Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 38(13): e23765, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38934372

RESUMEN

The importance of autophagy in atherosclerosis has garnered significant attention regarding the potential applications of autophagy inducers. However, the impact of TAT-Beclin1, a peptide inducer of autophagy, on the development of atherosclerotic plaques remains unclear. Single-cell omics analysis indicates a notable reduction in GAPR1 levels within fibroblasts, stromal cells, and macrophages during atherosclerosis. Tat-beclin1 (T-B), an autophagy-inducing peptide derived from Beclin1, could selectively bind to GAPR1, relieving its inhibition on Beclin1 and thereby augmenting autophagosome formation. To investigate its impact on atherosclerosic plaque progression, we established the ApoE-/- mouse model of carotid atherosclerotic plaques. Surprisingly, intravenous administration of Tat-beclin1 dramatically accelerated the development of carotid artery plaques. Immunofluorescence analysis suggested that macrophage aggregation and autophagosome formation within atherosclerotic plaques were significantly increased upon T-B treatment. However, immunofluorescence and transmission electron microscopy (TEM) analysis revealed a reduction in autophagy flux through lysosomes. In vitro, the interaction between T-B and GAPR1 was confirmed in RAW264.7 cells, resulting in the increased accumulation of p62/SQSTM1 and LC3-II in the presence of ox-LDL. Additionally, T-B treatment elevated the protein levels of p62/SQSTM1, LC3-II, and cleaved caspase 1, along with the secretion of IL-1ß in response to ox-LDL exposure. In summary, our study underscores that T-B treatment amplifies abnormal autophagy and inflammation, consequently exacerbating atherosclerotic plaque development in ApoE-/- mice.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Autofagia , Beclina-1 , Placa Aterosclerótica , Animales , Ratones , Beclina-1/metabolismo , Beclina-1/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Autofagia/efectos de los fármacos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Células RAW 264.7 , Ratones Endogámicos C57BL , Masculino , Ratones Noqueados , Macrófagos/metabolismo
2.
Eur J Pharmacol ; 974: 176570, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38688398

RESUMEN

Mitochondrial dynamics play a crucial role in myocardial ischemia-reperfusion (I/R) injury, where an imbalance between fusion and fission processes occurs. However, effective measures to regulate mitochondrial dynamics in this context are currently lacking. Peptide derived from the 40 S ribosomal protein S6 (PDRPS6), a peptide identified via peptidomics, is associated with hypoxic stress. This study aimed to investigate the function and mechanism of action of PDRPS6 in I/R injury. In vivo, PDRPS6 ameliorated myocardial tissue injury and cardiomyocyte apoptosis and decreased cardiac function induced by I/R injury in rats. PDRPS6 supplementation significantly reduced apoptosis in vitro. Mechanistically, PDRPS6 improved mitochondrial function by decreasing reactive oxygen species (ROS) levels, maintaining mitochondrial membrane potential (MMP), and inhibiting mitochondrial fission. Pull-down assay analyses revealed that phosphoglycerate mutase 5 (PGAM5) may be the target of PDRPS6, which can lead to the dephosphorylation of dynamin-related protein1 (Drp1) at ser616 site. Overexpression of PGAM5 partially eliminated the effect of PDRPS6 on improving mitochondrial function. These findings suggest that PDRPS6 supplementation is a novel method for treating myocardial injuries caused by I/R.


Asunto(s)
Apoptosis , Dinámicas Mitocondriales , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Animales , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Ratas , Dinámicas Mitocondriales/efectos de los fármacos , Apoptosis/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Especies Reactivas de Oxígeno/metabolismo , Proteína S6 Ribosómica/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Dinaminas/metabolismo , Dinaminas/genética , Péptidos/farmacología , Péptidos/uso terapéutico , Fosforilación/efectos de los fármacos
3.
Clin Interv Aging ; 19: 503-515, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38525316

