Molecular Targeted Therapy

分子靶向治疗
  • 文章类型: Journal Article
    卵巢纤维化,以卵巢成纤维细胞的过度增殖和细胞外基质(ECM)的积累为特征,是卵巢功能障碍的主要原因之一。尽管卵巢纤维化在维持哺乳动物卵巢的正常生理功能方面具有关键作用,对这种情况的研究被大大低估了,这导致临床上对纤维化引起的卵巢功能障碍缺乏治疗选择。本文综述了卵巢纤维化分子机制的研究进展,包括TGF-β,细胞外基质,炎症,和其他促纤维化因素导致卵巢异常纤维化。此外,我们总结了目前针对卵巢纤维化的卵巢功能障碍的治疗方法,包括抗纤维化药物,干细胞移植,和外泌体疗法。本文就卵巢纤维化的研究进展作一综述,提出针对卵巢纤维化治疗卵巢功能障碍的潜在治疗策略。
    Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-β, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.
    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
    T-BOX因子属于进化保守的转录因子家族。T-BOX因子不仅在生长发育中起关键作用,而且还参与免疫。癌症开始,和进步。此外,相同的T-BOX分子在各种发育过程和肿瘤微环境中表现出不同甚至相反的作用。了解环境依赖性T-BOX因子在恶性肿瘤中的多种作用对于揭示T-BOX靶向癌症治疗的潜力至关重要。我们总结了T-BOX因子在不同发育过程中的生理作用及其表达失调时观察到的病理作用。我们还讨论了它们在肿瘤免疫微环境(TIME)中的调节作用以及尚未解决的新出现的问题。本综述将有助于系统全面地了解T-BOX转录因子家族在肿瘤生理学中的重要作用。病理学,和豁免权。目的是提供有价值的信息来支持T-BOX靶向治疗的发展。
    T-BOX factors belong to an evolutionarily conserved family of transcription factors. T-BOX factors not only play key roles in growth and development but are also involved in immunity, cancer initiation, and progression. Moreover, the same T-BOX molecule exhibits different or even opposite effects in various developmental processes and tumor microenvironments. Understanding the multiple roles of context-dependent T-BOX factors in malignancies is vital for uncovering the potential of T-BOX-targeted cancer therapy. We summarize the physiological roles of T-BOX factors in different developmental processes and their pathological roles observed when their expression is dysregulated. We also discuss their regulatory roles in tumor immune microenvironment (TIME) and the newly arising questions that remain unresolved. This review will help in systematically and comprehensively understanding the vital role of the T-BOX transcription factor family in tumor physiology, pathology, and immunity. The intention is to provide valuable information to support the development of T-BOX-targeted therapy.
    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
    乳腺癌(BC)是世界范围内高度流行的恶性肿瘤,具有复杂的发病机制和治疗挑战。研究表明,甲基转移酶样3(METTL3)通过其靶RNA的甲基化而广泛参与多种肿瘤的发病机制。及其在BC中的作用和机制也被广泛研究。在这次审查中,我们旨在对现有研究进行全面解释,并阐明METTL3与BC之间的关系.这篇综述表明,高水平的METTL3与发病机制有关。预后不良,和BC的耐药性,提示METTL3是潜在的诊断或预后生物标志物和治疗靶点。总的来说,这篇综述提供了对METTL3如何通过RNA甲基化发挥作用的全面理解,为今后的基础研究和临床应用提供了有价值的参考。
    Breast cancer (BC) is a highly prevalent malignancy worldwide, with complex pathogenesis and treatment challenges. Research reveals that methyltransferase-like 3 (METTL3) is widely involved in the pathogenesis of several tumors through methylation of its target RNAs, and its role and mechanisms in BC are also extensively studied. In this review, we aim to provide a comprehensive interpretation of available studies and elucidate the relationship between METTL3 and BC. This review suggests that high levels of METTL3 are associated with the pathogenesis, poor prognosis, and drug resistance of BC, suggesting METTL3 as a potential diagnostic or prognostic biomarker and therapeutic target. Collectively, this review provides a comprehensive understanding of how METTL3 functions through RNA methylation, which provides a valuable reference for future fundamental studies and clinical applications.
    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
    多发性骨髓瘤(MM)是第二常见的浆细胞恶性血液病,以溶骨性骨病变为特征,贫血,高钙血症,肾功能衰竭,和恶性浆细胞的积累。MM的发病机制涉及MM细胞通过可溶性细胞因子和细胞粘附分子与骨髓微环境相互作用,激活各种信号通路,如PI3K/AKT/mTOR,RAS/MAPK,JAK/STAT,Wnt/β-catenin,和NF-κB通路。这些途径的异常激活有助于增殖,生存,迁移,和骨髓瘤细胞的耐药性,使它们成为有吸引力的治疗干预目标。目前,在MM中针对这些信号通路的批准药物是有限的,许多抑制剂和诱导剂仍处于临床前或临床研究阶段。MM的治疗选择包括非靶向药物,如烷化剂,皮质类固醇,免疫调节药物,蛋白酶体抑制剂,和组蛋白脱乙酰酶抑制剂。此外,靶向药物如单克隆抗体,嵌合抗原受体T细胞,双特异性T细胞衔接者,和双特异性抗体正在用于MM治疗。尽管在MM治疗方面取得了重大进展,这种疾病仍然无法治愈,强调需要根据新兴的理论知识开发新的或联合的靶向疗法,技术,和平台。在这次审查中,我们强调了信号通路在MM的恶性进展和治疗中的关键作用,探索靶向治疗和潜在治疗的进展,为改善MM管理和结局提供进一步的见解。
    Multiple myeloma (MM) is the second most common hematological malignancy of plasma cells, characterized by osteolytic bone lesions, anemia, hypercalcemia, renal failure, and the accumulation of malignant plasma cells. The pathogenesis of MM involves the interaction between MM cells and the bone marrow microenvironment through soluble cytokines and cell adhesion molecules, which activate various signaling pathways such as PI3K/AKT/mTOR, RAS/MAPK, JAK/STAT, Wnt/β-catenin, and NF-κB pathways. Aberrant activation of these pathways contributes to the proliferation, survival, migration, and drug resistance of myeloma cells, making them attractive targets for therapeutic intervention. Currently, approved drugs targeting these signaling pathways in MM are limited, with many inhibitors and inducers still in preclinical or clinical research stages. Therapeutic options for MM include non-targeted drugs like alkylating agents, corticosteroids, immunomodulatory drugs, proteasome inhibitors, and histone deacetylase inhibitors. Additionally, targeted drugs such as monoclonal antibodies, chimeric antigen receptor T cells, bispecific T-cell engagers, and bispecific antibodies are being used in MM treatment. Despite significant advancements in MM treatment, the disease remains incurable, emphasizing the need for the development of novel or combined targeted therapies based on emerging theoretical knowledge, technologies, and platforms. In this review, we highlight the key role of signaling pathways in the malignant progression and treatment of MM, exploring advances in targeted therapy and potential treatments to offer further insights for improving MM management and outcomes.
    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
    滋养层细胞表面抗原2(Trop2)在一系列实体瘤和多个致癌信号通路的参与者中过度表达,使其成为有吸引力的治疗靶点。在过去的十年里,各种Trop2靶向疗法的快速发展,特别是以抗体-药物偶联物(ADC)的出现为标志,彻底改变了治疗意见有限的Trop2阳性肿瘤患者的结果,例如三阴性乳腺癌(TNBC)。这篇综述全面总结了Trop2靶向治疗的进展,包括ADC,抗体,多特异性药物,免疫疗法,癌症疫苗,和小分子抑制剂,随着对他们设计的深入讨论,行动机制(MOA),和限制。此外,我们强调了这些新兴的Trop2靶向药物的临床研究进展,重点关注其临床应用和对肿瘤的治疗效果。此外,我们提出了未来研究的方向,例如加强我们对Trop2的结构和生物学的理解,探索最佳组合策略,并根据Trop2测试方法定制精确处理。
    Trophoblast cell surface antigen 2 (Trop2) is overexpressed in a range of solid tumors and participants in multiple oncogenic signaling pathways, making it an attractive therapeutic target. In the past decade, the rapid development of various Trop2-targeted therapies, notably marked by the advent of the antibody-drug conjugate (ADC), revolutionized the outcome for patients facing Trop2-positive tumors with limited treatment opinions, such as triple-negative breast cancer (TNBC). This review provides a comprehensive summary of advances in Trop2-targeted therapies, including ADCs, antibodies, multispecific agents, immunotherapy, cancer vaccines, and small molecular inhibitors, along with in-depth discussions on their designs, mechanisms of action (MOAs), and limitations. Additionally, we emphasize the clinical research progress of these emerging Trop2-targeted agents, focusing on their clinical application and therapeutic efficacy against tumors. Furthermore, we propose directions for future research, such as enhancing our understanding of Trop2\'s structure and biology, exploring the best combination strategies, and tailoring precision treatment based on Trop2 testing methodologies.
