Mechanosensitive channel

机械敏感通道
  • 文章类型: Journal Article
    生物医学研究的最新进展强调了非侵入性细胞操作技术的重要性。声遗传学,一种利用基因工程在靶细胞中产生超声敏感蛋白的方法,随着光遗传学越来越突出,电遗传学,和磁遗传学。用超声波刺激后,这些蛋白质引发细胞活动和功能的级联反应。与传统的超声模式不同,声遗传学提供了增强的空间选择性,提高疾病治疗的精度和安全性。这项技术在广泛的临床研究和治疗应用中拓宽了非手术干预的范围,包括神经调节,肿瘤治疗,干细胞疗法,和超越。尽管目前的文献主要强调超声神经调节,这篇综述提供了对超声遗传学的全面探索。我们讨论超声特性,超声遗传学中使用的特定超声敏感蛋白,以及该技术在管理神经系统疾病等疾病方面的潜力,癌症,和眼科疾病,和干细胞疗法。我们的目标是激发在这个有前途的领域进一步研究的新观点。
    Recent advancements in biomedical research have underscored the importance of noninvasive cellular manipulation techniques. Sonogenetics, a method that uses genetic engineering to produce ultrasound-sensitive proteins in target cells, is gaining prominence along with optogenetics, electrogenetics, and magnetogenetics. Upon stimulation with ultrasound, these proteins trigger a cascade of cellular activities and functions. Unlike traditional ultrasound modalities, sonogenetics offers enhanced spatial selectivity, improving precision and safety in disease treatment. This technology broadens the scope of non-surgical interventions across a wide range of clinical research and therapeutic applications, including neuromodulation, oncologic treatments, stem cell therapy, and beyond. Although current literature predominantly emphasizes ultrasonic neuromodulation, this review offers a comprehensive exploration of sonogenetics. We discuss ultrasound properties, the specific ultrasound-sensitive proteins employed in sonogenetics, and the technique\'s potential in managing conditions such as neurological disorders, cancer, and ophthalmic diseases, and in stem cell therapies. Our objective is to stimulate fresh perspectives for further research in this promising field.
    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
    来自身体不同器官的细胞经历一系列机械和渗透压,这些压力在各种疾病中发生变化,包括神经学,心血管,眼科,和肾脏疾病。这里,我们展示了使用来自大电导(MscL)的机械敏感通道的微生物来源的工程传感器-执行器-调制器(SAM)来感测外部机械应力和调节哺乳动物细胞的活动。SAM在哺乳动物细胞膜中可靠表达,并充当张力激活的压力释放阀。Further,哺乳动物细胞中异源表达SAM的活性可以通过渗透压调节。来自不同微生物来源的SAM变体的机械敏感性活性的比较显示,响应于低渗透休克施加的机械应力,内向电流和染料吸收存在差异。在哺乳动物细胞中使用SAM通道作为机械应力激活的调节剂可以提供用于治疗与机械或渗透压相关的病症的新的治疗方法。
    Cells from different organs in the body experience a range of mechanical and osmotic pressures that change in various diseases, including neurological, cardiovascular, ophthalmological, and renal diseases. Here, we demonstrate the use of an engineered Sensor-Actuator-Modulator (SAM) of microbial origin derived from a mechanosensitive channel of large conductance (MscL) for sensing external mechanical stress and modulating activities of mammalian cells. SAM is reliably expressed in the mammalian cell membrane and acts as a tension-activated pressure release valve. Further, the activities of heterologously expressed SAM in mammalian cells could be modulated by osmotic pressure. A comparison of the mechanosensitive activities of SAM-variants from different microbial origins shows differential inward current and dye uptake in response to mechanical stress exerted by hypo-osmotic shock. The use of SAM channels as mechanical stress-activated modulators in mammalian cells could provide new therapeutic approaches for treating disorders related to mechanical or osmotic pressure.
    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
    大电导(MscL)的机械敏感通道充当“紧急释放阀”,保护细菌细胞免受急性低渗应激,它可以作为研究机械力传导机制的范例。提出了MSCL门控以不打开的扩展启动,随后通过许多中间子状打开孔隙,并以完整的开口结束。然而,门控过程的细节在很大程度上仍然未知。使用体内活力测定,单通道膜片钳记录,半胱氨酸交联,和色氨酸荧光猝灭方法,我们鉴定并表征了孔结构域中收缩区占比不同的MscL突变体。结果表明,在门控时,收缩点沿着门控途径向细胞质侧从残基G26(尽管G22)向L19转移,表明紧密堆积的疏水性收缩区域的膨胀的封闭-膨胀过渡耦合,以响应膜张力进行初始离子渗透。此外,这些转变是由疏水和脂类相互作用与收缩的“热点”调节。我们的数据揭示了从MscL的封闭到开放子状态的过渡的新分辨率,提供对MSCL门控机制的见解。
    The mechanosensitive channel of large conductance (MscL) acts as an \"emergency release valve\" that protects bacterial cells from acute hypoosmotic stress, and it serves as a paradigm for studying the mechanism underlying the transduction of mechanical forces. MscL gating is proposed to initiate with an expansion without opening, followed by subsequent pore opening via a number of intermediate substates, and ends in a full opening. However, the details of gating process are still largely unknown. Using in vivo viability assay, single channel patch clamp recording, cysteine cross-linking, and tryptophan fluorescence quenching approach, we identified and characterized MscL mutants with different occupancies of constriction region in the pore domain. The results demonstrated the shifts of constriction point along the gating pathway towards cytoplasic side from residue G26, though G22, to L19 upon gating, indicating the closed-expanded transitions coupling of the expansion of tightly packed hydrophobic constriction region to conduct the initial ion permeation in response to the membrane tension. Furthermore, these transitions were regulated by the hydrophobic and lipidic interaction with the constricting \"hot spots\". Our data reveal a new resolution of the transitions from the closed to the opening substate of MscL, providing insights into the gating mechanisms of MscL.
    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
    目的:本研究调查了上尿路功能障碍(UUTD)后上尿路尿动力学(UUTU)的变化。方法:通过单侧输尿管梗阻建立UUTD模型。为了测量肾盂体积,休息压力。进行了输尿管肌电图(EMG)和原位输尿管收缩实验。获得输尿管组织进行HE和masson染色,IF染色和IHC研究探索Piezo1的分布,并使用Westernblot研究Piezo1的表达。结果:实验组术后肾盂体积和静息压逐渐升高。输尿管组织水肿的程度,细胞坏死和纤维化逐渐增多。术后实验组输尿管最年夜收缩力和频率明显高于假手术组。Westernblotting显示Piezo1的表达强度逐渐增加,并明显高于假手术组。对输尿管各亚层的进一步分析表明,Piezo1在尿道上皮层中高度表达,其次是上皮层,在平滑肌细胞层有低表达。结论:本研究观察到上尿路的形态学和电生理改变可能是UUTU异常的重要机制。Piezo1的表达增加可能是UUTD后尿动力学异常的新分子机制。
    Objects: This study investigated changes in upper urinary tract urodynamics (UUTU) after upper urinary tract dysfunction (UUTD). Methods: The UUTD model was induced through unilateral ureteral obstruction. To measure the renal pelvis volume, and resting pressure. Ureteral electromyography (EMG) and in situ ureteral constriction experiments were performed. Ureteral tissue was obtained for HE and masson staining, IF staining and IHC research to explore the distribution of Piezo1, and the expression of Piezo1 was studied using Western blotting. Results: The study showed that the renal pelvis volumes and the renal pelvis resting pressures gradually increased post surgery in the experimental group. The degree of ureteral tissue edema, cell necrosis and fibrosis gradually increased. The maximum contraction force and frequency of ureter in the experimental group post surgery were significantly higher than in the sham group. Western blotting showed that the expression intensity of Piezo1 gradually increased and was significantly higher than in the sham group. Further analysis of each sub-layer of the ureter revealed that Piezo1 was highly expressed in the urothelium layer, followed by the suburothelium layer, and had low expression in the smooth muscle cell layer. Conclusion: The study observed that morphological and electrophysiological changes in the upper urinary tract may be important mechanisms of abnormal UUTU. Increased expression of the Piezo1 may be a new molecular mechanism of abnormal urodynamics after UUTD.
    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
    超声已被证明可以激活机械敏感通道,这被认为是超声神经调节的主要机制。目前,所有对超声敏感的通道都是阳离子通道。除了阳离子通道,阴离子通道在神经功能中也起着不可或缺的作用。然而,到目前为止,还没有关于超声波调节阴离子通道的研究。如果阴离子通道也能被超声波激活,它们将不可避免地导致超声神经调节的多功能性。囊性纤维化跨膜转导调节因子(CFTR)已被证明是一种机械敏感的通道,介导阴离子跨膜流动。为了确定CFTR对超声敏感,CFTR在HEK293T细胞中外源表达,并受到低强度超声刺激。使用全细胞膜片钳在超声(0.8MHz,将0.20MPa)递送至这些电池。当CFTR抑制剂(GlyH101)应用于溶液或氯离子从溶液中清除时,这些电流消失。同时,当应用CFTR激动剂(Forskolin)时,这些电流的幅度增加。这些结果表明超声刺激可以激活CFTR以在单细胞水平介导氯离子的跨膜流动。这些发现可能会扩展超声在神经调节领域的应用。
    Ultrasound has been demonstrated to activate mechanosensitive channels, which is considered the main mechanism of ultrasound neuromodulation. Currently, all channels that have been shown to be sensitive to ultrasound are cation channels. In addition to cation channels, anion channels also play indispensable roles in neural function. However, there have been no research on ultrasound regulation of anion channels until now. If anion channels can be activated by ultrasound as well, they will inevitably lead to more versatility in ultrasound neuromodulation. Cystic fibrosis transmembrane transduction regulator (CFTR) has been demonstrated to be a mechanically sensitive channel, mediating anionic transmembrane flow. To identify that CFTR is sensitive to ultrasound, CFTR was exogenously expressed in HEK293T cells and was stimulated by low intensity ultrasound. Outward currents in CFTR-expressed HEK293T cells were observed by using whole-cell patch clamp when ultrasound (0.8 MHz, 0.20 MPa) was delivered to these cells. These currents were abolished when the CFTR inhibitor (GlyH101) was applied to the solution or chloride ions was cleared from the solution. Meanwhile, the amplitude of these currents increased when the CFTR agonist (Forskolin) was applied. These results suggest that ultrasound stimuli can activate the CFTR to mediate transmembrane flowing of chloride ions at the single cell level. These findings may expand the application of ultrasound in the neuromodulation field.
    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
    机械敏感性PIEZO通道构成多种临床病症的潜在药理靶标,推动寻找有效的化学PIEZO调节剂。其中包括Yoda1,一种通过基于细胞的高通量筛选发现的广泛使用的合成小分子PIEZO1激活剂。Yoda1被认为与PIEZO1的机械感觉臂域结合,夹在通道孔附近的两个跨膜区之间。然而,Yoda1与该区域的结合如何促进通道激活仍然难以捉摸。这里,我们首先证明了交联PIEZO1重复A和B与二硫键以氧化还原依赖性方式降低Yoda1的作用,这表明Yoda1通过扰乱这些重复之间的接触来发挥作用。使用基于分子动力学的绝对结合自由能模拟,我们接下来显示Yoda1优先占据更深的位置,在PIEZO1开放状态下具有较高亲和力的两亲性结合位点。在打开和关闭状态下使用Yoda1\的绑定姿势,在膜环境中进行了相对结合自由能模拟,概述已知Yoda1类似物的结构-活性关系。通过使用Yoda1结合位点的计算片段图虚拟筛选800万种化合物文库,我们随后鉴定了两种对PIEZO1具有激动剂活性的化学支架。这项研究支持一种药理学模型,其中Yoda1通过楔入重复序列A和B来激活PIEZO1,为PIEZO1调制器的合理设计提供了结构和热力学框架。除了PIEZO频道,这里采用的三种正交计算方法代表了在高度异质的膜蛋白系统中发现药物的有希望的途径。
    Mechanosensitive PIEZO channels constitute potential pharmacological targets for multiple clinical conditions, spurring the search for potent chemical PIEZO modulators. Among them is Yoda1, a widely used synthetic small molecule PIEZO1 activator discovered through cell-based high-throughput screening. Yoda1 is thought to bind to PIEZO1\'s mechanosensory arm domain, sandwiched between two transmembrane regions near the channel pore. However, how the binding of Yoda1 to this region promotes channel activation remains elusive. Here, we first demonstrate that cross-linking PIEZO1 repeats A and B with disulfide bridges reduces the effects of Yoda1 in a redox-dependent manner, suggesting that Yoda1 acts by perturbing the contact between these repeats. Using molecular dynamics-based absolute binding free energy simulations, we next show that Yoda1 preferentially occupies a deeper, amphipathic binding site with higher affinity in PIEZO1 open state. Using Yoda1\'s binding poses in open and closed states, relative binding free energy simulations were conducted in the membrane environment, recapitulating structure-activity relationships of known Yoda1 analogs. Through virtual screening of an 8 million-compound library using computed fragment maps of the Yoda1 binding site, we subsequently identified two chemical scaffolds with agonist activity toward PIEZO1. This study supports a pharmacological model in which Yoda1 activates PIEZO1 by wedging repeats A and B, providing a structural and thermodynamic framework for the rational design of PIEZO1 modulators. Beyond PIEZO channels, the three orthogonal computational approaches employed here represent a promising path toward drug discovery in highly heterogeneous membrane protein systems.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    蛋白质的跨膜63(TMEM63)家族最初被鉴定为植物中渗透敏感性钙可渗透(OSCA)通道的同源物。除了通过高/低渗透压提出的激活机制外,最近还证明了OSCA和TMEM63蛋白的机械敏感性。TMEM63蛋白存在于所有动物中,果蝇中只有一个成员(TMEM63),哺乳动物中有三个成员(TMEM63A/B/C)。在人类中,据报道,TMEM63A的单等位基因变异体在婴儿期会导致短暂的髓鞘减少,或严重的骨髓溶解不足和整体发育迟缓。TMEM63B的杂合变体在患有智力障碍和运动功能和脑形态异常的患者中发现。TMEM63C的双等位基因变异体与伴有轻度或无智力障碍的遗传性痉挛性截瘫相关。到目前为止,已明确认可的TMEM63蛋白的生理功能包括检测果蝇中的食物沙质和环境湿度,通过调节耳蜗毛细胞的存活来支持小鼠的听力。在这次审查中,我们总结了有关TMEM63通道的激活机制和生物学功能的最新知识,并为有兴趣研究该蛋白质家族在体内普遍存在的更多生理和致病作用的研究人员提供了简明的参考。
    The transmembrane 63 (TMEM63) family of proteins are originally identified as homologs of the osmosensitive calcium-permeable (OSCA) channels in plants. Mechanosensitivity of OSCA and TMEM63 proteins are recently demonstrated in addition to their proposed activation mechanism by hyper/hypo-osmolarity. TMEM63 proteins exist in all animals, with a single member in Drosophila (TMEM63) and three members in mammals (TMEM63 A/B/C). In humans, monoallelic variants of TMEM63A have been reported to cause transient hypomyelination during infancy, or severe hypomyelination and global developmental delay. Heterozygous variants of TMEM63B are found in patients with intellectual disability and abnormal motor function and brain morphology. Biallelic variants of TMEM63C are associated with hereditary spastic paraplegias accompanied by mild or no intellectual disability. Physiological functions of TMEM63 proteins clearly recognized so far include detecting food grittiness and environmental humidity in Drosophila, and supporting hearing in mice by regulating survival of cochlear hair cells. In this review, we summarize current knowledge about the activation mechanisms and biological functions of TMEM63 channels, and provide a concise reference for researchers interested in investigating more physiological and pathogenic roles of this family of proteins with ubiquitous expression in the body.
    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
    细胞感知和响应其物理环境的能力在广泛的生物过程中起着基本作用。作为细胞膜中最重要的分子力传感器和换能器之一,机械敏感(MS)离子通道可以将机械输入转换为生化或电信号,以介导各种感觉。显示细胞样组织的细胞大小的隔室的自下而上的结构,行为,和复杂性,也被称为合成细胞,作为单独表征生物功能的实验平台,已经越来越受欢迎。通过重建合成脂质双层中的MS通道,我们设想将机械敏感性合成细胞用于多种医学应用。这里,我们描述了使用超声波的三个不同概念,剪切应力,和压缩应力作为机械刺激,以激活药物从机械敏感性合成细胞中释放,用于疾病治疗。
    The ability of cells to sense and respond to their physical environment plays a fundamental role in a broad spectrum of biological processes. As one of the most essential molecular force sensors and transducers found in cell membranes, mechanosensitive (MS) ion channels can convert mechanical inputs into biochemical or electrical signals to mediate a variety of sensations. The bottom-up construction of cell-sized compartments displaying cell-like organization, behaviors, and complexity, also known as synthetic cells, has gained popularity as an experimental platform to characterize biological functions in isolation. By reconstituting MS channels in the synthetic lipid bilayers, we envision using mechanosensitive synthetic cells for several medical applications. Here, we describe three different concepts for using ultrasound, shear stress, and compressive stress as mechanical stimuli to activate drug release from mechanosensitive synthetic cells for disease treatments.
    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
    大电导MscL的细菌机械敏感通道仅通过膜双层中增加的张力而激活。尽管提出了许多MscL开放的模型,其精确的机械门控机构,特别是在张力传感器处接收到的力如何传递到闸门仍然是不完整的。以前的研究表明,随着两亲性N端位于膜的细胞质表面附近,外表面附近的Phe78残留物也用作张力传感器,而Gly22是疏水门的中心组成部分。“本研究的重点是通过将膜片钳和分子动力学(MD)模拟应用于MscL的外跨膜螺旋(TM2)中的传感器Phe78到MscL的内跨膜螺旋(TM1)中的门的力传递机制。野生型MscL通道及其单个突变体在传感器(F78N)上,门(G22N)及其组合(G22N/F78N)双突变体。F78NMscL导致严重的功能丧失,而G22NMscL导致功能增益通道在静息膜张力下表现出自发开口。我们最初推测,G22N突变体的自发开放可能在没有张力作用于Phe78残基的情况下发生。为了检验这个假设,我们检测了(G22N/F78N)双突变体,出乎意料地既没有表现出自发活动,也没有表现出相对较高的膜张力的活动。要了解潜在的机制,我们进行了MD模拟并分析了力传递途径。结果表明,TM2中张力传感器(F78N)的突变不仅导致该残基与脂质的相互作用减少,但在邻近的TM1螺旋中也有一组氨基酸(Ile32-Leu36-Ile40),这导致向TM1上的栅极构成氨基酸的力传递效率低下。这种变化也引起了TM1向膜平面的轻微倾斜,并减小了门处通道孔的大小。这似乎是抑制双突变通道自发开放的主要机制。更重要的是,新发现的TM2(Phe78)和相邻TM1(Ile32-Leu36-Ile40)螺旋之间的相互作用似乎是MscL拉伸依赖性激活的重要力传递机制,因为这四种氨基酸中的任何一种被Asn取代会导致严重的MscL功能丧失。
    The bacterial mechanosensitive channel of large conductance MscL is activated exclusively by increased tension in the membrane bilayer. Despite many proposed models for MscL opening, its precise mechano-gating mechanism, particularly how the received force at the tension sensor transmits to the gate remains incomplete. Previous studies have shown that along with amphipathic N-terminus located near the cytoplasmic surface of the membrane, Phe78 residue near the outer surface also acts as a \"tension sensor,\" while Gly22 is a central constituent of the \"hydrophobic gate.\" Present study focused on elucidating the force transmission mechanism from the sensor Phe78 in the outer transmembrane helix (TM2) to the gate in the inner transmembrane helix (TM1) of MscL by applying the patch clamp and molecular dynamics (MD) simulations to the wild type MscL channel and its single mutants at the sensor (F78N), the gate (G22N) and their combination (G22N/F78N) double mutant. F78N MscL resulted in a severe loss-of-function, while G22N MscL caused a gain-of-function channel exhibiting spontaneous openings at the resting membrane tension. We initially speculated that the spontaneous opening in G22N mutant might occur without tension acting on Phe78 residue. To test this hypothesis, we examined the (G22N/F78N) double mutant, which unexpectedly exhibited neither spontaneous activity nor activity by a relatively high membrane tension. To understand the underlying mechanism, we conducted MD simulations and analyzed the force transduction pathway. Results showed that the mutation at the tension sensor (F78N) in TM2 caused decreased interaction of this residue not only with lipids, but also with a group of amino acids (Ile32-Leu36-Ile40) in the neighboring TM1 helix, which resulted in an inefficient force transmission to the gate-constituting amino acids on TM1. This change also induced a slight tilting of TM1 towards the membrane plane and decreased the size of the channel pore at the gate, which seems to be the major mechanism for the inhibition of spontaneous opening of the double mutant channel. More importantly, the newly identified interaction between the TM2 (Phe78) and adjacent TM1 (Ile32-Leu36-Ile40) helices seems to be an essential force transmitting mechanism for the stretch-dependent activation of MscL given that substitution of any one of these four amino acids with Asn resulted in severe loss-of-function MscL as reported in our previous work.
    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
    Piezo1机械敏感性离子通道(MSC)在人体生理中起着重要作用。尽管对Piezo1在神经系统中的功能和表达进行了一些研究,其在神经炎性星形胶质细胞中的电生理特性仍然未知。我们使用电记录测试星形细胞的神经炎症状态是否调节Piezo1,钙成像,和培养的星形胶质细胞的伤口愈合试验。在这项研究中,我们确定了神经炎症状态是否调节星形胶质细胞中的星形胶质细胞Piezo1电流.首先,我们在脂多糖(LPS)诱导的神经炎症条件下对小鼠小脑星形胶质细胞(C8-S)进行了电生理记录。我们发现LPS处理显著增加了C8-S中的MSC电流。LPS处理的MSC电流的半最大压力左移,但斜率灵敏度未被LPS处理改变。LPS诱导的MSC电流增加被Piezo1激动剂进一步增强,Yoda1,但被Piezo1抑制剂归一化,GsMTx4。此外,在LPS处理的C8-S中沉默Piezo1不仅使MSC电流归一化,而且使钙内流和细胞迁移速度归一化。一起,我们的结果表明,LPS致敏C8-S星形胶质细胞中的Piezo1通道。这些发现将表明星形胶质细胞Piezo1是神经炎症发病机理的决定因素,并且可能反过来成为进一步研究治愈几种神经元疾病和神经元细胞损伤相关炎症的基础。
    Piezo1 mechanosensitive ion channel (MSC) plays a significant role in human physiology. Despite several research on the function and expression of Piezo1 in the nervous system, its electrophysiological properties in neuroinflammatory astrocytes remain unknown. We tested whether astrocytic neuroinflammatory state regulates Piezo1 using electrical recordings, calcium imaging, and wound healing assays on cultured astrocytes. In this study, we determined whether neuroinflammatory condition regulates astrocytic Piezo1 currents in astrocytes. First, we performed electrophysiological recordings on the mouse cerebellum astrocytes (C8-S) under lipopolysaccharide (LPS)-induced neuroinflammatory condition. We found that LPS treatment significantly increased MSC currents in C8-S. The half-maximal pressure of LPS treated MSC currents was left-shifted but the slope sensitivity was not altered by LPS treatment. LPS-induced increase of MSC currents were further augmented by Piezo1 agonist, Yoda1 but were normalized by Piezo1 inhibitor, GsMTx4. Furthermore, silencing Piezo1 in LPS treated C8-S normalized not only MSC currents but also calcium influx and cell migration velocity. Together, our results show that LPS sensitized Piezo1 channel in C8-S astrocytes. These findings will suggest that astrocytic Piezo1 is a determinant of neuroinflammation pathogenesis and may in turn become the foundation of further research into curing several neuronal illnesses and injury related inflammation of neuronal cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号