RESUMEN

Objective: This study aimed to explore the association of preoperative neutrophil percentage (NEUT%) with the risk of acute kidney injury (AKI) in patients with acute myocardial infarction (AMI) having undergone coronary interventional therapy. Methods: A single-center, retrospective and observational study was conducted. From December 2012 to June 2021, patients with AMI were enrolled and divided into AKI group and non-AKI group. The NEUT% in the two groups was compared. The association between NEUT% with the risk of post-AMI AKI was analyzed by univariate and multivariable logistic regression. Kaplan-Meier survival curve was drawn to evaluate the prognostic ability of NEUT% for short-term all-cause death following AMI. Results: A total of 3001 consecutive patients were enrolled with an average age of 64.38 years. AKI occurred in 327 (10.9%) patients. The NEUT% was higher in the AKI group than in the non-AKI group ([76.65±11.43]% versus [73.22±11.83]%, P<0.001). NEUT% was also identified as an independent risk factor for AKI in AMI patients after adjustment (OR=1.021, 95% CI: 1.010-1.033, P < 0.001). Compared with those at the lowest quartile of NEUT%, the patients at quartiles 2-4 had a higher risk of AKI (P for trend = 0.003). The odds of AKI increased by 29.0% as NEUT% increased by 1 standard deviation (OR=1.290, 95% CI: 1.087-1.531, P = 0.004). After a median of 35 days follow-up, 93 patients died. Patients with a higher NEUT% presented a higher risk of all-cause death after AMI (Log rank: χ2 =24.753, P<0.001). Conclusion: In AMI patients, the peripheral blood NEUT% was positively associated with the odds of AKI and short-term all-cause mortality. NEUT% may provide physicians with more information about disease development and prognosis.


Asunto(s)
Lesión Renal Aguda , Infarto del Miocardio , Humanos , Anciano , Neutrófilos , Estudios Retrospectivos , Pronóstico , Infarto del Miocardio/complicaciones , Biomarcadores , Factores de Riesgo , Lesión Renal Aguda/epidemiología , Lesión Renal Aguda/etiología
4.
Lab Invest ; 103(6): 100126, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36889540

RESUMEN

Sepsis-associated acute kidney injury (S-AKI) is a frequent complication in patients who are critically ill, which is often initiated by glomerular endothelial cell dysfunction. Although transient receptor vanilloid subtype 4 (TRPV4) ion channels are known to be permeable to Ca2+ and are widely expressed in the kidneys, the role of TRPV4 on glomerular endothelial inflammation in sepsis remains elusive. In the present study, we found that TRPV4 expression in mouse glomerular endothelial cells (MGECs) increased after lipopolysaccharide (LPS) stimulation or cecal ligation and puncture challenge, which increased intracellular Ca2+ in MGECs. Furthermore, the inhibition or knockdown of TRPV4 suppressed LPS-induced phosphorylation and translocation of inflammatory transcription factors NF-κB and IRF-3 in MGECs. Clamping intracellular Ca2+ mimicked LPS-induced responses observed in the absence of TRPV4. In vivo experiments showed that the pharmacologic blockade or knockdown of TRPV4 reduced glomerular endothelial inflammatory responses, increased survival rate, and improved renal function in cecal ligation and puncture-induced sepsis without altering renal cortical blood perfusion. Taken together, our results suggest that TRPV4 promotes glomerular endothelial inflammation in S-AKI and that its inhibition or knockdown alleviates glomerular endothelial inflammation by reducing Ca2+ overload and NF-κB/IRF-3 activation. These findings provide insights that may aid in the development of novel pharmacologic strategies for the treatment of S-AKI.


Asunto(s)
Lesión Renal Aguda , Antineoplásicos , Sepsis , Ratones , Animales , FN-kappa B/metabolismo , Células Endoteliales/metabolismo , Lipopolisacáridos/farmacología , Canales Catiónicos TRPV/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Inflamación/metabolismo , Sepsis/complicaciones , Sepsis/metabolismo
5.
BMC Med ; 20(1): 308, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36127705

RESUMEN

BACKGROUND: The relationship between vitamin D status and mortality in patients with osteoarthritis (OA) is unknown. This study investigated the associations of serum 25-hydroxyvitamin D [25(OH)D] concentrations with all-cause and cause-specific mortality among American adults with OA. METHODS: This study included 2556 adults with OA from the National Health and Nutrition Examination Survey (2001-2014). Death outcomes were ascertained by linkage to National Death Index (NDI) records through 31 December 2015. Cox proportional hazards model and two-piecewise Cox proportional hazards model were used to elucidate the nonlinear relationship between serum 25(OH)D concentrations and mortality in OA patients, and stratified analyses were performed to identify patients with higher mortality risk. RESULTS: During 16,606 person-years of follow-up, 438 all-cause deaths occurred, including 74 cardiovascular disease (CVD)-related and 78 cancer deaths. After multivariable adjustment, lower serum 25(OH)D levels were significantly and nonlinearly associated with higher risks of all-cause and CVD mortality among participants with OA. Furthermore, we discovered L-shaped associations between serum 25(OH)D levels and all-cause and CVD mortality, with mortality plateauing at 54.40 nmol/L for all-cause mortality and 27.70 nmol/L for CVD mortality. Compared to participants with 25(OH)D levels below the inflection points, those with higher levels had a 2% lower risk for all-cause mortality (hazard ratio [HR] 0.98, 95% confidence interval [CI] 0.96-0.99) and 17% lower risk for CVD mortality (HR 0.83, 95% CI 0.72-0.95). CONCLUSIONS: Nonlinear associations of serum 25(OH)D levels with all-cause and CVD mortality were observed in American patients with OA. The thresholds of 27.70 and 54.40 nmol/L for CVD and all-cause mortality, respectively, may represent intervention targets for lowering the risk of premature death and cardiovascular disease, but this needs to be confirmed in large clinical trials.


Asunto(s)
Enfermedades Cardiovasculares , Osteoartritis , Deficiencia de Vitamina D , Adulto , Causas de Muerte , Humanos , Encuestas Nutricionales , Estudios Prospectivos , Estados Unidos/epidemiología , Vitamina D/análogos & derivados
6.
EBioMedicine ; 82: 104164, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35843176

RESUMEN

BACKGROUND: Exercise training protects the heart against pathological cardiac remodeling and confers cardioprotection from heart failure. However, the underlying mechanism is still elusive. METHODS: An integrative analysis of multi-omics data of the skeletal muscle in response to exercise is performed to search for potential exerkine. Then, CCDC80tide is examined in humans after acute exercise. The role of CCDC80tide is assessed in a mouse model of hypertensive cardiac remodeling and in hypertension-mediated cell injury models. The transcriptomic analysis and immunoprecipitation assay are conducted to explore the mechanism. FINDINGS: The coiled-coil domain-containing protein 80 (CCDC80) is found strongly positively associated with exercise. Interestingly, exercise stimuli induce the secretion of C-terminal CCDC80 (referred as CCDC80tide hereafter) via EVs-encapsulated CCDC80tide into the circulation. Importantly, cardiac-specific expression of CCDC80tide protects against angiotensin II (Ang II)-induced cardiac hypertrophy and fibrosis in mice. In in vitro studies, the expression of CCDC80tide reduces Ang II-induced cardiomyocyte hypertrophy, cardiac microvascular endothelial cell (CMEC) inflammation, and mitigated vascular smooth muscle cell (VSMC) proliferation and collagen formation. To understand the cardioprotective effect of CCDC80tide, a transcriptomic analysis reveals a dramatic inhibition of the STAT3 (Signal transducer and activator of transcription 3) signaling pathway in CCDC80tide overexpressing cells. Mechanistically, CCDC80tide selectively interacts with the kinase-active form of JAK2 (Janus kinase 2) and consequently inhibits its kinase activity to phosphorylate and activate STAT3. INTERPRETATION: The results provide new insights into exercise-afforded cardioprotection in pathological cardiac remodeling and highlight the therapeutic potential of CCDC80tide in heart failure treatment. FUNDING: This work was supported by the National Natural Science Foundation of China [Grant/Award Numbers: 81770428, 81830010, 82130012, 81900438, 82100447); Shanghai Science and Technology Committee [Grant/Award Numbers: 21S11903000, 19JC1415702]; Emerging and Advanced Technology Programs of Hospital Development Center of Shanghai [Grant/Award Number: SHDC12018129]; China Postdoctoral Science Foundation [2021M692108]; and China National Postdoctoral Program for Innovative Talents [BX20200211].


Asunto(s)
Insuficiencia Cardíaca , Hipertensión , Angiotensina II/farmacología , Animales , China , Proteínas de la Matriz Extracelular/metabolismo , Corazón , Insuficiencia Cardíaca/metabolismo , Humanos , Hipertensión/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Remodelación Ventricular
7.
Oxid Med Cell Longev ; 2022: 2017815, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35464766

RESUMEN

Purpose: Activation of NLR (nucleotide-binding and leucine-rich repeat immune receptor) family pyrin domain containing 3 (NLRP3) inflammasome mediating interleukin- (IL-) 1ß secretion has emerged as an important component of inflammatory processes in atherogenesis. The nuclear receptor Nur77 is highly expressed in human atherosclerotic lesions; however, its functional role in macrophage NLRP3 inflammasome activation has not yet been clarified. Methods, Materials, and Results. Eight-week-old apolipoprotein E (ApoE)-/- and ApoE-/- Nur77-/- mice that were fed a Western diet underwent partial ligation of the left common carotid artery (LCCA) and left renal artery (LRA) to induce atherogenesis. Four weeks later, severe plaque burden associated with increased lipid deposition, reduced smooth muscle cells, macrophage infiltration, and decreased collagen expression was identified in ApoE-/- Nur77-/- mice compared with those in ApoE-/- mice. ApoE-/- Nur77-/- mice showed increased macrophage inflammatory responses in carotid atherosclerotic lesions. In vitro studies demonstrated that oxidized low-density lipoprotein cholesterol (ox-LDL) increased the release of lactate dehydrogenase (LDH) and upregulated the expressions of cleaved caspase-1, cleaved IL-1ß and gasdermin D (GSMD) in WT peritoneal macrophages (PMs) in a NLRP3-dependent manner. Nur77-/- PMs exhibited a further increased level of NLRP3 inflammasome-mediated inflammation under ox-LDL treatment compared with WT PMs. Mechanistically, Nur77 could bind to the promoter of NLRP3 and inhibit its transcriptional activity. Conclusions: This study demonstrated that Nur77 deletion promotes atherogenesis by exacerbating NLRP3 inflammasome-mediated inflammation.


Asunto(s)
Aterosclerosis , Inflamasomas , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/patología , Colesterol/metabolismo , Inflamasomas/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares
8.
Cardiovasc Drugs Ther ; 36(6): 1061-1073, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34410548

RESUMEN

PURPOSE: Macrophage apoptosis coupled with a defective phagocytic clearance of the apoptotic cells promotes plaque necrosis in advanced atherosclerosis, which causes acute atherothrombotic vascular disease. Nonsteroidal anti-inflammatory drug sulindac derivative K-80003 treatment was previously reported to dramatically attenuate atherosclerotic plaque progression and destabilization. However, the underlying mechanisms are not fully understood. This study aimed to determine the role of K-80003 on macrophage apoptosis and elucidate the underlying mechanism. METHODS: The mouse model of vulnerable carotid plaque in ApoE-/- mice was developed in vivo. Consequently, mice were randomly grouped into two study groups: the control group and the K-80003 group (30 mg/kg/day). Samples of carotid arteries were collected to determine atherosclerotic necrotic core area, cellular apoptosis, and oxidative stress. The effects of K-80003 on RAW264.7 macrophage apoptosis, oxidative stress, and autophagic flux were also examined in vitro. RESULTS: K-80003 significantly suppressed necrotic core formation and inhibited cellular apoptosis of vulnerable plaques. K-80003 can also inhibit 7-ketocholesterol-induced macrophage apoptosis in vitro. Furthermore, K-80003 inhibited intraplaque cellular apoptosis mainly through the suppression of oxidative stress, which is a key cause of advanced lesional macrophage apoptosis. Mechanistically, K-80003 prevented 7-ketocholesterol-induced impairment of autophagic flux in macrophages, evidenced by the decreased LC3II and SQSTM1/p62 expression, GFP-RFP-LC3 cancellation upon K-80003 treatment. CONCLUSION: Inhibition of macrophage apoptosis and necrotic core formation by autophagy-mediated reduction of oxidative stress is one mechanism of the suppression of plaque progression and destabilization by K-80003.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Ratones , Apoptosis , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Necrosis/metabolismo , Placa Aterosclerótica/metabolismo , Sulindac/metabolismo , Sulindac/farmacología
9.
Pharmacol Res ; 175: 105988, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34808368

RESUMEN

Myocardial ischemia-reperfusion (I/R) is a severe disease,but its underlying mechanism is not fully elucidated and no effective clinical treatment is available. Utilizing intracellular peptidomics, we identified a novel native peptide PDRL23A (Peptide Derived from RPL23A), that is intimately related to hypoxic stress. We further show that PDRL23A effectively alleviates hypoxia-induced cardiomyocyte injury in vitro, along with improvements in mitochondrial function and redox homeostasis, including ROS accumulation, oxidative phosphorylation, and mitochondrial membrane potential. Strikingly, the in vivo results indicate that, short-term pretreatment with PDRL23A could effectively inhibit I/R-induced cardiomyocyte death, myocardial fibrosis and decreased cardiac function. Interestingly, PDRL23A was found to interact with 60 S ribosomal protein L26 (RPL26), hampering RPL26-governed p53 translation, and resulting in a reduction in the level of p53 protein, which in turn reduced p53-mediated apoptosis under hypoxic conditions. Collectively, a native peptide, PDRL23A, which translationally regulates p53 to protect against myocardial I/R injury, has been identified for the first time. Our findings provide insight into the adaptive mechanisms of hypoxia and present a potential new treatment for myocardial I/R.


Asunto(s)
Hipoxia de la Célula , Miocitos Cardíacos/metabolismo , Péptidos/metabolismo , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Cultivadas , Masculino , Daño por Reperfusión Miocárdica , Péptidos/química , Ratas Sprague-Dawley , Proteínas Ribosómicas/química , Proteína p53 Supresora de Tumor/genética
10.
J Mol Cell Cardiol ; 159: 91-104, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34147480

RESUMEN

AIMS: Pathological cardiac hypertrophy induced by activation of the renin-angiotensin-aldosterone system (RAAS) is one of the leading causes of heart failure. However, in current clinical practice, the strategy for targeting the RAAS is not sufficient to reverse hypertrophy. Here, we investigated the effect of prostaglandin E1 (PGE1) on angiotensin II (AngII)-induced cardiac hypertrophy and potential molecular mechanisms underlying the effect. METHODS AND RESULTS: Adult male C57 mice were continuously infused with AngII or saline and treated daily with PGE1 or vehicle for two weeks. Neonatal rat cardiomyocytes were cultured to detect AngII-induced hypertrophic responses. We found that PGE1 ameliorated AngII-induced cardiac hypertrophy both in vivo and in vitro. The RNA sequencing (RNA-seq) and expression pattern analysis results suggest that Netrin-1 (Ntn1) is the specific target gene of PGE1. The protective effect of PGE1 was eliminated after knockdown of Ntn1. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the PGE1-mediated signaling pathway changes are associated with the mitogen-activated protein kinase (MAPK) pathway. PGE1 suppressed AngII-induced activation of the MAPK signaling pathway, and such an effect was attenuated by Ntn1 knockdown. Blockade of MAPK signaling rescued the phenotype of cardiomyocytes caused by Ntn1 knockdown, indicating that MAPK signaling may act as the downstream effector of Ntn1. Furthermore, inhibition of the E-prostanoid (EP) 3 receptor, as opposed to the EP1, EP2, or EP4 receptor, in cardiomyocytes reversed the effect of PGE1, and activation of EP3 by sulprostone, a specific agonist, mimicked the effect of PGE1. CONCLUSION: In conclusion, PGE1 ameliorates AngII-induced cardiac hypertrophy through activation of the EP3 receptor and upregulation of Ntn1, which inhibits the downstream MAPK signaling pathway. Thus, targeting EP3, as well as the Ntn1-MAPK axis, may represent a novel approach for treating pathological cardiac hypertrophy.


Asunto(s)
Alprostadil/farmacología , Angiotensina II/farmacología , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Netrina-1/genética , Subtipo EP3 de Receptores de Prostaglandina E/genética , Regulación hacia Arriba/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/genética
11.
Biochim Biophys Acta Gen Subj ; 1865(3): 129836, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33370564

RESUMEN

BACKGROUND: Emerging evidence revealed peptides within breast milk may be an abundant source of potential candidates for metabolism regulation. Our previous work identified numerous peptides existed in breast milk, but its function has not been validated. Thus, our study aims to screen for novel peptides that have the potential to antagonize obesity and diabetes. METHODS: A function screen was designed to identify the candidate peptide and then the peptide effect was validated by assessing lipid storage. Afterwards, the in vivo study was performed in two obese models: high-fat diet (HFD)-induced obese mice and obese ob/ob mice. For mechanism study, a RNA-seq analysis was conducted to explore the pathway that account for the biological function of peptide. RESULTS: By performing a small scale screening, a peptide (AVPVQALLLNQ) termed AOPDM1 (anti-obesity peptide derived from breast milk 1) was identified to reduce lipid storage in adipocytes. Further study showed AOPDM1 suppressed adipocyte differentiation by sustaining ERK activity at later stage of differentiation which down-regulated PPARγ expression. In vivo, AOPDM1 effectively reduced fat mass and improved glucose metabolism in high-fat diet (HFD)-induced obese mice and obese ob/ob mice. CONCLUSIONS: We identified a novel peptide AOPDM1 derived from breast milk could restrict adipocyte differentiation and ameliorate obesity through regulating MAPK pathway. GENERAL SIGNIFICANCE: Our findings may provide a potential candidate for the discovery of therapeutic drugs for obesity and type 2 diabetes.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Leche Humana/química , Obesidad/prevención & control , Péptidos/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Fármacos Antiobesidad/síntesis química , Diferenciación Celular/efectos de los fármacos , Dieta Alta en Grasa , Femenino , Regulación de la Expresión Génica , Humanos , Leptina/genética , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Obesidad/etiología , Obesidad/genética , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Péptidos/síntesis química , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Int J Radiat Oncol Biol Phys ; 109(2): 540-552, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32942003

RESUMEN

PURPOSE: Autophagy inhibition is a novel therapeutic strategy suggested for patients with advanced cancer, especially those who have undergone radiation therapy. In the present study, we investigated whether autophagy inhibitors accelerate the progression of radiation-associated atherosclerosis (RAA). METHODS AND MATERIALS: Eight-week-old apolipoprotein (ApoE-/-) mice were fed a Western diet, and their left common carotid arteries were partially ligated to induce atherogenesis. Four weeks later, local ionizing radiation (IR) at a dose of 5 or 10 Gy was used to induce RAA in the left common carotid artery. After another 4 weeks, severe plaque burden associated with increased macrophage infiltration and lipid deposition, reduced smooth muscle cells, and decreased collagen expression was observed. In addition, these changes occurred in a dose-dependent manner. Improved autophagic flux caused by IR was observed in both macrophages of the atherosclerotic plaque and peritoneal macrophages in vitro. The inhibition of autophagic flux by chloroquine (50 mg/kg/d) further accelerated the progression of RAA in the left common carotid arteries of ApoE-/- mice. Furthermore, chloroquine treatment exacerbated IR-induced p65 nuclear translocation, IκBα degradation, and transcription of nuclear factor-κB (NF-κB) target genes in peritoneal macrophages. CONCLUSIONS: IR promotes atherogenesis and increases autophagic flux. In addition, autophagy inhibition by chloroquine accelerates the progression of RAA lesions by stimulating NF-κB-mediated inflammatory responses in macrophages.


Asunto(s)
Antineoplásicos/efectos adversos , Aterosclerosis/patología , Autofagia/efectos de los fármacos , Traumatismos por Radiación/patología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Cloroquina/efectos adversos , Progresión de la Enfermedad , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Inhibidor NF-kappaB alfa/metabolismo , Proteolisis/efectos de los fármacos , Traumatismos por Radiación/etiología , Traumatismos por Radiación/metabolismo , Factor de Transcripción ReIA/metabolismo
13.
Mol Med Rep ; 19(1): 302-308, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30431112

RESUMEN

Acute myocardial infarction (AMI) is a life­threatening disease and seriously influences patient quality of life. Long non­coding RNAs (lncRNAs), an emerging class of non­coding genes, have attracted attention in research, however, whether lncRNAs serve a function in acute ischemic hypoxia remains to be elucidated. In the present study, an lncRNA microarray was used to analyze differential lncRNA expression in acute ischemic hypoxia. A total of 323 lncRNAs were identified, 168 of which were upregulated and 155 of which were downregulated. Gene Ontology and Pathway analyses were also used to identify the potential functions of dysregulated lncRNAs; it was predicted that these dysregulated lncRNAs may contribute to the initiation of AMI. It was demonstrated that an lncRNA termed sloyfley may influence acute ischemic hypoxia through its neighboring gene Peg3, which has been linked to brain ischemia hypoxia. In summary, the present study identified numerous lncRNAs, which may provide further opportunities for the development of novel therapeutic strategies.


Asunto(s)
Perfilación de la Expresión Génica , Hipoxia/fisiopatología , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , Enfermedad Aguda , Animales , Células Cultivadas , Masculino , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley
14.
Biomed Res Int ; 2018: 4658024, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30003100

RESUMEN

In previous studies, we have demonstrated the function of uc.167 in the heart development. DNA methylation plays a crucial role in regulating the expression of developmental genes during embryonic development. In this study, the methylomic landscape was investigated in order to identify the DNA methylation alterations. Methylated DNA immunoprecipitation (MeDIP) was performed to examine the differences in methylation status of overexpressed uc.167 in P19 cells. GO and KEGG pathway analyses of differentially methylated genes were also conducted. We found that the distribution of differentially methylated regions (DMRs) peaks in different components of genome was mainly located in intergenic regions and intron. The biological process associated with uc.167 was focal adhesion and Rap1 signaling pathway. MEF2C was significantly decreased in uc.167 overexpressed group, suggesting that uc.167 may influence the P19 differentiation through MEF2C reduction. Taken together, our findings revealed that the effect of uc.167 on P19 differentiation may be attributed to the altered methylation of specific genes.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Corazón/crecimiento & desarrollo , ARN Largo no Codificante , Animales , Línea Celular , Genoma , Ratones , Transducción de Señal
15.
Int J Biol Macromol ; 118(Pt A): 1142-1148, 2018 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-30001601

RESUMEN

OBJECTIVES: Oxidative stress plays an important role in myocardial ischemia-reperfusion (I/R) injury. And pNaKtide is known to inhibit Na/K-ATPase/Src/reactive oxygen species (ROS) amplification signaling. Accordingly, we aimed to investigate the effect of pNaKtide on myocardial I/R injury. METHODS: We first determine the effect of pNaKtide on hypoxia- or cobalt chloride-induced injury in embryonic heart-derived H9c2 cells via measuring lactate dehydrogenase (LDH) level and trypan blue stain assay. In addition, TUNEL stain assay and western blot analysis of cleaved-PARP and cleaved-caspase3 were performed to detect apoptosis level. Meanwhile, ROS accumulation was assessed by dichlorofluorescin diacetate (DCFH-DA) assay. Then we conducted cell counting kit-8 (CCK-8) and flow cytometry to examine cell proliferation and cell cycle respectively. We next generated rat I/R model and determined the effect of pNaKtide by measuring serum LDH and evaluating heart pathology. At last, the activities of Src and ERK1/2 were examined via western blot to clarify molecular mechanism. RESULTS: In vitro, pNaKtide exposure significantly attenuated the H9c2 cells death and ROS accumulation induced by hypoxia or cobalt chloride. And no significant effect was detected on cell cycle and proliferation upon pNaKtide administration. In vivo, pNaKtide distinctly decreased serum LDH level and ameliorated I/R induced myocardial injury in the rats. Western blot analysis revealed pNaKtide decreased Src and ERK1/2 activities robustly. CONCLUSIONS: The results provided evidence that pNaKtide exhibited cardioprotective effect against hypoxia-induced injury in vitro and in vivo. And pNaKtide might be a potential molecular for therapy of I/R related heart disease.


Asunto(s)
Daño por Reperfusión Miocárdica/prevención & control , Péptidos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular , Humanos , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Familia-src Quinasas/antagonistas & inhibidores
16.
Exp Mol Med ; 50(2): e447, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29504607

RESUMEN

In a previous study, we screened thousands of long non-coding RNAs (lncRNAs) to assess their potential relationship with congenital heart disease (CHD). In this study, uc.4 attracted our attention because of its high level of evolutionary conservation and its antisense orientation to the CASZ1 gene, which is vital for heart development. We explored the function of uc.4 in cells and in zebrafish, and describe a potential mechanism of action. P19 cells were used to investigate the function of uc.4. We studied the effect of uc.4 overexpression on heart development in zebrafish. The overexpression of uc.4 influenced cell differentiation by inhibiting the TGF-beta signaling pathway and suppressed heart development in zebrafish, resulting in cardiac malformation. Taken together, our findings show that uc.4 is involved in heart development, thus providing a potential therapeutic target for CHD.


Asunto(s)
Diferenciación Celular/genética , ARN Largo no Codificante/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis , Biomarcadores , Ciclo Celular/genética , Línea Celular , Biología Computacional/métodos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Bases de Datos Genéticas , Expresión Génica , Perfilación de la Expresión Génica , Genes Letales , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Humanos , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...