    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
    原理:代谢的协同重编程主导神经母细胞瘤(NB)的进展。基于阐明代谢重编程的分子机制,开发具有分层指导的NB治疗选择的个性化风险预测方法具有重要的临床意义。方法:采用基于机器学习的多步骤程序,在单细胞和代谢物通量维度阐明了代谢重编程驱动NB恶性进展的协同机制.随后,我们开发了一种有前景的代谢重编程相关预后特征(MPS)和基于MPS分层的个体化治疗方法,并使用临床前模型进行了进一步独立验证.结果:MPS鉴定的MPS-INB显示出明显高于MPS-II对应物的代谢重编程活性。与目前的临床特征相比,MPS显示出更高的准确性[AUC:0.915vs.0.657(MYCN),0.713(INSS阶段),和0.808(INRG分层)]预测预后。AZD7762和依托泊苷被确定为针对MPS-I和IINB的有效治疗剂,分别。随后的生物学测试表明AZD7762基本上抑制了生长,迁移,MPS-INB细胞的侵袭,比MPS-II细胞更有效。相反,依托泊苷对MPS-Ⅱ型NB细胞有较好的治疗作用。更令人鼓舞的是,AZD7762和依托泊苷显著抑制体内皮下肿瘤发生,扩散,MPS-I和MPS-II样本中的肺转移,分别;从而延长荷瘤小鼠的生存期。机械上,AZD7762和依托泊苷诱导的MPS-I和MPS-II细胞凋亡,分别,通过线粒体依赖性途径;MPS-INB通过谷氨酸代谢成瘾和乙酰辅酶A抵抗依托泊苷诱导的细胞凋亡。MPS-INB进展受多种代谢重编程驱动因素的推动,包括多药耐药,免疫抑制和促进肿瘤的炎症微环境。免疫学,MPS-INB通过MIF和THBS信号通路抑制免疫细胞。代谢,MPS-INB细胞的恶性增殖得到了重新编程的谷氨酸代谢的显著支持,三羧酸循环,尿素循环,等。此外,MPS-INB细胞表现出独特的肿瘤促进发育谱系和自我沟通模式,通过发育和自我通讯激活的致癌信号通路增强证明了这一点。结论:本研究为代谢重编程介导的NB恶性进展的分子机制提供了深刻的见解。它还揭示了在新的精确风险预测方法的指导下开发靶向药物,这可能有助于明显改善NB的治疗策略。
    Rationale: Synergic reprogramming of metabolic dominates neuroblastoma (NB) progression. It is of great clinical implications to develop an individualized risk prognostication approach with stratification-guided therapeutic options for NB based on elucidating molecular mechanisms of metabolic reprogramming. Methods: With a machine learning-based multi-step program, the synergic mechanisms of metabolic reprogramming-driven malignant progression of NB were elucidated at single-cell and metabolite flux dimensions. Subsequently, a promising metabolic reprogramming-associated prognostic signature (MPS) and individualized therapeutic approaches based on MPS-stratification were developed and further validated independently using pre-clinical models. Results: MPS-identified MPS-I NB showed significantly higher activity of metabolic reprogramming than MPS-II counterparts. MPS demonstrated improved accuracy compared to current clinical characteristics [AUC: 0.915 vs. 0.657 (MYCN), 0.713 (INSS-stage), and 0.808 (INRG-stratification)] in predicting prognosis. AZD7762 and etoposide were identified as potent therapeutics against MPS-I and II NB, respectively. Subsequent biological tests revealed AZD7762 substantially inhibited growth, migration, and invasion of MPS-I NB cells, more effectively than that of MPS-II cells. Conversely, etoposide had better therapeutic effects on MPS-II NB cells. More encouragingly, AZD7762 and etoposide significantly inhibited in-vivo subcutaneous tumorigenesis, proliferation, and pulmonary metastasis in MPS-I and MPS-II samples, respectively; thereby prolonging survival of tumor-bearing mice. Mechanistically, AZD7762 and etoposide-induced apoptosis of the MPS-I and MPS-II cells, respectively, through mitochondria-dependent pathways; and MPS-I NB resisted etoposide-induced apoptosis by addiction of glutamate metabolism and acetyl coenzyme A. MPS-I NB progression was fueled by multiple metabolic reprogramming-driven factors including multidrug resistance, immunosuppressive and tumor-promoting inflammatory microenvironments. Immunologically, MPS-I NB suppressed immune cells via MIF and THBS signaling pathways. Metabolically, the malignant proliferation of MPS-I NB cells was remarkably supported by reprogrammed glutamate metabolism, tricarboxylic acid cycle, urea cycle, etc. Furthermore, MPS-I NB cells manifested a distinct tumor-promoting developmental lineage and self-communication patterns, as evidenced by enhanced oncogenic signaling pathways activated with development and self-communications. Conclusions: This study provides deep insights into the molecular mechanisms underlying metabolic reprogramming-mediated malignant progression of NB. It also sheds light on developing targeted medications guided by the novel precise risk prognostication approaches, which could contribute to a significantly improved therapeutic strategy for NB.
    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
    靶向治疗对于遗传驱动因素阳性的晚期结直肠癌(CRC)至关重要。随着深度测序技术和新型靶向药物的进步,现有的标准分子病理学检测系统和治疗策略已不能满足对晚期CRC患者进行精心管理的要求.因此,罕见的遗传变异在临床实践中需要诊断和靶向治疗。罕见的基因突变,扩增,和重排通常与预后差和对常规治疗的反应差相关。本文综述了罕见遗传变异的临床诊断和治疗,在包括erb-b2受体酪氨酸激酶2(ERBB2)的基因中,B-Raf原癌基因,丝氨酸/苏氨酸激酶(BRAF),ALK受体酪氨酸激酶/ROS原癌基因1,受体酪氨酸激酶(ALK/ROS1),神经营养受体酪氨酸激酶(NTRKs),ret原癌基因(RET),成纤维细胞生长因子受体2(FGFR2),和表皮生长因子受体(EGFR),以增强对罕见遗传变异患者的理解并确定更准确的个性化治疗方法。
    Targeted therapy is crucial for advanced colorectal cancer (CRC) positive for genetic drivers. With advances in deep sequencing technology and new targeted drugs, existing standard molecular pathological detection systems and therapeutic strategies can no longer meet the requirements for careful management of patients with advanced CRC. Thus, rare genetic variations require diagnosis and targeted therapy in clinical practice. Rare gene mutations, amplifications, and rearrangements are usually associated with poor prognosis and poor response to conventional therapy. This review summarizes the clinical diagnosis and treatment of rare genetic variations, in genes including erb-b2 receptor tyrosine kinase 2 (ERBB2), B-Raf proto-oncogene, serine/threonine kinase (BRAF), ALK receptor tyrosine kinase/ROS proto-oncogene 1, receptor tyrosine kinase (ALK/ROS1), neurotrophic receptor tyrosine kinases (NTRKs), ret proto-oncogene (RET), fibroblast growth factor receptor 2 (FGFR2), and epidermal growth factor receptor (EGFR), to enhance understanding and identify more accurate personalized treatments for patients with rare genetic variations.
    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
    Ferroptosis,一种受调节的细胞死亡形式,与铁依赖性脂质过氧化密切相关。最近的证据强烈支持铁凋亡的诱导作为治疗对常规疗法有抗性的癌症的有希望的策略。铁凋亡调节的关键参与者是铁凋亡抑制蛋白1(FSP1),其通过促进辅酶Q10的抗氧化剂形式的产生来促进癌细胞抗性。值得注意的是,FSP1独立于谷胱甘肽(GSH)和谷胱甘肽过氧化物酶-4途径赋予铁死亡抗性。因此,靶向FSP1以削弱其对铁凋亡的抑制作用可能是治疗难治性癌症的可行策略。这篇综述旨在阐明铁死亡的分子机制,FSP1抑制铁凋亡的具体途径以及FSP1抑制剂对癌细胞的影响。
    Ferroptosis, a regulated form of cell death, is intricately linked to iron‑dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase‑4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer cells.
    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
    针对干燥综合征(SS)的靶向治疗已成为临床医生的重要关注点。多组学广泛的孟德尔随机化(MR)分析为识别潜在的药物靶标提供了新思路。
    我们进行了基于汇总数据的孟德尔随机化(SMR)分析,以通过整合DNA甲基化来评估与SS相关的治疗靶标,基因表达和蛋白质数量性状基因座(mQTL,eQTL,和pQTL,分别)。与SS的遗传关联来源于FinnGen研究(发现)和GWAS目录(复制)。采用共定位分析来确定两种潜在相关表型在给定区域中是否共享相同的遗传因素。此外,深入研究DNA甲基化之间的潜在调控,基因表达,和蛋白质丰富,我们进行了MR分析,以探讨候选基因甲基化与表达之间的因果关系,以及基因表达和蛋白质丰度之间。进一步采用药物预测和分子对接来验证候选药物靶标的药理活性。
    在整合多组数据后,我们确定了与SS风险相关的三个基因:TNFAIP3,BTN3A1和PLAU.BTN3A1中cg22068371的甲基化与蛋白水平呈正相关,与cg22068371甲基化对SS风险的负面影响一致。此外,PLAU(cg04939496)基因甲基化与表达呈正相关,以及表达和蛋白质水平之间。这种一致性阐明了PLAU在DNA甲基化时对SS风险的促进作用,基因表达,和蛋白质水平。在蛋白质水平,遗传预测的TNFAIP3(OR2.47,95%CI1.56-3.92)与SS风险呈正相关,而BTN3A1(OR2.96E-03,95%CI2.63E-04-3.33E-02)与SS风险呈负相关。分子对接显示候选药物和靶蛋白的稳定结合。
    我们的研究揭示了治疗SS的有希望的治疗目标,为SS的靶向治疗提供有价值的见解。然而,有必要通过未来的实验进一步验证.
    UNASSIGNED: Targeted therapy for Sjögren\'s syndrome (SS) has become an important focus for clinicians. Multi-omics-wide Mendelian randomization (MR) analyses have provided new ideas for identifying potential drug targets.
    UNASSIGNED: We conducted summary-data-based Mendelian randomization (SMR) analysis to evaluate therapeutic targets associated with SS by integrating DNA methylation, gene expression and protein quantitative trait loci (mQTL, eQTL, and pQTL, respectively). Genetic associations with SS were derived from the FinnGen study (discovery) and the GWAS catalog (replication). Colocalization analyses were employed to determine whether two potentially relevant phenotypes share the same genetic factors in a given region. Moreover, to delve deeper into potential regulation among DNA methylation, gene expression, and protein abundance, we conducted MR analysis to explore the causal relationship between candidate gene methylation and expression, as well as between gene expression and protein abundance. Drug prediction and molecular docking were further employed to validate the pharmacological activity of the candidate drug targets.
    UNASSIGNED: Upon integrating the multi-omics data, we identified three genes associated with SS risk: TNFAIP3, BTN3A1, and PLAU. The methylation of cg22068371 in BTN3A1 was positively associated with protein levels, consistent with the negative effect of cg22068371 methylation on the risk of SS. Additionally, positive correlations were observed between the gene methylation of PLAU (cg04939496) and expression, as well as between expression and protein levels. This consistency elucidates the promotional effects of PLAU on SS risk at the DNA methylation, gene expression, and protein levels. At the protein level, genetically predicted TNFAIP3 (OR 2.47, 95% CI 1.56-3.92) was positively associated with SS risk, while BTN3A1 (OR 2.96E-03, 95% CI 2.63E-04-3.33E-02) was negatively associated with SS risk. Molecular docking showed stable binding for candidate drugs and target proteins.
    UNASSIGNED: Our study reveals promising therapeutic targets for the treatment of SS, providing valuable insights into targeted therapy for SS. However, further validation through future experiments is warranted.
    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
    Tau蛋白是一种微管相关蛋白,广泛分布于中枢神经系统,维持和调节神经元的形态和功能。Tau蛋白在神经退行性疾病中异常聚集并形成神经原纤维缠结,破坏神经元的结构和功能,导致神经元死亡,这引发了神经系统疾病的开始和进展。tau蛋白在神经退行性疾病中的聚集与翻译后修饰有关,这可能会影响亲水性,空间构象,和tau蛋白的稳定性,促进tau蛋白聚集和神经原纤维缠结的形成。因此,研究tau蛋白在神经退行性疾病中的作用和异常聚集的机制对于理解神经退行性疾病的机制和寻找治疗方法具有重要意义。这篇综述描述了tau蛋白促进神经退行性疾病的可能机制。tau蛋白翻译后修饰及相关影响因素,以及与tau蛋白相关的药物发现和开发的现状,这可能有助于开发新的治疗方法来缓解或治疗神经退行性疾病。
    Tau protein is a microtubule-associated protein that is widely distributed in the central nervous system and maintains and regulates neuronal morphology and function. Tau protein aggregates abnormally and forms neurofibrillary tangles in neurodegenerative diseases, disrupting the structure and function of neurons and leading to neuronal death, which triggers the initiation and progression of neurological disorders. The aggregation of tau protein in neurodegenerative diseases is associated with post-translational modifications, which may affect the hydrophilicity, spatial conformation, and stability of tau protein, promoting tau protein aggregation and the formation of neurofibrillary tangles. Therefore, studying the role of tau protein in neurodegenerative diseases and the mechanism of aberrant aggregation is important for understanding the mechanism of neurodegenerative diseases and finding therapeutic approaches. This review describes the possible mechanisms by which tau protein promotes neurodegenerative diseases, the post-translational modifications of tau protein and associated influencing factors, and the current status of drug discovery and development related to tau protein, which may contribute to the development of new therapeutic approaches to alleviate or treat neurodegenerative diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号