Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetes Metab Res Rev ; 40(1): e3764, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38287717

RESUMEN

AIMS: Insulin resistance (IR) is associated with cardiometabolic multimorbidity (CMM). We aimed to explore the predictive value of six surrogate IR indexes-Chinese visceral adiposity index (CVAI), lipid accumulation product (LAP), triglyceride-glucose (TyG), atherogenic index of plasma (AIP), TyG-body mass index (TyGBMI), and TyG-waist circumference (TyGwaist)-to establish the CMM incidence in Chinese middle-aged and older populations. MATERIAL AND METHODS: To estimate the odds ratio (OR) with a 95% confidence interval (95% CI) for incident CMM using six surrogates, we analysed data from the China Health and Retirement Longitudinal Study using multivariate logistic regression models. The nonlinear dose-response correlation was evaluated using restricted cubic spline analysis; predictive performance was assessed using receiver operator characteristic curves. RESULTS: Among 6451 eligible participants, 268 (4.2%) developed CMM during the 4-year follow-up period. The ORs (95% CI) for incident CMM increased with increasing CVAI quartiles (Q) [Q2: 1.71, 1.03-2.90; Q3: 2.72, 1.70-4.52; Q4: 5.16, 3.29-8.45; all p < 0.05] after full adjustment, with Q1 as the reference. Other indexes yielded similar results. These associations remained significant in individuals with a normal body mass index. Notably, CVAI, AIP, and TyG exhibited a linear dose-response relationship with CMM (Pnonlinear ≥0.05), whereas LAP, TyGBMI, and TyGwaist displayed significant nonlinear correlations (Pnonlinear <0.05). The area under the curve for the CVAI (0.691) was significantly superior to that of other indexes (all p < 0.05). CONCLUSIONS: The six IR surrogates were independently associated with CMM incidence. CVAI may be the most appropriate indicator for predicting CMM in middle-aged and older Chinese populations.


Asunto(s)
Enfermedades Cardiovasculares , Resistencia a la Insulina , Persona de Mediana Edad , Humanos , Anciano , Estudios Longitudinales , Jubilación , Incidencia , Multimorbilidad , Glucosa , China/epidemiología , Triglicéridos
2.
Cell Mol Life Sci ; 81(1): 4, 2023 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-38070059

RESUMEN

Obesity is characterized by chronic low-grade inflammation, which is driven by macrophage infiltration in adipose tissue and leads to elevated cytokines such as interleukin-1ß (IL-1ß) in the circulation and tissues. Previous studies demonstrate that SENP3, a redox-sensitive SUMO2/3-specific protease, is strongly implicated in the development and progression of cancer and cardiovascular diseases. However, the role of SENP3 in obesity-associated inflammation remains largely unknown. To better understand the effects of SENP3 on adipose tissue macrophage (ATM) activation and function within the context of obesity, we generated mice with myeloid-specific deletion of SENP3 (Senp3flox/flox;Lyz2-Cre mice). We found that the expression of SENP3 is dramatically increased in ATMs during high-fat diet (HFD)-induced obesity in mice. Senp3flox/flox;Lyz2-Cre mice show lower body weight gain and reduced adiposity and adipocyte size after challenged with HFD and during aging. Myeloid-specific SENP3 deletion attenuates macrophage infiltration in adipose tissue and reduces serum levels of inflammatory factors during diet and age-induced obesity. Furthermore, we found that SENP3 knockout markedly inhibits cytokine release from macrophage after lipopolysaccharide and palmitic acid treatment in vitro. Mechanistically, in cultured peritoneal macrophages, SENP3 protein level is enhanced by IL-1ß, in parallel with the upregulation of Yes-associated protein 1 (YAP1). Moreover, we demonstrated that SENP3 modulates de-SUMO modification of YAP1 and SENP3 deletion abolishes the upregulation of YAP1 induced by IL-1ß. Most importantly, SENP3 deficiency reduces YAP1 protein level in adipose tissue during obesity. Our results highlight the important role of SENP3 in ATM inflammation and diet and age-induced obesity.


Asunto(s)
Resistencia a la Insulina , Sumoilación , Animales , Ratones , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Inflamación/metabolismo , Dieta Alta en Grasa/efectos adversos , Citocinas/metabolismo , Factores de Transcripción/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo
3.
Acta Biochim Biophys Sin (Shanghai) ; 55(2): 285-294, 2023 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-36786072

RESUMEN

Septic cardiomyopathy is associated with mechanisms such as excessive inflammation, oxidative stress, regulation of calcium homeostasis, endothelial dysfunction, mitochondrial dysfunction, and cardiomyocyte death, and there is no effective treatment at present. MOTS-c is a mitochondria-derived peptide (MDP) encoded by mitochondrial DNA (mtDNA) that protects cells from stresses in an AMPK-dependent manner. In the present study, we aim to explore the protective effect of MOTS-c on lipopolysaccharide (LPS)-induced septic cardiomyopathy. LPS is used to establish a model of septic cardiomyopathy. Our results demonstrate that MOTS-c treatment reduces the mRNA levels of inflammatory cytokines ( IL-1ß, IL-4, IL-6, and TNFα) in cardiomyocytes and the levels of circulating myocardial injury markers, such as CK-MB and TnT, alleviates cardiomyocyte mitochondrial dysfunction and oxidative stress, reduces cardiomyocyte apoptosis, activates cardioprotection-related signaling pathways, including AMPK, AKT, and ERK, and inhibits the inflammation-related signaling pathways JNK and STAT3. However, treatment with the AMPK pathway inhibitor compound C (CC) abolishes the positive effect of MOTS-c on LPS stress. Collectively, our research suggests that MOTS-c may attenuate myocardial injury in septic cardiomyopathy by activating AMPK and provides a new idea for therapeutic strategies in septic cardiomyopathy.


Asunto(s)
Cardiomiopatías , Lipopolisacáridos , Humanos , Lipopolisacáridos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/etiología , Cardiomiopatías/prevención & control , Citocinas , Inflamación
4.
Eur J Pharmacol ; 947: 175451, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-36502962

RESUMEN

BACKGROUND: Jujuboside A (JuA), as a main effective component of Jujubogenin, has long been known as a sedative-hypnotic drug. The aim of the current study was to investigate the potential effect of JuA on sepsis-induced cardiomyopathy (SIC) induced by lipopolysaccharide (LPS). METHOD: Wide type C57BL/6 mice and neonatal rat cardiomyocytes (NRCMs) were exposed to LPS to establish myocardial toxicity models. Cardiac function of septic mice was detected by echocardiography. Moreover, the survival rate was calculated for 7 days. ELISA assays were used to analyze inflammatory factors in serum. Furthermore, western blotting, flow cytometry and TUNEL staining were performed to assess cell apoptosis and transmission electron microscopy detect the number of autophagosomes in myocardium. Finally, the expression of proteins related to pyroptosis, autophagy and oxidative stress was analyzed by western blotting and immunohistochemistry staining. RESULTS: Results showed that JuA pretreatment significantly improved the survival rate and cardiac function, and suppressed systemic inflammatory response in septic mice. Further study revealed that JuA could decrease cell apoptosis and pyroptosis; instead, it strengthened autophagy in SIC. Moreover, JuA also significantly decreased oxidative stress and nitrodative stress, as evidenced by suppressing the superoxide production and downregulating iNOS and gp91 expression in vivo. In addition, the autophagy inhibitor 3-MA significantly abolished the effect of JuA on autophagic activity in SIC. CONCLUSION: In conclusion, the findings indicated that JuA attenuates cardiac function via blocking inflammasome-mediated apoptosis and pyroptosis, at the same time by enhancing autophagy in SIC, heralding JuA as a potential therapy for sepsis.


Asunto(s)
Cardiomiopatías , Sepsis , Ratas , Ratones , Animales , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/etiología , Autofagia , Sepsis/complicaciones , Sepsis/tratamiento farmacológico
5.
Comput Biol Chem ; 87: 107272, 2020 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-32438115

RESUMEN

The human cardiac troponin (hcTn) has been implicated in diverse cardiovascular diseases (CDs). The protein function is regulated by the inter-subunit interaction between the N-terminal domain of hcTnC and the C-terminal switch peptide of hcTnI; disruption of the interaction has been recognized as a potential therapeutic strategy for CDs. Here, we report use of biogenic medicines as small-molecule competitors to directly disrupt the protein-protein interaction by competitively targeting the core binding site (CBS) of hcTnC NTD domain. A multistep virtual screening protocol is performed against a biogenic compound library to identify competitor candidates and competition assay is employed to verify the screening results. Consequently, two compounds Collismycin and Compound e are identified as strong competitors (CC50 < 10 µM) with hcTnI for hcTnC CBS site, while other tested compounds are found to have moderate (CC50 = 10-100 µM), low (CC50 > 100 µM) or no (CC50 = N.D.) potency. The competitor ligands are anchored at the core groove of hcTnC CBS site through aromatic and hydrophobic interactions, while few peripheral hydrogen bonds are formed to further confer specificity for domain-compound recognition. These molecular-level findings would benefit from further in vitro and in vivo studies at cellular and animal levels, which can help to practice the ultimate therapeutic purpose.

