DG granule neurons, implying that enhancing or dampening DG neuronal excitability may cause resistance to or facilitation of extinction. Therefore, our findings provide a functional link between adiponectin/AdipoR2 activation, DG neuronal excitability and contextual fear extinction, and suggest that targeting adiponectin/AdipoR2 may be used to strengthen extinction-based exposure therapies for PTSD.


Asunto(s)
Adiponectina/metabolismo , Giro Dentado/metabolismo , Receptores de Adiponectina/metabolismo , Potenciales de Acción/fisiología , Adiponectina/deficiencia , Adiponectina/genética , Animales , Condicionamiento Clásico/fisiología , Modelos Animales de Enfermedad , Extinción Psicológica/fisiología , Miedo/fisiología , Femenino , Hipocampo , Aprendizaje/fisiología , Sistema Límbico , Masculino , Potenciales de la Membrana , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Técnicas de Placa-Clamp , Corteza Prefrontal/fisiología , Trastornos por Estrés Postraumático/metabolismo
18.
Br J Anaesth ; 120(6): 1356-1367, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29793601

RESUMEN

BACKGROUND: Adiponectin, a cytokine secreted by adipocytes, plays an important role in regulating glucose and lipid metabolism. However, the role of adiponectin in pain conditions is largely unknown. This study aimed to identify the role and mechanism of adiponectin in nociceptive sensitivity under physiological and pathological states utilising adiponectin knockout (KO) mice. METHODS: Wild type (WT) and adiponectin KO mice were subjected to partial sciatic nerve ligation (pSNL) or sham operation. Pain-like behavioural tests, including thermal allodynia, hyperalgesia, and mechanical allodynia, were performed before and after pSNL from Day 3-21. Dorsal root ganglions (DRGs), lumbar spinal segments at L3-5, and somatosensory cortex were collected for protein measurement via western blotting and immunofluorescence staining. RESULTS: Compared with WT mice, KO mice had significantly lower (40-50%) paw withdrawal latency to innocuous and noxious stimuli before and after pSNL. In DRG neurones from KO mice, where adiponectin receptor (AdipoR) 2 is located, phosphorylated p38 mitogen-activated protein kinase (p-p38 MAPK) and heat-sensitive transient receptor potential cation channel subfamily V member 1 (TRPV1) were significantly higher (by two- to three-fold) than from WT mice. In spinal microglia and somatosensory cortical neurones, where AdipoR1 is mainly located, p-p38 MAPK and TRPV1 were also higher (by two- to three-fold) in KO compared with WT mice, and altered signalling of these molecules was exacerbated (1.2- to 1.3-fold) by pSNL. CONCLUSIONS: Our results show that adiponectin regulates thermal nociceptive sensitivity by inhibiting activation of DRG neurones, spinal microglia, and somatosensory cortical neurones in physiological and neuropathic pain states. This study has relevance for patients with adiponectin disorders, such as obesity and diabetes.


Asunto(s)
Adiponectina/fisiología , Hiperalgesia/fisiopatología , Neuralgia/fisiopatología , Nocicepción/fisiología , Adiponectina/deficiencia , Animales , Modelos Animales de Enfermedad , Calor , Hiperalgesia/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones Noqueados , Neuralgia/metabolismo , Receptores de Adiponectina/fisiología , Corteza Somatosensorial/metabolismo , Médula Espinal/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Biochim Biophys Acta ; 1861(5): 430-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26923434

RESUMEN

Mechanistic target of rapamycin complex 1 (mTORC1) loss of function reduces adiposity whereas partial mTORC1 inhibition enhances fat deposition. Herein we evaluated how constitutive mTORC1 activation in adipocytes modulates adiposity in vivo. Mice with constitutive mTORC1 activation in adipocytes induced by tuberous sclerosis complex (Tsc)1 deletion and littermate controls were evaluated for body mass, energy expenditure, glucose and fatty acid metabolism, mitochondrial function, mRNA and protein contents. Adipocyte-specific Tsc1 deletion reduced visceral, but not subcutaneous, fat mass, as well as adipocyte number and diameter, phenotypes that were associated with increased lipolysis, UCP-1 content (browning) and mRNA levels of pro-browning transcriptional factors C/EBPß and ERRα. Adipocyte Tsc1 deletion enhanced mitochondrial oxidative activity, fatty acid oxidation and the expression of PGC-1α and PPARα in both visceral and subcutaneous fat. In brown adipocytes, however, Tsc1 deletion did not affect UCP-1 content and basal respiration. Adipocyte Tsc1 deletion also reduced visceral adiposity and enhanced glucose tolerance, liver and muscle insulin signaling and adiponectin secretion in mice fed with purified low- or high-fat diet. In conclusion, adipocyte-specific Tsc1 deletion enhances mitochondrial activity, induces browning and reduces visceral adiposity in mice.


Asunto(s)
Adipocitos Marrones/enzimología , Adipocitos Blancos/enzimología , Tejido Adiposo Pardo/enzimología , Adiposidad , Grasa Intraabdominal/enzimología , Mitocondrias/enzimología , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Adipocitos Marrones/ultraestructura , Adipocitos Blancos/ultraestructura , Adiponectina/deficiencia , Adiponectina/genética , Tejido Adiposo Pardo/ultraestructura , Adiposidad/genética , Animales , Respiración de la Célula , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Metabolismo Energético , Activación Enzimática , Regulación de la Expresión Génica , Genotipo , Glucosa/metabolismo , Insulina/metabolismo , Grasa Intraabdominal/ultraestructura , Lipólisis , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/ultraestructura , Oxidación-Reducción , Fenotipo , Transducción de Señal , Factores de Tiempo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
20.
Kidney Int ; 91(2): 324-337, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27914707

RESUMEN

The kidney controls systemic calcium and phosphate levels and disturbances of its control mechanisms can lead to a variety of diseases. The insulin-sensitizing adipokine adiponectin is renoprotective and accelerates functional recovery following renal injury. However, unlike other adipokines, adiponectin is reduced in obesity. High adiponectin levels are also correlated with bone loss, suggestive of an additional action in mineral metabolism. Using knockout, wild-type, and adiponectin-overexpressing transgenic mice, we sought to identify the mechanistic basis for adiponectin's ability to regulate calcium and phosphate balance at the level of the kidney. Adiponectin knockout mice exhibited lower serum calcium, lower urinary calcium excretion, and markedly lower serum fibroblast growth factor 23 (FGF23) levels, although circulating klotho concentrations were significantly higher than in wild-type littermates. The transgenic mice exhibited lower bone mass and strength, particularly compared to adiponectin knockout mice. The transgenic mice were hyper-responsive to a 2% phosphate-enriched diet, exhibiting 2-fold higher serum FGF23 and concomitantly higher fractional phosphate excretion. These mice also excreted more calcium with calcium-enriched diet and had less renal klotho protein expression. In contrast, the knockout mice exhibited a smaller increase in FGF23 and maintained elevated klotho levels on both mineral challenges. Kidney-specific adiponectin expression in doxycycline-inducible adiponectin mice and adiponectin addition in vitro confirmed adiponectin's ability to reduce tubular epithelial cell klotho secretion. Thus, adiponectin alters calcium and phosphate balance and renal mineral excretion, in part, through klotho. This work highlights the profound effects of adipose tissue on renal function and has identified a new mechanism by which adiponectin may regulate bone mass.


Asunto(s)
Adiponectina/metabolismo , Calcio de la Dieta/metabolismo , Glucuronidasa/sangre , Riñón/metabolismo , Fosfatos/metabolismo , Fósforo Dietético/metabolismo , Eliminación Renal , Adiponectina/deficiencia , Adiponectina/genética , Animales , Fenómenos Biomecánicos , Calcio de la Dieta/sangre , Calcio de la Dieta/orina , Colágeno/metabolismo , Perros , Fémur/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Fibrosis , Genotipo , Glucuronidasa/genética , Homeostasis , Hormonas/sangre , Riñón/patología , Túbulos Renales/metabolismo , Proteínas Klotho , Células de Riñón Canino Madin Darby , Masculino , Ratones Noqueados , Osteogénesis , Fenotipo , Fosfatos/sangre , Fosfatos/orina , Fósforo Dietético/sangre , Fósforo Dietético/orina , Columna Vertebral/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA