heart regeneration

心脏再生
  • 文章类型: Journal Article
    Cardiomyocyte proliferation is a challenging metric to assess. Current methodologies have limitations in detecting the generation of new cardiomyocytes and technical challenges that reduce widespread applicability. Here, we describe an improved cell suspension and imaging-based methodology that can be broadly employed to assess cardiomyocyte cell division in standard laboratories across a multitude of model organisms and experimental conditions. We highlight additional metrics that can be gathered from the same cell preparations to enable additional relevant analyses to be performed. We incorporate additional antibody stains to address potential technical concerns of miscounting. Finally, we employ this methodology with a dual-thymidine analog-labeling approach to a post-infarction murine model, which allowed us to robustly identify unique cycling events, such as cardiomyocytes undergoing multiple rounds of cell division.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心血管疾病(CVD)是一个重大的全球健康挑战。仍然是全球疾病和死亡的主要原因。成人心脏有限的再生能力在修复由心肌梗塞等疾病引起的广泛损伤方面构成了主要障碍。为了应对这些挑战,纳米医学已成为一个有希望的领域,旨在通过创新的药物输送策略改善治疗效果。纳米载体,如纳米粒子(NPs),通过促进治疗剂直接向心脏的靶向递送提供革命性的方法。这种精确的递送系统通过解决潜在的机制,如炎症,在治疗各种心脏疾病方面具有巨大的潜力。氧化应激,细胞死亡,细胞外基质重塑,促生存信号,以及与缺血再灌注损伤相关的血管生成途径。在这次审查中,我们提供了与心脏重塑和再生有关的基本机制的简明摘要。我们探索基于纳米颗粒的药物递送系统如何有效地靶向上述机制。此外,我们讨论了临床试验,利用纳米颗粒为基础的药物输送系统,专门为心脏应用设计。这些试验证明了纳米医学在临床环境中的潜力,通过精确和有效的药物递送,为心脏疗法的未来发展铺平了道路。总的来说,纳米医学通过提供针对心脏损伤的复杂病理生理学的有针对性和有效的治疗策略,有望彻底改变心血管疾病的治疗前景。
    Cardiovascular disease (CVD) represents a significant global health challenge, remaining the leading cause of illness and mortality worldwide. The adult heart\'s limited regenerative capacity poses a major obstacle in repairing extensive damage caused by conditions like myocardial infarction. In response to these challenges, nanomedicine has emerged as a promising field aimed at improving treatment outcomes through innovative drug delivery strategies. Nanocarriers, such as nanoparticles (NPs), offer a revolutionary approach by facilitating targeted delivery of therapeutic agents directly to the heart. This precise delivery system holds immense potential for treating various cardiac conditions by addressing underlying mechanisms such as inflammation, oxidative stress, cell death, extracellular matrix remodeling, prosurvival signaling, and angiogenic pathways associated with ischemia-reperfusion injury. In this review, we provide a concise summary of the fundamental mechanisms involved in cardiac remodeling and regeneration. We explore how nanoparticle-based drug delivery systems can effectively target the afore-mentioned mechanisms. Furthermore, we discuss clinical trials that have utilized nanoparticle-based drug delivery systems specifically designed for cardiac applications. These trials demonstrate the potential of nanomedicine in clinical settings, paving the way for future advancements in cardiac therapeutics through precise and efficient drug delivery. Overall, nanomedicine holds promise in revolutionizing the treatment landscape of cardiovascular diseases by offering targeted and effective therapeutic strategies that address the complex pathophysiology of cardiac injuries.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    哺乳动物心肌细胞具有有限的再生能力。心脏病,比如先天性心脏病和心肌梗塞,通过调节细胞死亡(RCD)引起心肌细胞的初始损失。了解在受损心肌中控制RCD的机制对于开发促进心脏再生的疗法至关重要。我们以前报道过铁死亡,一种非凋亡和铁依赖性的RCD,是受伤心脏心肌细胞死亡的主要原因。为了研究心肌细胞铁凋亡偏好的机制,我们研究了抗铁蛋白试剂在梗塞小鼠心脏中的作用。结果表明,抗铁蛋白试剂不能改善新生儿心脏再生,并进一步损害青少年心脏的心脏功能。另一方面,铁细胞心肌细胞在伤口愈合过程中通过释放促血管生成因子发挥支持作用。再生小鼠心脏中铁死亡的抑制改变了免疫和血管生成反应。我们的研究提供了对应激心肌细胞中铁性凋亡优于其他类型RCD的见解,以及设计用于治疗心脏病的抗细胞死亡疗法的指导。
    Mammalian cardiomyocytes have limited regenerative ability. Cardiac disease, such as congenital heart disease and myocardial infarction, causes an initial loss of cardiomyocytes through regulated cell death (RCD). Understanding the mechanisms that govern RCD in the injured myocardium is crucial for developing therapeutics to promote heart regeneration. We previously reported that ferroptosis, a non-apoptotic and iron-dependent form of RCD, is the main contributor to cardiomyocyte death in the injured heart. To investigate the mechanisms underlying the preference for ferroptosis in cardiomyocytes, we examined the effects of anti-ferroptotic reagents in infarcted mouse hearts. The results revealed that the anti-ferroptotic reagent did not improve neonatal heart regeneration, and further compromised the cardiac function of juvenile hearts. On the other hand, ferroptotic cardiomyocytes played a supportive role during wound healing by releasing pro-angiogenic factors. The inhibition of ferroptosis in the regenerating mouse heart altered the immune and angiogenic responses. Our study provides insights into the preference for ferroptosis over other types of RCD in stressed cardiomyocytes, and guidance for designing anti-cell-death therapies for treating heart disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    与人类和其他哺乳动物不同,斑马鱼在一生中都表现出非凡的能力来再生受伤的心脏。线粒体脂肪酸β-氧化(FAO)有助于成人心脏在生理条件下的主要能量需求;然而,其在调节心脏再生中的功能和潜在机制尚未完全了解。针对粮农组织的不同战略产生了不同的结果。这里,我们证明了用米膦酸盐(MD)对线粒体FAO的药理抑制作用会导致斑马鱼幼虫中的脂质积累并抑制心室再生。MD治疗阻碍心源性因子再激活和心肌细胞(CM)增殖,补充外源性左旋肉碱可以挽救受损的心室再生。此外,与野生型鱼的消融心脏相比,心室再生,在肉碱棕榈酰转移酶-1b(cpt1b)基因敲除斑马鱼的消融心脏中,心源性因子再激活和CM增殖被显着阻断。进一步的实验表明,NF-κB信号传导和炎症增加可能与系统性线粒体FAO抑制引起的心室再生障碍有关。总的来说,我们的研究证明了线粒体FAO在斑马鱼心室再生中的重要作用,并重申了FAO在不同损伤模型和FAO抑制手段方面在心脏再生中的复杂和多方面的作用。
    Unlike humans and other mammals, zebrafish demonstrate a remarkable capacity to regenerate their injured hearts throughout life. Mitochondrial fatty acid β-oxidation (FAO) contributes to major energy demands of the adult hearts under physiological conditions; however, its functions in regulating cardiac regeneration and the underlying mechanisms are not completely understood. Different strategies targeting FAO have yield mixed outcomes. Here, we demonstrated that pharmacological inhibition of mitochondrial FAO with mildronate (MD) caused lipid accumulation in zebrafish larvae and suppressed ventricle regeneration. MD treatment impeded cardiogenic factor reactivation and cardiomyocyte (CM) proliferation, and impaired ventricle regeneration could be rescued by exogenous l-carnitine supplementation. Moreover, compared with the ablated hearts of wild-type fish, ventricle regeneration, cardiogenic factor reactivation and CM proliferation were significantly blocked in the ablated hearts of carnitine palmitoyltransferase-1b (cpt1b) knockout zebrafish. Further experiments suggested that NF-κB signaling and increased inflammation may be involved in the impediment of ventricle regeneration caused by systemic mitochondrial FAO inhibition. Overall, our study demonstrates the essential roles of mitochondrial FAO in zebrafish ventricle regeneration and reaffirms the sophisticated and multifaceted roles of FAO in heart regeneration with regard to different injury models and means of FAO inhibition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:干细胞衍生的细胞外囊泡(EV)是一类具有优异生物相容性的新兴治疗剂,生物活性和促再生能力。基于EV的药物的潜在目标之一是心血管疾病(CVD)。在这项工作中,我们使用了在不同氧气浓度(21、5和3%O2)下培养的人诱导多能干细胞(hiPSCs;hiPPS-EV)衍生的EV来剖析负责心脏保护的分子机制。
    方法:通过超滤结合尺寸排阻色谱(UFSEC)分离电动汽车,然后通过纳米粒子跟踪分析进行表征,原子力显微镜(AFM)和蛋白质印迹方法。液相色谱和串联质谱结合生物信息学分析用于鉴定各种氧气条件下的差异富集蛋白。我们直接比较了这些EV在心肌细胞(CM)损伤的氧-葡萄糖剥夺/复氧(OGD/R)模型中的心脏保护作用。利用先进的分子生物学,荧光显微镜,原子力谱和生物信息学技术,我们研究了参与细胞存活调节的细胞内信号通路,细胞凋亡和抗氧化反应。在用ML385抑制NRF2后的CM中评估EV对NRF2调节的信号传导的直接作用。
    结果:我们证明,与其他测试的氧气条件(常氧;EV-N和低氧3%O2;EV-H3)衍生自5%O2的生理性缺氧(EV-H5)的hiPS-EV对受损CM的细胞保护功能增强。这是由于转移后受体细胞中Akt激酶的磷酸化率较高,调节AMPK活性和减少细胞凋亡。此外,我们提供了使用AFM测量的EV-H5治疗的CM的钙信号改善和持续收缩性的直接证据.机械上,我们的质谱和生物信息学分析显示,EV-H5中的差异富集蛋白与NRF2调节的抗氧化途径相关.在这方面,EV-H5在OGD/R时增加NRF2蛋白的核易位并增强其在CMs中的转录。相比之下,用ML385抑制NRF2消除了电动汽车对CMs的保护作用。
    结论:在这项工作中,与EV-N和EV-H3相比,我们证明了EV-H5具有更好的心脏保护功能.这样的电动汽车在恢复应力CM的氧化还原平衡方面最有效,保持其收缩功能并防止细胞死亡。我们的数据支持生理性缺氧产生的hiPS-EV的潜在用途,作为具有再生特性的无细胞疗法,用于治疗心脏病。
    BACKGROUND: Stem cell-derived extracellular vesicles (EVs) are an emerging class of therapeutics with excellent biocompatibility, bioactivity and pro-regenerative capacity. One of the potential targets for EV-based medicines are cardiovascular diseases (CVD). In this work we used EVs derived from human induced pluripotent stem cells (hiPSCs; hiPS-EVs) cultured under different oxygen concentrations (21, 5 and 3% O2) to dissect the molecular mechanisms responsible for cardioprotection.
    METHODS: EVs were isolated by ultrafiltration combined with size exclusion chromatography (UF + SEC), followed by characterization by nanoparticle tracking analysis, atomic force microscopy (AFM) and Western blot methods. Liquid chromatography and tandem mass spectrometry coupled with bioinformatic analyses were used to identify differentially enriched proteins in various oxygen conditions. We directly compared the cardioprotective effects of these EVs in an oxygen-glucose deprivation/reoxygenation (OGD/R) model of cardiomyocyte (CM) injury. Using advanced molecular biology, fluorescence microscopy, atomic force spectroscopy and bioinformatics techniques, we investigated intracellular signaling pathways involved in the regulation of cell survival, apoptosis and antioxidant response. The direct effect of EVs on NRF2-regulated signaling was evaluated in CMs following NRF2 inhibition with ML385.
    RESULTS: We demonstrate that hiPS-EVs derived from physiological hypoxia at 5% O2 (EV-H5) exert enhanced cytoprotective function towards damaged CMs compared to EVs derived from other tested oxygen conditions (normoxia; EV-N and hypoxia 3% O2; EV-H3). This resulted from higher phosphorylation rates of Akt kinase in the recipient cells after transfer, modulation of AMPK activity and reduced apoptosis. Furthermore, we provide direct evidence for improved calcium signaling and sustained contractility in CMs treated with EV-H5 using AFM measurements. Mechanistically, our mass spectrometry and bioinformatics analyses revealed differentially enriched proteins in EV-H5 associated with the antioxidant pathway regulated by NRF2. In this regard, EV-H5 increased the nuclear translocation of NRF2 protein and enhanced its transcription in CMs upon OGD/R. In contrast, inhibition of NRF2 with ML385 abolished the protective effect of EVs on CMs.
    CONCLUSIONS: In this work, we demonstrate a superior cardioprotective function of EV-H5 compared to EV-N and EV-H3. Such EVs were most effective in restoring redox balance in stressed CMs, preserving their contractile function and preventing cell death. Our data support the potential use of hiPS-EVs derived from physiological hypoxia, as cell-free therapeutics with regenerative properties for the treatment of cardiac diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:心肌缺血再灌注损伤(MIRI)定义为短暂中断血液供应后恢复到心肌的血流,对缺血心肌造成更严重的损害。然而,目前,再灌注疗法是缺血性心肌病的首选疗法,这无疑会导致MIRI,因此,它已成为影响冠状动脉疾病预后的挑战性问题。
    方法:在WebofScienceCoreCollection数据库中检索了2000年1月1日至2023年10月1日发表的与MIRI治疗相关的论文。使用VOSviewer和CiteSpace进行了文献计量分析,以阐明进展和热点。
    结果:来自64个国家的3304篇论文,2134个研究机构和13228名作者参加了这项研究。其中,中国贡献的论文最多,影响最大,而美国拥有最广泛的伙伴关系。第四军医大学是主要研究机构。最有价值的作者包括Chattipakorn,尼蓬,Chattipakorn,Siripornc,杨,简和杨,杨。
    结论:在过去的20年里,MIRI疗法的研究取得了重大进展。需要进一步的研究来探索各种治疗方案之间的相互作用。未来的调查将强调纳米载体,心脏再生,和干细胞疗法。我们的研究从文献计量的角度确定了MIRI的研究热点,预测未来趋势,并为MIRI治疗研究提供了新的见解。
    BACKGROUND: Myocardial ischemia-reperfusion injury (MIRI) is defined as the restoration of blood flow to the myocardium after a brief interruption of blood supply, causing more severe damage to the ischemic myocardium. However, currently, reperfusion therapy is the preferred therapy for ischemic cardiomyopathy, which undoubtedly causes MIRI, and thus it has become a challenging issue affecting the prognosis of coronary artery disease.
    METHODS: A search was conducted in the Web of Science Core Collection database for papers relevant to MIRI therapy published between 1 January 2000 and 1 October 2023. Bibliometric analyses were performed using VOSviewer and CiteSpace to elucidate the progress and hotspots.
    RESULTS: 3304 papers from 64 countries, 2134 research institutions and 13,228 authors were enrolled in the study. Of these, China contributed the most papers and had the biggest impact, while the United States had the most extensive partnership. The Fourth Military Medical University was the primary research institution. The most valuable authors include Chattipakorn, Nipon, Chattipakorn, Siriporn c, Yang, Jian and Yang, Yang.
    CONCLUSIONS: Over the past 20 years, research on MIRI therapies has made significant strides. Further studies are necessary to explore the interactions between various therapeutic options. Future investigations will emphasize nanocarriers, cardiac regeneration, and stem cell therapies. Our study identifies MIRI research hotspots from a bibliometric perspective, forecasts future trends, and offers fresh insights into MIRI therapy research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    心血管疾病是世界范围内死亡的主要原因,心肌梗塞最为普遍。目前,目前尚无治疗方法可以预防或逆转心肌梗死后心肌细胞的大量死亡.成年哺乳动物的心脏显示出有限的再生能力,但不足以让心肌完全恢复.相比之下,受损的斑马鱼心肌通过先前存在的心肌细胞的强大增殖而有效再生。因此,斑马鱼可以利用它来研究心脏再生背后的遗传程序,可能存在,尽管处于休眠状态,在成年人的心脏。为此,我们建立了ZebraReg,一种新型的多功能自动化平台,用于研究斑马鱼幼虫心肌细胞特定消融后的心脏再生动力学。结合自动心脏成像,该平台可以与遗传或药理学方法整合,并用于中等通量筛选假定的心脏再生调节剂。我们通过鉴定基因和药物的抗再生和促再生作用来证明平台的多功能性。总之,我们提出了一种工具,可以用来简化再生的新基因调节因子的目标验证过程,以及发现新的药物疗法来使心肌梗死后的心脏再生。
    Cardiovascular disease is the leading cause of death worldwide with myocardial infarction being the most prevalent. Currently, no cure is available to either prevent or revert the massive death of cardiomyocytes that occurs after a myocardial infarction. Adult mammalian hearts display a limited regeneration capacity, but it is insufficient to allow complete myocardial recovery. In contrast, the injured zebrafish heart muscle regenerates efficiently through robust proliferation of pre-existing myocardial cells. Thus, zebrafish allows its exploitation for studying the genetic programs behind cardiac regeneration, which may be present, albeit dormant, in the adult human heart. To this end, we have established ZebraReg, a novel and versatile automated platform for studying heart regeneration kinetics after the specific ablation of cardiomyocytes in zebrafish larvae. In combination with automated heart imaging, the platform can be integrated with genetic or pharmacological approaches and used for medium-throughput screening of presumed modulators of heart regeneration. We demonstrate the versatility of the platform by identifying both anti- and pro-regenerative effects of genes and drugs. In conclusion, we present a tool which may be utilised to streamline the process of target validation of novel gene regulators of regeneration, and the discovery of new drug therapies to regenerate the heart after myocardial infarction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    氧化磷酸化(OXPHOS)系统是复杂的组织,呼吸复合物形成超组装的四级结构,其组装机制和生理作用仍在研究中。Cox7a2l,也称为Scaf1,促进复杂III和复杂IV(CIII-CIV)超组装,提高各种物种的能量效率。我们检查了Cox7a1的作用,另一个Cox7a家族成员,在超复合物组装和肌肉生理学中。缺乏Cox7a1的斑马鱼表现出减少的CIV2形成,代谢改变,和非病理性肌肉性能下降。此外,cox7a1-/-心脏表现出促进再生的代谢特征,影响心脏再生反应。cox7a1-/-和cox7a2l-/-的独特表型效应强调了受损的超复合物形成的不同代谢和生理后果,强调Cox7a1在OXPHOS系统内肌肉成熟中的重要性。
    The oxidative phosphorylation (OXPHOS) system is intricately organized, with respiratory complexes forming super-assembled quaternary structures whose assembly mechanisms and physiological roles remain under investigation. Cox7a2l, also known as Scaf1, facilitates complex III and complex IV (CIII-CIV) super-assembly, enhancing energetic efficiency in various species. We examined the role of Cox7a1, another Cox7a family member, in supercomplex assembly and muscle physiology. Zebrafish lacking Cox7a1 exhibited reduced CIV2 formation, metabolic alterations, and non-pathological muscle performance decline. Additionally, cox7a1-/- hearts displayed a pro-regenerative metabolic profile, impacting cardiac regenerative response. The distinct phenotypic effects of cox7a1-/- and cox7a2l-/- underscore the diverse metabolic and physiological consequences of impaired supercomplex formation, emphasizing the significance of Cox7a1 in muscle maturation within the OXPHOS system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:人类心肌梗塞(MI)后的愈合过程涉及复杂的事件,这些事件将受损组织替换为纤维化疤痕。受影响的心脏组织可能永久丧失其功能。相比之下,斑马鱼显示出显着的无疤痕心脏再生能力。先前的研究表明,syndecan-4(SDC4)调节高等脊椎动物心脏损伤后的炎症反应和成纤维细胞活性。然而,Sdc4是否以及如何调节高度再生斑马鱼的心脏再生仍然未知。
    结果:这项研究表明,通过转录分析,sdc4表达在斑马鱼心脏再生过程中受到差异调节。具体来说,sdc4表达在心室冷冻损伤的早期再生阶段迅速和短暂增加。此外,sdc4的敲低导致细胞外基质蛋白沉积的显著减少,免疫细胞积累,和病变部位的细胞增殖。tgfb1a和col1a1a的表达式,以及纤维连接蛋白的表达,在sdc4敲低下都下调。此外,通过体内心电图分析,我们证实sdc4的表达是斑马鱼心脏修复所必需的。sdc4表达缺失导致明显的病理性Q波和ST段抬高,这是人类MI患者的体征。
    结论:我们的发现支持Sdc4在斑马鱼心脏再生的早期阶段是介导多效性修复反应所必需的。
    BACKGROUND: The healing process after a myocardial infarction (MI) in humans involves complex events that replace damaged tissue with a fibrotic scar. The affected cardiac tissue may lose its function permanently. In contrast, zebrafish display a remarkable capacity for scar-free heart regeneration. Previous studies have revealed that syndecan-4 (SDC4) regulates inflammatory response and fibroblast activity following cardiac injury in higher vertebrates. However, whether and how Sdc4 regulates heart regeneration in highly regenerative zebrafish remains unknown.
    RESULTS: This study showed that sdc4 expression was differentially regulated during zebrafish heart regeneration by transcriptional analysis. Specifically, sdc4 expression increased rapidly and transiently in the early regeneration phase upon ventricular cryoinjury. Moreover, the knockdown of sdc4 led to a significant reduction in extracellular matrix protein deposition, immune cell accumulation, and cell proliferation at the lesion site. The expression of tgfb1a and col1a1a, as well as the protein expression of Fibronectin, were all down-regulated under sdc4 knockdown. In addition, we verified that sdc4 expression was required for cardiac repair in zebrafish via in vivo electrocardiogram analysis. Loss of sdc4 expression caused an apparent pathological Q wave and ST elevation, which are signs of human MI patients.
    CONCLUSIONS: Our findings support that Sdc4 is required to mediate pleiotropic repair responses in the early stage of zebrafish heart regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    斑马鱼在受损后具有终生的心脏再生能力,而哺乳动物在出生后早期发育过程中会失去这种能力。这项研究调查了出生后哺乳动物发育过程中生长因子表达的下降是否有助于心肌细胞再生潜力的降低。除了证实神经调节素1(NRG1)的增殖能力外,白细胞介素(IL)1b,核因子κB受体活化因子配体(RANKL),胰岛素生长因子(IGF)2和IL6,我们确定了其他潜在的促再生因子,BMP7表现出最显著的功效。新生小鼠心肌细胞Bmp7敲低和成年斑马鱼在心脏再生过程中功能丧失减少心肌细胞增殖,表明Bmp7在小鼠和斑马鱼心脏的再生阶段至关重要。相反,再生斑马鱼中的bmp7过表达或在有丝分裂后幼年和成年小鼠阶段施用,心肌梗死后的体外和体内,增强心肌细胞循环。机械上,BMP7通过BMPR1A/ACVR1和ACVR2A/BMPR2受体和下游SMAD5,ERK,和AKT信号。总的来说,BMP7给药是一种有希望的心脏再生策略。
    Zebrafish have a lifelong cardiac regenerative ability after damage, whereas mammals lose this capacity during early postnatal development. This study investigated whether the declining expression of growth factors during postnatal mammalian development contributes to the decrease of cardiomyocyte regenerative potential. Besides confirming the proliferative ability of neuregulin 1 (NRG1), interleukin (IL)1b, receptor activator of nuclear factor kappa-Β ligand (RANKL), insulin growth factor (IGF)2, and IL6, we identified other potential pro-regenerative factors, with BMP7 exhibiting the most pronounced efficacy. Bmp7 knockdown in neonatal mouse cardiomyocytes and loss-of-function in adult zebrafish during cardiac regeneration reduced cardiomyocyte proliferation, indicating that Bmp7 is crucial in the regenerative stages of mouse and zebrafish hearts. Conversely, bmp7 overexpression in regenerating zebrafish or administration at post-mitotic juvenile and adult mouse stages, in vitro and in vivo following myocardial infarction, enhanced cardiomyocyte cycling. Mechanistically, BMP7 stimulated proliferation through BMPR1A/ACVR1 and ACVR2A/BMPR2 receptors and downstream SMAD5, ERK, and AKT signaling. Overall, BMP7 administration is a promising strategy for heart regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号