6.
Acta Biochim Biophys Sin (Shanghai) ; 52(2): 141-149, 2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-31942916

RESUMEN

Pathological studies have shown that the vulnerability of plaques affects outcomes in patients with atherosclerosis (AS), a chronic inflammatory disease and common cause of morbidity and mortality worldwide. Although emerging technologies have enabled early diagnosis of AS with high-risk vulnerable plaques, more accurate and noninvasive diagnostic methods are urgently required. To this end, molecules involved in genetic or epigenetic regulation of the vulnerability of atherosclerotic plaques have been extensively studied. Here, we evaluated long noncoding RNA (lncRNA) variability by microarray assay in murine aortic endothelial cells (MAECs) bearing vulnerable plaques and identified the novel functional lncRNA UC.98, whose expression pattern was associated with the vulnerability of atherosclerotic plaques. Consistent with this, clinical statistics comparing the peripheral blood specimens from sets of patients with AS with or without vulnerable plaques confirmed the linear relationship between the expression pattern of UC.98 and plaque instability. Moreover, MTT assays and western blot analysis showed that silencing of intrinsic UC.98 in MAECs not only suppressed cell proliferation but also decreased the expressions of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, thereby inactivating the nuclear factor-κB pathway. In conclusion, our results highlighted the pivotal role of UC.98 in regulating the vulnerability of plaques during AS progression and suggested that UC.98 may be a biomarker of the early diagnosis and prognosis of AS with vulnerable plaques and a potential therapeutic target for slowing AS progression.


Asunto(s)
Aorta/citología , Células Endoteliales/patología , Placa Aterosclerótica/patología , ARN Largo no Codificante/fisiología , Animales , Biomarcadores/análisis , Adhesión Celular , Proliferación Celular , Progresión de la Enfermedad , Ratones , ARN Largo no Codificante/sangre
7.
Metabolism ; 105: 154046, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31837300

RESUMEN

BACKGROUND: Hepatic ER stress is a risk factor of insulin resistance and type 2 diabetes. X-box binding protein 1 spliced (XBP1s), a transcription factor, plays a key role in ameliorating insulin resistance and maintaining glucose homeostasis. Unfortunately, the short half-life of the protein dampens its clinical application, and the specific site of lysine residue that could be ubiquitinated and involved in the degradation of XBP1s remains elusive. METHODS AND RESULTS: Here, we identified K60 and K77 on XBP1s as two pivotal ubiquitin sites required for its proteasome-dependent degradation. We also constructed a double mutant form of XBP1s (K60/77R) and found that it showed higher capacity in resisting against ubiquitin-mediated protein degradation, increasing nuclear translocation, enhancing transcriptional activity, suppressing ER stress and promoting Foxo1 degradation, compared to that of wild type XBP1s (WT). Consistently, overexpression of the K60/77R XBP1s mutant in DIO mice increased the ability to reduce ER stress and decrease Foxo1 levels, thus contributed to maintaining glucose homeostasis. CONCLUSION: Our results suggest that delaying the degradation of XBP1s by preventing ubiquitination might provide a strategic approach for reducing ER stress as an anti-diabetes therapy.


Asunto(s)
Ubiquitinación/genética , Proteína 1 de Unión a la X-Box/genética , Animales , Dieta Alta en Grasa , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Proteína Forkhead Box O1/biosíntesis , Proteína Forkhead Box O1/genética , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Células HEK293 , Homeostasis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Obesidad/genética , Complejo de la Endopetidasa Proteasomal , Translocación Genética , Ubiquitinación/fisiología
8.
Front Cell Neurosci ; 11: 391, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29311826

RESUMEN

The spinal cord injury (SCI) is a detrimental neurological disease involving the primary mechanical injury and secondary inflammatory damage. Curtailing the detrimental neuroinflammation would be beneficial for spinal cord function recovery. Microglia reside in the spinal cord and actively participate in the onset, progression and perhaps resolution of post-SCI neuroinflammation. In the current study, we tested the effects of methylene blue on microglia both in vitro and in a rat SCI model. We found that methylene blue inhibited the protein levels of IL-1ß and IL-18 rather than their mRNA levels in activated microglia. Further investigation indicated that methylene blue deceased the activation of NLRP3 inflammasome and NLRC4 inflammasome in microglia in vitro. Moreover, in the rat SCI model, the similar effect of methylene blue on post-SCI microglia was also observed, except that the activation of NLRC4 inflammasome was not seen. The inhibition of microglia NLRP3 inflammasome was associated with down-regulation of intracellular reactive oxygen species (ROS). The administration of methylene blue mitigated the overall post-SCI neuroinflammation, demonstrated by decreased pro-inflammatory cytokine production and leukocyte infiltrates. Consequently, the neuronal apoptosis was partially inhibited and the hind limb locomotor function was ameliorated by methylene blue treatment. Our research highlights the role of methylene blue in inhibiting post-SCI neuroinflammation, and suggests that methylene blue might be used for SCI therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA