Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Circulation ; 149(9): 684-706, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-37994595

RESUMEN

BACKGROUND: The majority of people with diabetes are susceptible to cardiac dysfunction and heart failure, and conventional drug therapy cannot correct diabetic cardiomyopathy progression. Herein, we assessed the potential role and therapeutic value of USP28 (ubiquitin-specific protease 28) on the metabolic vulnerability of diabetic cardiomyopathy. METHODS: The type 2 diabetes mouse model was established using db/db leptin receptor-deficient mice and high-fat diet/streptozotocin-induced mice. Cardiac-specific knockout of USP28 in the db/db background mice was generated by crossbreeding db/m and Myh6-Cre+/USP28fl/fl mice. Recombinant adeno-associated virus serotype 9 carrying USP28 under cardiac troponin T promoter was injected into db/db mice. High glucose plus palmitic acid-incubated neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes were used to imitate diabetic cardiomyopathy in vitro. The molecular mechanism was explored through RNA sequencing, immunoprecipitation and mass spectrometry analysis, protein pull-down, chromatin immunoprecipitation sequencing, and chromatin immunoprecipitation assay. RESULTS: Microarray profiling of the UPS (ubiquitin-proteasome system) on the basis of db/db mouse hearts and diabetic patients' hearts demonstrated that the diabetic ventricle presented a significant reduction in USP28 expression. Diabetic Myh6-Cre+/USP28fl/fl mice exhibited more severe progressive cardiac dysfunction, lipid accumulation, and mitochondrial disarrangement, compared with their controls. On the other hand, USP28 overexpression improved systolic and diastolic dysfunction and ameliorated cardiac hypertrophy and fibrosis in the diabetic heart. Adeno-associated virus serotype 9-USP28 diabetic mice also exhibited less lipid storage, reduced reactive oxygen species formation, and mitochondrial impairment in heart tissues than adeno-associated virus serotype 9-null diabetic mice. As a result, USP28 overexpression attenuated cardiac remodeling and dysfunction, lipid accumulation, and mitochondrial impairment in high-fat diet/streptozotocin-induced type 2 diabetes mice. These results were also confirmed in neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes. RNA sequencing, immunoprecipitation and mass spectrometry analysis, chromatin immunoprecipitation assays, chromatin immunoprecipitation sequencing, and protein pull-down assay mechanistically revealed that USP28 directly interacted with PPARα (peroxisome proliferator-activated receptor α), deubiquitinating and stabilizing PPARα (Lys152) to promote Mfn2 (mitofusin 2) transcription, thereby impeding mitochondrial morphofunctional defects. However, such cardioprotective benefits of USP28 were largely abrogated in db/db mice with PPARα deletion and conditional loss-of-function of Mfn2. CONCLUSIONS: Our findings provide a USP28-modulated mitochondria homeostasis mechanism that involves the PPARα-Mfn2 axis in diabetic hearts, suggesting that USP28 activation or adeno-associated virus therapy targeting USP28 represents a potential therapeutic strategy for diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Cardiomiopatías Diabéticas , Células Madre Pluripotentes Inducidas , Ubiquitina Tiolesterasa , Animales , Humanos , Ratones , Ratas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Lípidos , Ratones Noqueados , Miocitos Cardíacos/metabolismo , PPAR alfa/metabolismo , Estreptozocina/metabolismo , Estreptozocina/uso terapéutico , Ubiquitina Tiolesterasa/análisis , Ubiquitina Tiolesterasa/metabolismo
2.
J Mol Cell Cardiol ; 186: 81-93, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37995517

RESUMEN

AIM: Doxorubicin-induced cardiotoxicity (DIC) is an increasing problem, occurring in many cancer patients receiving anthracycline chemotherapy, ultimately leading to heart failure (HF). Unfortunately, DIC remains difficult to manage due to an ignorance regarding pathophysiological mechanisms. Our work aimed to evaluate the role of HSP47 in doxorubicin-induced HF, and to explore the molecular mechanisms. METHODS AND RESULTS: Mice were exposed to multi-intraperitoneal injection of doxorubicin (DOX, 4mg/kg/week, for 6 weeks continuously) to produce DIC. HSP47 expression was significantly upregulated in serum and in heart tissue in DOX-treated mice and in isolated cardiomyocytes. Mice with cardiac-specific HSP47 overexpression and knockdown were generated using recombinant adeno-associated virus (rAVV9) injection. Importantly, cardiac-specific HSP47 overexpression exacerbated cardiac dysfunction in DIC, while HSP47 knockdown prevented DOX-induced cardiac dysfunction, cardiac atrophy and fibrosis in vivo and in vitro. Mechanistically, we identified that HSP47 directly interacted with IRE1α in cardiomyocytes. Furthermore, we provided powerful evidence that HSP47-IRE1α complex promoted TXNIP/NLRP3 inflammasome and reinforced USP1-mediated NLRP3 ubiquitination. Moreover, NLRP3 deficiency in vivo conspicuously abolished HSP47-mediated cardiac atrophy and fibrogenesis under DOX condition. CONCLUSION: HSP47 was highly expressed in serum and cardiac tissue after doxorubicin administration. HSP47 contributed to long-term anthracycline chemotherapy-associated cardiac dysfunction in an NLRP3-dependent manner. HSP47 therefore represents a plausible target for future therapy of doxorubicin-induced HF.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Ratones , Humanos , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas del Choque Térmico HSP47/metabolismo , Cardiotoxicidad/metabolismo , Doxorrubicina/farmacología , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Miocitos Cardíacos/metabolismo , Antibióticos Antineoplásicos/efectos adversos , Atrofia/inducido químicamente , Atrofia/metabolismo , Atrofia/patología , Apoptosis , Estrés Oxidativo
3.
PLoS Biol ; 19(8): e3001304, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34437534

RESUMEN

Tumor necrosis factor receptor-1 (TNFR1) signaling, apart from its pleiotropic functions in inflammation, plays a role in embryogenesis as deficiency of varieties of its downstream molecules leads to embryonic lethality in mice. Caspase-8 noncleavable receptor interacting serine/threonine kinase 1 (RIPK1) mutations occur naturally in humans, and the corresponding D325A mutation in murine RIPK1 leads to death at early midgestation. It is known that both the demise of Ripk1D325A/D325A embryos and the death of Casp8-/- mice are initiated by TNFR1, but they are mediated by apoptosis and necroptosis, respectively. Here, we show that the defects in Ripk1D325A/D325A embryos occur at embryonic day 10.5 (E10.5), earlier than that caused by Casp8 knockout. By analyzing a series of genetically mutated mice, we elucidated a mechanism that leads to the lethality of Ripk1D325A/D325A embryos and compared it with that underlies Casp8 deletion-mediated lethality. We revealed that the apoptosis in Ripk1D325A/D325A embryos requires a scaffold function of RIPK3 and enzymatically active caspase-8. Unexpectedly, caspase-1 and caspase-11 are downstream of activated caspase-8, and concurrent depletion of Casp1 and Casp11 postpones the E10.5 lethality to embryonic day 13.5 (E13.5). Moreover, caspase-3 is an executioner of apoptosis at E10.5 in Ripk1D325A/D325A mice as its deletion extends life of Ripk1D325A/D325A mice to embryonic day 11.5 (E11.5). Hence, an unexpected death pathway of TNFR1 controls RIPK1 D325A mutation-induced lethality at E10.5.


Asunto(s)
Caspasa 8/fisiología , Desarrollo Embrionario , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Caspasas/metabolismo , Muerte Celular , Ratones , Cultivo Primario de Células , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
4.
Comput Biol Med ; 166: 107549, 2023 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-37839222

RESUMEN

To address the scarcity and class imbalance of abnormal electrocardiogram (ECG) databases, which are crucial in AI-driven diagnostic tools for potential cardiovascular disease detection, this study proposes a novel quantum conditional generative adversarial algorithm (QCGAN-ECG) for generating abnormal ECG signals. The QCGAN-ECG constructs a quantum generator based on patch method. In this method, each sub-generator generates distinct features of abnormal heartbeats in different segments. This patch-based generative algorithm conserves quantum resources and makes QCGAN-ECG practical for near-term quantum devices. Additionally, QCGAN-ECG introduces quantum registers as control conditions. It encodes information about the types and probability distributions of abnormal heartbeats into quantum registers, rendering the entire generative process controllable. Simulation experiments on Pennylane demonstrated that the QCGAN-ECG could generate completely abnormal heartbeats with an average accuracy of 88.8%. Moreover, the QCGAN-ECG can accurately fit the probability distribution of various abnormal ECG data. In the anti-noise experiments, the QCGAN-ECG showcased outstanding robustness across various levels of quantum noise interference. These results demonstrate the effectiveness and potential applicability of the QCGAN-ECG for generating abnormal ECG signals, which will further promote the development of AI-driven cardiac disease diagnosis systems. The source code is available at github.com/VanSWK/QCGAN_ECG.

5.
Eur J Pharmacol ; 940: 175482, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36587888

RESUMEN

BACKGROUND: Increased inflammatory mediators produced by inflamed cells are often connected with pressure-induced cardiac remodelling and heart failure. Interleukin-21 (IL-21) serves as an immunomodulator involved in multiple pathological processes, while the role of IL-21 in pressure-induced cardiac remodelling remains unclear. EXPERIMENT APPROACH: Cardiac function, CD4+T-cell infiltration, and IL-21 and IL-21 receptor expression levels were investigated in a pressure overload mouse model induced by aortic banding (AB) surgery. Western blotting and qPCR were used to detect the effects of IL-21 on inflammation, apoptosis, and fibrosis in the myocardium after AB surgery. In addition, the signal transduction mechanisms underlying these effects were investigated in vivo and in vitro by qPCR and western blotting. KEY RESULTS: IL-21 levels in mice rapidly increased in the acute phase after AB surgery. Compared with those in the control group, the transverse aortas of mice in the AB surgery group contracted. However, it must be noted that neutralizing IL-21 could reduce myocardial injury and remodelling, while the administration of exogenous IL-21 recombinant protein had the opposite effect. Mechanistically, we learned that IL-21 is effective in inducing the activation of tissue inhibitor of metalloproteinase 4 (TIMP4) and matrix metalloproteinase 9 (MMP-9) signalling in vitro and in vivo. We believe that increased activation and secretion of IL-21 and CD4+ T cells may contribute to stress overload-induced cardiac remodelling. CONCLUSION: These findings reveal a novel mechanism by which IL-21 stimulates myocardial inflammation, apoptosis, and fibrosis to induce stress-overload-induced myocardial remodelling by activating the TIMP4/MMP9 signalling pathway.


Asunto(s)
Metaloproteinasa 9 de la Matriz , Remodelación Ventricular , Ratones , Animales , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Noqueados , Miocardio/metabolismo , Transducción de Señal , Inflamación/metabolismo , Fibrosis , Miocitos Cardíacos , Ratones Endogámicos C57BL , Inhibidor Tisular de Metaloproteinasa-4
6.
Artículo en Inglés | MEDLINE | ID: mdl-37399158

RESUMEN

Electrocardiogram (ECG) is the main criterion for arrhythmia detection. As a means of identification, ECG leakage seems to be a common occurrence due to the development of the Internet of Medical Things (IoMT). The advent of the quantum era makes it difficult for classical blockchain technology to provide security for ECG data storage. Therefore, from the perspective of safety and practicality, this article proposes a quantum arrhythmia detection system named QADS, which achieves secure storage and sharing of ECG data based on quantum blockchain technology. Furthermore, a quantum neural network is used in QADS to recognize abnormal ECG data, which contributes to further cardiovascular disease diagnosis. Each quantum block stores the hash of the current and previous block to construct a quantum block network. The new quantum blockchain algorithm introduces a controlled quantum walk hash function and a quantum authentication protocol to guarantee legitimacy and security while creating new blocks. In addition, this article constructs a hybrid quantum convolutional neural network nameded HQCNN to extract the temporal features of ECG to detect abnormal heartbeats. The simulation experimental results show that HQCNN achieves an average training and testing accuracy of 94.7% and 93.6%. And the detection stability is much higher than classical CNN with the same structure. HQCNN also has certain robustness under the perturbation of quantum noise. Besides, this article demonstrates through mathematical analysis that the proposed quantum blockchain algorithm has strong security and can effectively resist various quantum attacks, such as external attacks, Entanglement-Measure attack and Interception-Measurement-Repeat attack.

7.
Acta Pharm Sin B ; 12(11): 4138-4153, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36386478

RESUMEN

Despite complications were significantly reduced due to the popularity of percutaneous coronary intervention (PCI) in clinical trials, reperfusion injury and chronic cardiac remodeling significantly contribute to poor prognosis and rehabilitation in AMI patients. We revealed the effects of HSP47 on myocardial ischemia-reperfusion injury (IRI) and shed light on the underlying molecular mechanism. We generated adult mice with lentivirus-mediated or miRNA (mi1/133TS)-aided cardiac fibroblast-selective HSP47 overexpression. Myocardial IRI was induced by 45-min occlusion of the left anterior descending (LAD) artery followed by 24 h reperfusion in mice, while ischemia-mediated cardiac remodeling was induced by four weeks of reperfusion. Also, the role of HSP47 in fibrogenesis was evaluated in cardiac fibroblasts following hypoxia-reoxygenation (HR). Extensive HSP47 was observed in murine infarcted hearts, human ischemic hearts, and cardiac fibroblasts and accelerated oxidative stress and apoptosis after myocardial IRI. Cardiac fibroblast-selective HSP47 overexpression exacerbated cardiac dysfunction caused by chronic myocardial IRI and presented deteriorative fibrosis and cell proliferation. HSP47 upregulation in cardiac fibroblasts promoted TGFß1-Smad4 pathway activation and Smad4 deubiquitination by recruiting ubiquitin-specific peptidase 10 (USP10) in fibroblasts. However, cardiac fibroblast specific USP10 deficiency abolished HSP47-mediated fibrogenesis in hearts. Moreover, blockage of HSP47 with Col003 disturbed fibrogenesis in fibroblasts following HR. Altogether, cardiac fibroblast HSP47 aggravates fibrosis post-myocardial IRI by enhancing USP10-dependent Smad4 deubiquitination, which provided a potential strategy for myocardial IRI and cardiac remodeling.

8.
EBioMedicine ; 86: 104359, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36395739

RESUMEN

BACKGROUND: Arachidonate 5-lipoxygenase (Alox5) belongs to a class of nonheme iron-containing dioxygenases involved in the catalysis of leukotriene biosynthesis. However, the effects of Alox5 itself on pathological cardiac remodeling and heart failure remain elusive. METHODS: The role of Alox5 in pathological cardiac remodeling was investigated by Alox5 genetic depletion, AAV9-mediated overexpression in cardiomyocytes, and a bone marrow (BM) transplantation approach. Neonatal rat cardiomyocytes were used to explore the effects of Alox5 in vitro. Molecular and signaling pathways were revealed by CUT &Tag, IP-MS, RNA sequencing and bioinformatic analyses. FINDINGS: Untargeted metabolomics showed that serum 5-HETE (a primary product of Alox5) levels were little changed in patients with cardiac hypertrophy, while Alox5 expression was significantly upregulated in murine hypertensive cardiac samples and human cardiac samples of hypertrophy, which prompted us to test whether high Alox5 levels under hypertensive stimuli were directly associated with pathologic myocardium in an enzymatic activity-independent manner. Herein, we revealed that Alox5 deficiency significantly ameliorated transverse aortic constriction (TAC)-induced hypertrophy. Cardiomyocyte-specific Alox5 depletion attenuated hypertensive ventricular remodeling. Conversely, cardiac-specifical Alox5 overexpression showed a pro-hypertrophic cardiac phenotype. Ablation of Alox5 in bone marrow-derived cells did not affect pathological cardiac remodeling and heart failure. Mechanically, Runx2 was identified as a target of Alox5. In this regard, Alox5 PEST domain could directly bind to Runx2 PTS domain, promoting nuclear localization of Runx2 in an enzymatic activity-independent manner, simultaneously contributed to liquid-liquid phase separation (LLPS) of Runx2 at specific domain in the nucleus and increased transcription of EGFR in cardiomyocytes. Runx2 depletion alleviated hypertrophy in Ang II-pretreated Alox5-overexpressing cardiomyocytes. INTERPRETATION: Overall, our study demonstrated that targeting Alox5 exerted a protective effect against cardiac remodeling and heart failure under hypertensive stimuli by disturbing LLPS of Runx2 and substantial reduction of EGFR transcription activation in cardiomyocytes. Our findings suggest that negative modulation of Alox5-Runx2 may provide a therapeutic approach against pathological cardiac remodeling and heart failure. FUNDING: National Natural Science Foundation of China.


Asunto(s)
Araquidonato 5-Lipooxigenasa , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Insuficiencia Cardíaca , Hipertensión , Remodelación Ventricular , Animales , Humanos , Ratones , Ratas , Araquidonato 5-Lipooxigenasa/genética , Cardiomegalia/genética , Cardiomegalia/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Insuficiencia Cardíaca/metabolismo , Hipertensión/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Remodelación Ventricular/genética
9.
J Am Heart Assoc ; 11(19): e026728, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36172969

RESUMEN

Background Mechanistic insights of glucagon-like peptide-1 receptor agonists remain incompletely identified, despite the efficacy in heart failure observed in clinical trials. Here, we evaluated the effects of dulaglutide on heart complications and illuminated its underlying mechanism. Methods and Results We used mice with high-fat diet (HFD)/streptozotocin-induced type 2 diabetes to investigate the effects of dulaglutide upon diabetic cardiac dysfunction. After the onset of diabetes, control and diabetic mice were injected subcutaneously with either dulaglutide (type 2 diabetes-dulaglutide and control-dulaglutide groups) or vehicle (type 2 diabetes-vehicle and control-vehicle groups) for 8 weeks. Subsequently, heart characteristics, cardiometabolic profile and mitochondrial morphology and function were evaluated. Also, we analyzed the effects of dulaglutide on neonatal rat ventricular myocytes treated with high glucose plus palmitic acid. In addition, wild type and AMP-activated protein kinase α2 mutant mice were used to evaluate the underlying mechanism. In type 2 diabetes mouse model, dulaglutide ameliorated insulin resistance, improved glucose tolerance, reduced hyperlipidemia, and promoted fatty acid use in the myocardium. Dulaglutide treatment functionally attenuated cardiac remodeling and dysfunction and promoted metabolic reprogramming in diabetic mice. Furthermore, dulaglutide improved mitochondria fragmentation in myocytes, and simultaneously reinstated mitochondrial morphology and function in diabetic hearts. We also found that dulaglutide preserved AMP-activated protein kinase α2-dependent mitochondrial homeostasis, and the protective effects of dulaglutide on diabetic heart was almost abated by AMP-activated protein kinase α2 knockout. Conclusions Dulaglutide prevents diabetic heart failure and favorably affects myocardial metabolic remodeling by impeding mitochondria fragmentation, and we suggest a potential strategy to develop a long-term activation of glucagon-like peptide-1 receptor-based therapy to treat diabetes associated cardiovascular complications.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Insuficiencia Cardíaca , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Péptidos Similares al Glucagón/análogos & derivados , Glucosa , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/prevención & control , Hipoglucemiantes/uso terapéutico , Fragmentos Fc de Inmunoglobulinas , Ratones , Ácido Palmítico , Ratas , Proteínas Recombinantes de Fusión/farmacología , Estreptozocina/uso terapéutico
10.
Front Pharmacol ; 12: 794982, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899356

RESUMEN

Background: As a plant-derived polycyclic phenolic carboxylic acid isolated from Salvia miltiorrhiza, lithospermic acid (LA) has been identified as the pharmacological management for neuroprotection and hepatoprotection. However, the role and mechanism of lithospermic acid in the pathological process of myocardial ischemia-reperfusion injury are not fully revealed. Methods: C57BL/6 mice were subjected to myocardial ischemia and reperfusion (MI/R) surgery and pretreated by LA (50 mg/kg, oral gavage) for six consecutive days before operation. The in vitro model of hypoxia reoxygenation (HR) was induced by hypoxia for 24 h and reoxygenation for 6 h in H9C2 cells, which were subsequently administrated with lithospermic acid (100 µM). Nrf2 siRNA and dorsomorphin (DM), an inhibitor of AMPKα, were used to explore the function of AMPKα/Nrf2 in LA-mediated effects. Results: LA pretreatment attenuates infarct area and decreases levels of TnT and CK-MB in plasm following MI/R surgery in mice. Echocardiography and hemodynamics indicate that LA suppresses MI/R-induced cardiac dysfunction. Moreover, LA ameliorates oxidative stress and cardiomyocytes apoptosis following MI/R operation or HR in vivo and in vitro. In terms of mechanism, LA selectively activates eNOS, simultaneously increases nuclear translocation and phosphorylation of Nrf2 and promotes Nrf2/HO-1 pathway in vivo and in vitro, while cardioprotection of LA is abolished by pharmacological inhibitor of AMPK or Nrf2 siRNA in H9C2 cells. Conclusion: LA protects against MI/R-induced cardiac injury by promoting eNOS and Nrf2/HO-1 signaling via phosphorylation of AMPKα.

11.
Free Radic Biol Med ; 166: 348-357, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33705958

RESUMEN

Sepsis rapidly contributed to multiorgan failure affecting most commonly of the cardiovascular and respiratory systems and yet there were no effective therapies. The current study aimed at providing evidence on the cardioprotection of suppression of 5-Lipoxygenase (5-Lox) and identifying the possible mechanism in the mouse model of sepsis. The cecal ligation-perforation (CLP) model was applied to C57BL/6 wild-type (WT) and 5-Lox deficient (5-Lox-/-) mice to induce sepsis. 5-Lox expression was up-regulated in mouse myocardium and leukotriene B4 (LTB4) level was increased in serum after sepsis. Subsequently, we utilized a recombinant adenoviral expression vector (rAAV9) to overexpress Alox5 gene in adult mice. Compared to WT mice, 5-Lox overexpression accelerated CLP-induced myocardial injury and cardiac dysfunction. Oppositely, 5-Lox deficiency offered protection against myocardial injury in a mouse model of sepsis and attenuated sepsis-mediated inflammation, oxidative stress and apoptosis in the mouse heart. Mechanically, 5-Lox promoted LTB4 production, which in turn contributed to the activation of leukotriene B4 receptor 1 (BLT1)/interleukin-12p35 (IL-12p35) pathway and enhanced M1 macrophage polarization. However, the suppression of BLT1 by either gene mutation or antagonist U75302 significantly inhibited the adverse effect of 5-Lox in sepsis. Further study demonstrated that pharmacological inhibition of 5-Lox prevented CLP-induced septic cardiomyopathy (SCM). Our study identified 5-Lox exacerbated sepsis-associated myocardial injury through activation of LTB4 production and promoting BLT1/IL-12p35 pathway. Hence, inhibition of 5-Lox may be a potential candidate strategy for septic cardiac dysfunction treatment.


Asunto(s)
Receptores de Leucotrieno B4 , Sepsis , Animales , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Regulación hacia Abajo , Subunidad p35 de la Interleucina-12 , Ratones , Ratones Endogámicos C57BL , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/metabolismo , Sepsis/complicaciones , Sepsis/tratamiento farmacológico
12.
ACS Appl Mater Interfaces ; 11(50): 46783-46791, 2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31769644

RESUMEN

Solid polymer electrolytes can be used to construct solid-state lithium batteries (SSLBs) using lithium metals as the anode. However, the lifespan and safety problems of SSLBs caused by lithium dendrite growth have hindered their practical application. Here, we have designed and prepared a rigid-flexible asymmetric solid electrolyte (ASE) that is used in building SSLBs. The ASE can inhibit efficiently the growth of lithium dendrites and lead to a long cycle life of SSLBs due to the hierarchical structure of a combination of "polymer-in-ceramic" (i.e., rigid ceramic layer of Li6.4La3Zr1.4Ta0.6O12) and "LiBOB-in-polymer" (i.e., soft polymer-layer of polyethylene oxide and LiBOB components). The results demonstrated that a symmetrical battery with ASE (Li|ASE|Li) can be steadily cycled for more than 2000 h and yielded a flat plating/stripping voltage profile under a current density of 0.1 mA cm-2. As a consequence, the SSLB of LiFePO4|ASE|Li delivered a specific capacity of 155.1 mA h g-1 with a capacity retention rate up to 90.2% after 200 cycles with the Coulombic efficiency over 99.6% per cycle. This asymmetric structure combines the advantages of ceramics and polymers, providing an ingenious solution for building rigid and flexible solid electrolytes.

13.
Huan Jing Ke Xue ; 38(7): 2718-2727, 2017 Jul 08.
Artículo en Zh | MEDLINE | ID: mdl-29964610

RESUMEN

A continuous measurement was conducted in Yixing city urban area from 24th August to 15th September using TH-300B continuous online GC-MS instrument during G20 summit in Hangzhou, 2016. The VOCs average mass concentrations of alkane, alkene, aromatic, acetylene, haloalkane hydrocarbons, OVOC and acetonitrile were 11.00×10-9, 1.93×10-9, 5.78×10-9, 1.23×10-9, 4.16×10-9, 10.37×10-9, 0.27×10-9, respectively. The photochemical reaction activity was calculated by using the maximum potential coefficient of Ozone Formation Potential. Alkene and aromatic hydrocarbons were the most active components of OFP. By applying the positive matrix factorization(PMF)model, five major factors were extracted to identify the sources of NMHCs in Yixing city, including industry(42.2%),vehicle exhaust(17.9%), fuel evaporation(20.8%), paint/solvent usage(7.0%)and plant(12.1%). Combined with the conditional probability function(CPF) analysis, source of anthropogenic pollution was related to the distribution of industrial enterprises in the northwest and southeast, while the plant source was related to the forest hilly region of Southwest Yixing city. The effect of air pollutant emission reduction showed that the primary emission air pollutants had declined significantly during the strict control period from 1th to 6th September in G20 summit,2016, and the industry proportion was reduced to 30.5%, whereas the plant proportion increased to 16.8%.

14.
Mol Cell Biol ; 37(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27920255

RESUMEN

Oligomerization of the mixed-lineage kinase domain-like protein (MLKL) is essential for its cation channel function in necroptosis. Here we show that the MLKL channel is an octamer comprising two previously identified tetramers most likely in their side-by-side position. Intermolecule disulfide bonds are present in the tetramer but are not required for octamer assembly and necroptosis. MLKL forms oligomers in the necrosome and is then released from the necrosome before or during its membrane translocation. We identified two MLKL mutants that could not oligomerize into octamers, although they formed a tetramer, and also, one MLKL mutant could spontaneously form a disulfide bond-linked octamer. Subsequent analysis revealed that the tetramers fail to translocate to the plasma membrane and that the MLKL octamer formation depends on α-helices 4 and 5. While MLKL could be detected from outside the cells, its N- and C-terminal ends could not be detected, indicating that the MLKL octamer spans across the plasma membrane, leaving its N and C termini inside the cell. These data allowed us to propose a 180° symmetry model of the MLKL octamer and conclude that the fully assembled MLKL octamers, but not the previously described tetramers, act as effectors of necroptosis.


Asunto(s)
Apoptosis , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Multimerización de Proteína , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Disulfuros/metabolismo , Células HeLa , Humanos , Ratones , Modelos Biológicos , Necrosis , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA