induced pluripotent stem cells

诱导多能干细胞
  • 文章类型: Journal Article
    核膜(NE)蛋白层粘连蛋白A/C(由LMNA编码)中的突变,引起严重形式的扩张型心肌病(DCM),伴有早发性危及生命的心律失常。然而,LMNA相关DCM(LMNA-DCM)中心律失常发生增加的分子机制仍然未知.在这里,我们显示LMNA中的移码突变会导致异常的Ca2处理,LMNA-DCM患者特异性iPSC衍生心肌细胞(iPSC-CMs)中的心律失常和NE畸形。机械上,层粘连蛋白A与沉默蛋白1(SIRT1)相互作用,其中突变体层粘连蛋白A/C加速SIRT1的降解,导致线粒体功能障碍和氧化应激。升高的活性氧(ROS)然后激活Ca2/钙调蛋白依赖性蛋白激酶II(CaMKII)-ryanodine受体2(RYR2)途径,并加剧突变体iPSC-CM中SUN1的积累,导致心律失常和NE变形,分别。一起来看,显示laminA/C缺乏介导的ROS障碍是LMNA-DCM发展的核心。操纵受损的SIRT1活性和过度的氧化应激是LMNA-DCM的潜在未来治疗策略。
    Mutations in the nuclear envelope (NE) protein lamin A/C (encoded by LMNA), cause a severe form of dilated cardiomyopathy (DCM) with early-onset life-threatening arrhythmias. However, molecular mechanisms underlying increased arrhythmogenesis in LMNA-related DCM (LMNA-DCM) remain largely unknown. Here we show that a frameshift mutation in LMNA causes abnormal Ca2+ handling, arrhythmias and disformed NE in LMNA-DCM patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). Mechanistically, lamin A interacts with sirtuin 1 (SIRT1) where mutant lamin A/C accelerates degradation of SIRT1, leading to mitochondrial dysfunction and oxidative stress. Elevated reactive oxygen species (ROS) then activates the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-ryanodine receptor 2 (RYR2) pathway and aggravates the accumulation of SUN1 in mutant iPSC-CMs, contributing to arrhythmias and NE deformation, respectively. Taken together, the lamin A/C deficiency-mediated ROS disorder is revealed as central to LMNA-DCM development. Manipulation of impaired SIRT1 activity and excessive oxidative stress is a potential future therapeutic strategy for LMNA-DCM.
    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
    免疫系统,充当身体的“国防军”,在监视中发挥作用,防御。免疫系统的任何破坏都可能导致免疫相关疾病的发展。广泛的研究表明间充质干细胞(MSCs)在这些疾病中至关重要的免疫调节作用。特别感兴趣的是在特定条件下诱导体细胞的能力,产生一种具有干细胞特征的新细胞类型,称为诱导多能干细胞(iPSC)。iPSCs向MSCs的分化,特异性诱导多能干细胞来源的间充质干细胞(iMSCs),作为解决MSC挑战的潜在解决方案,可能作为传统药物疗法的替代品。此外,iMSC的产品,称为诱导多能干细胞来源的间充质干细胞来源的细胞外囊泡(iMSC-EV),可能表现出类似于iMSC的功能。由于电动汽车的生物学优势,它们已成为“无细胞疗法”的焦点。这里,我们全面总结了iMSCs对免疫细胞的生物学影响,探讨了iMSC和iMSC-EV在疾病中的应用,并简要讨论了电动汽车的基本特征。最后,我们概述了目前与iMSC和iMSC-EV相关的优势和挑战.我们希望这篇与iMSCs和iMSC-EV相关的综述将有助于开发新的疾病治疗方法。
    The immune system, functioning as the body\'s \"defense army\", plays a role in surveillance, defense. Any disruptions in immune system can lead to the development of immune-related diseases. Extensive researches have demonstrated the crucial immunoregulatory role of mesenchymal stem cells (MSCs) in these diseases. Of particular interest is the ability to induce somatic cells under specific conditions, generating a new cell type with stem cell characteristics known as induced pluripotent stem cell (iPSC). The differentiation of iPSCs into MSCs, specifically induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs), hold promise as a potential solution to the challenges of MSCs, potentially serving as an alternative to traditional drug therapies. Moreover, the products of iMSCs, termed induced pluripotent stem cell-derived mesenchymal stem cell-derived extracellular vesicles (iMSC-EVs), may exhibit functions similar to iMSCs. With the biological advantages of EVs, they have become the focus of \"cell-free therapy\". Here, we provided a comprehensive summary of the biological impact of iMSCs on immune cells, explored the applications of iMSCs and iMSC-EVs in diseases, and briefly discussed the fundamental characteristics of EVs. Finally, we overviewed the current advantages and challenges associated with iMSCs and iMSC-EVs. It is our hope that this review related to iMSCs and iMSC-EVs will contribute to the development of new approaches for the treatment of 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
    Leber的遗传性视神经病变(LHON)是一种与线粒体DNA(mtDNA)突变相关的使人衰弱的线粒体疾病。不幸的是,由于线粒体替代的挑战,LHON患者的可用治疗选择有限.在我们的研究中,我们将LHON尿液细胞重编程为诱导多能干细胞(iPSCs),并将其分化为神经祖细胞(NPCs)和神经元,用于疾病建模.我们的研究表明,LHON神经元表现出显著较高水平的mtDNA突变和线粒体功能降低。确认疾病表型。然而,通过将LHONiPSC衍生的NPCs与间充质干细胞(MSCs)共培养,我们观察到突变mtDNA的显着拯救和LHON神经元线粒体代谢功能的显着改善。这些发现表明,与MSCs共培养可以增强LHONNPCs的线粒体功能,甚至在它们分化为神经元之后。这一发现有望成为LHON患者的潜在治疗策略。
    Leber\'s hereditary optic neuropathy (LHON) is a debilitating mitochondrial disease associated with mutations in mitochondrial DNA (mtDNA). Unfortunately, the available treatment options for LHON patients are limited due to challenges in mitochondrial replacement. In our study, we reprogramming LHON urine cells into induced pluripotent stem cells (iPSCs) and differentiating them into neural progenitor cells (NPCs) and neurons for disease modeling. Our research revealed that LHON neurons exhibited significantly higher levels of mtDNA mutations and reduced mitochondrial function, confirming the disease phenotype. However, through co-culturing LHON iPSC-derived NPCs with mesenchymal stem cells (MSCs), we observed a remarkable rescue of mutant mtDNA and a significant improvement in mitochondrial metabolic function in LHON neurons. These findings suggest that co-culturing with MSCs can enhance mitochondrial function in LHON NPCs, even after their differentiation into neurons. This discovery holds promise as a potential therapeutic strategy for LHON patients.
    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
    人胚胎干细胞和人诱导多能干细胞可用于创建称为脑类器官的3D组织。它们在结构和功能两方面更忠实地复制了人脑组织的生理和病理特征,它们更精确地类似于人类胚胎大脑的形态和细胞结构。这使它们成为有价值的模型,用于药物筛选和关于人脑和相关疾病发展的体外研究。脑类器官实现的技术突破对不同脑区的研究产生了重大影响,大脑发育和疾病,大脑与其他组织和器官之间的联系,和大脑进化。本文讨论了脑类器官的发育,它们在糖尿病研究中的应用,和他们的进步。
    Human embryonic stem cells and human induced pluripotent stem cells may be used to create 3D tissues called brain organoids. They duplicate the physiological and pathological characteristics of human brain tissue more faithfully in terms of both structure and function, and they more precisely resemble the morphology and cellular structure of the human embryonic brain. This makes them valuable models for both drug screening and in vitro studies on the development of the human brain and associated disorders. The technical breakthroughs enabled by brain organoids have a significant impact on the research of different brain regions, brain development and sickness, the connections between the brain and other tissues and organs, and brain evolution. This article discusses the development of brain organoids, their use in diabetes research, and their progress.
    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
    多发性硬化(MS)是一种异质性的炎症和神经退行性疾病,具有不可预测的进行性残疾过程。由于对潜在机制的见解有限,因此治疗进行性MS具有挑战性。我们使用交叉组织(血液和死后大脑)和多层数据(遗传,表观遗传,转录组)来自独立队列。在PPMS中,我们发现了1q21.1基因座的超甲基化,受PPMS特异性遗传变异控制,并影响近端基因的表达(CHD1L,PRKAB2)在大脑中。来自报告基因测定和CRISPR/dCas9实验的证据支持甲基化和表达之间的因果联系,并且相关网络分析进一步暗示这些基因在PPMS脑过程中。CHD1L在人类iPSC衍生的神经元中的敲除和chd1l在斑马鱼中的敲除导致神经元的发育和功能缺陷。因此,若干证据表明,1q21.1基因座中独特的遗传-表观遗传-转录相互作用可能导致PPMS发病.
    Multiple Sclerosis (MS) is a heterogeneous inflammatory and neurodegenerative disease with an unpredictable course towards progressive disability. Treating progressive MS is challenging due to limited insights into the underlying mechanisms. We examined the molecular changes associated with primary progressive MS (PPMS) using a cross-tissue (blood and post-mortem brain) and multilayered data (genetic, epigenetic, transcriptomic) from independent cohorts. In PPMS, we found hypermethylation of the 1q21.1 locus, controlled by PPMS-specific genetic variations and influencing the expression of proximal genes (CHD1L, PRKAB2) in the brain. Evidence from reporter assay and CRISPR/dCas9 experiments supports a causal link between methylation and expression and correlation network analysis further implicates these genes in PPMS brain processes. Knock-down of CHD1L in human iPSC-derived neurons and knock-out of chd1l in zebrafish led to developmental and functional deficits of neurons. Thus, several lines of evidence suggest a distinct genetic-epigenetic-transcriptional interplay in the 1q21.1 locus potentially contributing to PPMS pathogenesis.
    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
    心血管疾病的发病率一直在不断增加。因为心肌细胞(CM)是不可再生的细胞,很难找到合适的CM来源来修复受伤的心脏。人类诱导多能干细胞(hiPSC)分化和成熟为CM的研究对于心脏病的治疗具有重要意义。使用hiPSCs作为再生疗法可以治疗许多无法治愈的疾病,包括进行性心力衰竭.本综述有助于在细胞学水平上研究心脏修复和心血管疾病的靶向治疗。最近的研究表明,对于hiPSCs向CMs的分化和成熟,脂肪酸对细胞代谢有很大的影响,细胞器发育,特定基因的表达,和功能性能。本文就脂肪酸如何影响hiPSCs分化为CMs及其成熟的研究进展作一综述。
    The incidence of cardiovascular disease has been continuously increasing. Because cardiomyocytes (CM) are non-renewable cells, it is difficult to find appropriate CM sources to repair injured hearts. Research of human induced pluripotent stem cell (hiPSC) differentiation and maturation into CM has been invaluable for the treatment of heart diseases. The use of hiPSCs as regenerative therapy allows for the treatment of many diseases that cannot be cured, including progressive heart failure. This review contributes to the study of cardiac repair and targeted treatment of cardiovascular diseases at the cytological level. Recent studies have shown that for differentiation and maturation of hiPSCs into CMs, fatty acids have a strong influence on cellular metabolism, organelle development, expression of specific genes, and functional performance. This review describes the recent research progress on how fatty acids affect the differentiation of hiPSCs into CMs and their maturation.
    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:B细胞CLL/淋巴瘤6成员B(BCL6B)在细胞核内作为序列特异性转录抑制因子,在各种生物功能中发挥关键作用,包括肿瘤抑制,免疫反应,干细胞自我更新,血管生成。然而,BCL6B是否参与内皮细胞(EC)的发育在很大程度上还不清楚.已知ETS变体转录因子2(ETV2)促进EC分化。本研究旨在确定BCL6B在EC分化中的重要作用及其潜在机制。
    方法:建立具有BCL6B过表达或BCL6B敲低的多西环素诱导的人诱导多能干细胞(hiPSC)系,并分化成EC和血管类器官(VOs)。进行RNA测序分析以鉴定在EC从hiPSC分化期间由BCL6B调节的潜在信号通路。实时定量PCR(qRT-PCR)用于检测多能性和血管特异性标记基因的表达。通过流式细胞术分析确定EC分化效率。通过体外管形成测定评价EC的性能。使用免疫荧光分析或蛋白质印迹评估蛋白质表达和血管样结构。荧光素酶报告基因测定和染色质免疫沉淀(ChIP)-PCR分析用于确定BCL6B和ETV2之间的调节关系。
    结果:从hiPSC成功地产生了功能性ECs和V0s。值得注意的是,BCL6B的过表达被抑制,而BCL6B的敲低改善了从hiPSC的EC分化。此外,BCL6B的过表达减弱了衍生的hiPSC-EC形成管状结构的能力。此外,与对照VOs相比,BCL6B过表达抑制了VOs的生长,而BCL6B敲除对VOs大小影响不大。RNA测序分析证实,我们的分化方案诱导细胞/组织/系统发育过程的景观变化,特别是血管发育和管形态发生,BCL6B显著调节。随后的实验证实,BCL6B与ETV2启动子区的结合促进了BCL6B的抑制作用,从而抑制了ETV2的转录活性。重要的是,BCL6B过表达对hiPSCs向EC分化的抑制作用可以通过ETV2过表达来挽救。
    结论:BCL6B通过抑制ETV2的转录活性抑制EC分化并阻碍VO发育。
    BACKGROUND: B-cell CLL/lymphoma 6 member B (BCL6B) operates as a sequence-specific transcriptional repressor within the nucleus, playing crucial roles in various biological functions, including tumor suppression, immune response, stem cell self-renew, and vascular angiogenesis. However, whether BCL6B is involved in endothelial cell (EC) development has remained largely unknown. ETS variant transcription factor 2 (ETV2) is well known to facilitate EC differentiation. This study aims to determine the important role of BCL6B in EC differentiation and its potential mechanisms.
    METHODS: Doxycycline-inducible human induced pluripotent stem cell (hiPSC) lines with BCL6B overexpression or BCL6B knockdown were established and subjected to differentiate into ECs and vessel organoids (VOs). RNA sequencing analysis was performed to identify potential signal pathways regulated by BCL6B during EC differentiation from hiPSCs. Quantitative real-time PCR (qRT-PCR) was used to detect the expression of pluripotency and vascular-specific marker genes expression. EC differentiation efficiency was determined by Flow cytometry analysis. The performance of EC was evaluated by in vitro Tube formation assay. The protein expression and the vessel-like structures were assessed using immunofluorescence analysis or western blot. Luciferase reporter gene assay and chromatin immunoprecipitation (ChIP)-PCR analysis were used to determine the regulatory relationship between BCL6B and ETV2.
    RESULTS: Functional ECs and VOs were successfully generated from hiPSCs. Notably, overexpression of BCL6B suppressed while knockdown of BCL6B improved EC differentiation from hiPSCs. Additionally, the overexpression of BCL6B attenuated the capacity of derived hiPSC-ECs to form a tubular structure. Furthermore, compared to the control VOs, BCL6B overexpression repressed the growth of VOs, whereas BCL6B knockdown had little effect on the size of VOs. RNA sequencing analysis confirmed that our differentiation protocol induced landscape changes for cell/tissue/system developmental process, particularly vascular development and tube morphogenesis, which were significantly modulated by BCL6B. Subsequent experiments confirmed the inhibitory effect of BCL6B is facilitated by the binding of BCL6B to the promoter region of ETV2, led to the suppression of ETV2\'s transcriptional activity. Importantly, the inhibitory effect of BCL6B overexpression on EC differentiation from hiPSCs could be rescued by ETV2 overexpression.
    CONCLUSIONS: BCL6B inhibits EC differentiation and hinders VO development by repressing the transcriptional activity of ETV2.
    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:T细胞活化异常在心肌炎的发病机制中起重要作用,持续的T细胞反应会导致自身免疫和慢性心脏炎症,甚至扩张型心肌病。尽管以前的工作已经在动物模型中研究了T细胞在心肌炎中的作用,人心肌细胞的具体机制尚未被研究。
    方法:在本研究中,我们构建了人诱导多能干细胞来源的心肌细胞(hiPSC-CMs),并通过与外周单核血中分离的活化CD4+T或CD8+T细胞共培养,建立了T细胞介导的心脏损伤模型,以阐明炎症引起的心肌细胞损伤的发病机制.
    结果:通过定量蛋白质组学与组织和细胞免疫荧光检查相结合,我们从hiPSC-CMs建立了一个具有明显心肌细胞损伤和乳酸脱氢酶含量升高的炎性心肌的蛋白质组图谱,肌酸激酶同工酶MB和心肌肌钙蛋白。观察到hiPSC-CM的一系列分子功能障碍,表明CD4细胞可以通过激活NOD样受体信号通路产生直接的心肌细胞损伤。
    结论:我们研究中提供的数据基于hiPSC-CM损伤模型建立了炎性心肌的蛋白质组图。这些结果可以为发现心肌炎的临床治疗方法提供指导。
    BACKGROUND: Abnormalities in T cell activation play an important role in the pathogenesis of myocarditis, and persistent T cell responses can lead to autoimmunity and chronic cardiac inflammation, as well as even dilated cardiomyopathy. Although previous work has examined the role of T cells in myocarditis in animal models, the specific mechanism for human cardiomyocytes has not been investigated.
    METHODS: In this study, we constructed the human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and established the T cell-mediated cardiac injury model by co-culturing with activated CD4 + T or CD8 + T cells that were isolated from peripheral mononuclear blood to elucidate the pathogenesis of myocardial cell injury caused by inflammation.
    RESULTS: By combination of quantitative proteomics with tissue and cell immunofluorescence examination, we established a proteome profile of inflammatory myocardia from hiPSC-CMs with obvious cardiomyocyte injury and increased levels of lactate dehydrogenase content, creatine kinase isoenzyme MB and cardiac troponin. A series of molecular dysfunctions of hiPSC-CMs was observed and indicated that CD4 + cells could produce direct cardiomyocyte injury by activating the NOD-like receptor signals pathway.
    CONCLUSIONS: The data presented in our study established a proteome map of inflammatory myocardial based on hiPSC-CMs injury model. These results can provide guidance in the discovery of improved clinical treatments for myocarditis.
    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
    脊髓损伤(SCI)后,神经再生和回路重建仍然是一个挑战。皮质脊髓锥体神经元具有很强的轴突投射能力。在这项研究中,通过向培养物中添加小分子dorsomorphin,将人诱导多能干细胞(iPSCs)分化为锥体神经元前体(PNP)。在损伤的同一天,将iPSC衍生的PNP急性移植到大鼠挫伤SCI模型中。雕刻后,行为测试显示,与载体对照组相比,SCI大鼠的运动功能显着改善。植入八周后,PNP成熟为皮质脊髓锥体神经元,并延伸轴突进入远处宿主脊髓组织,主要是在尾部方向。病变部位的宿主神经元也将轴突生长到移植物中。作为桥接中继的可能的突触连接可能已经在宿主和移植物衍生的神经元之间形成,如突触前和突触后标记染色和化学遗传调节系统的调节所示。PNP移植物在损伤部位显示出抗炎作用,并且可能使小胶质细胞/巨噬细胞偏向M2表型。此外,PNP移植物是安全的,移植到免疫缺陷小鼠和SCI大鼠中后未检测到肿瘤形成。重建整个病变部位的神经元中继电路并调节SCI微环境的潜力使PNP成为治疗SCI的有希望的细胞候选者。
    Nerve regeneration and circuit reconstruction remain a challenge following spinal cord injury (SCI). Corticospinal pyramidal neurons possess strong axon projection ability. In this study, human induced pluripotent stem cells (iPSCs) were differentiated into pyramidal neuronal precursors (PNPs) by addition of small molecule dorsomorphin into the culture. iPSC-derived PNPs were transplanted acutely into a rat contusion SCI model on the same day of injury. Following engraftment, the SCI rats showed significantly improved motor functions compared with vehicle control group as revealed by behavioral tests. Eight weeks following engraftment, the PNPs matured into corticospinal pyramidal neurons and extended axons into distant host spinal cord tissues, mostly in a caudal direction. Host neurons rostral to the lesion site also grew axons into the graft. Possible synaptic connections as a bridging relay may have been formed between host and graft-derived neurons, as indicated by pre- and post-synaptic marker staining and the regulation of chemogenetic regulatory systems. PNP graft showed an anti-inflammatory effect at the injury site and could bias microglia/macrophages towards a M2 phenotype. In addition, PNP graft was safe and no tumor formation was detected after transplantation into immunodeficient mice and SCI rats. The potential to reconstruct a neuronal relay circuitry across the lesion site and to modulate the microenvironment in SCI makes PNPs a promising cellular candidate for treatment of SCI.
    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:心力衰竭(HF)的特征是氧化应激和线粒体功能障碍。这项研究调查了通过源自诱导多能干细胞(iPSC)的外泌体递送的Necrostatin-1(Nec-1)的治疗潜力,以解决HF中的这些病症。
    方法:建立HF大鼠模型,并使用超声心动图进行全面评估,血流动力学,和心室质量指数测量。iPSCs用于分离外泌体,装载有Nec-1,并表征为有效递送到心肌细胞中。Nec-1负载的外泌体(Nec-1-Exos)之间的相互作用,聚(ADP-核糖)聚合酶1(PARP1),并探讨了凋亡诱导因子线粒体相关因子1(AIFM1)。功能增益实验评估了心肌细胞参数的变化,并对心肌组织进行组织学分析。
    结果:心肌细胞成功内化了负载Nec-1的外泌体,导致PARP1下调,抑制AIFM1核易位,增加ATP和超氧化物歧化酶水平,降低活性氧和丙二醛水平,并恢复了线粒体膜电位.组织学检查证实了Nec-1对PARP1/AIFM1轴的调节,减轻了HF。
    结论:携带Nec-1的iPSC衍生的外泌体通过靶向PARP1/AIFM1轴减弱HF的氧化应激和线粒体功能障碍。这项研究提出了一种有前途的HF治疗策略,并强调了外泌体介导的药物递送的潜力。
    BACKGROUND: Heart failure (HF) is characterized by oxidative stress and mitochondrial dysfunction. This study investigates the therapeutic potential of Necrostatin-1 (Nec-1) delivered through exosomes derived from induced pluripotent stem cells (iPSCs) to address these pathologies in HF.
    METHODS: An HF rat model was established, and comprehensive assessments were performed using echocardiography, hemodynamics, and ventricular mass index measurements. iPSCs were used to isolate exosomes, loaded with Nec-1, and characterized for efficient delivery into cardiomyocytes. The interaction between Nec-1-loaded exosomes (Nec-1-Exos), poly (ADP-ribose) polymerase 1 (PARP1), and apoptosis-inducing factor mitochondria-associated 1 (AIFM1) was explored. Gain-of-function experiments assessed changes in cardiomyocyte parameters, and histological analyses were conducted on myocardial tissues.
    RESULTS: Cardiomyocytes successfully internalized Nec-1-loaded exosomes, leading to downregulation of PARP1, inhibition of AIFM1 nuclear translocation, increased ATP and superoxide dismutase levels, reduced reactive oxygen species and malonaldehyde levels, and restored mitochondrial membrane potential. Histological examinations confirmed the modulation of the PARP1/AIFM1 axis by Nec-1, mitigating HF.
    CONCLUSIONS: iPSC-derived exosomes carrying Nec-1 attenuate oxidative stress and mitochondrial dysfunction in HF by targeting the PARP1/AIFM1 axis. This study proposes a promising therapeutic strategy for HF management and highlights the potential of exosome-mediated drug delivery.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号