Mitophagy

线粒体自噬
  • 文章类型: Journal Article
    糖尿病(DM)是一种进行性、以高氧化应激为特征的慢性代谢紊乱,会导致心脏损伤.过量的活性氧(ROS)对蛋白质的甲硫氨酸亚砜化(MetO)会损害必需细胞蛋白质的基本功能,导致心力衰竭。蛋氨酸亚砜还原酶B2(MsrB2)可以逆转线粒体蛋白中MetO的氧化诱导,所以我们研究了它在糖尿病心肌病中的作用。我们观察到糖尿病小鼠模型中DM诱导的心脏损伤的特征是ROS增加,线粒体结构病理学增加的蛋白质MetO,和心脏纤维化。此外,MsrB2在小鼠DM心肌细胞中显著升高,支持诱导保护过程。Further,MsrB2直接诱导心肌细胞中的Parkin和LC3激活(线粒体自噬标志物)。在MsrB2基因敲除小鼠表现出异常的电生理功能,通过ECG分析确定。组织学分析证实在MsrB2敲除DM小鼠中增加的心脏纤维化和破坏的心脏组织。然后,我们证实了我们在人类DM心脏样本中的发现。我们的研究表明,心脏中MsrB2表达的增加可以预防糖尿病性心肌病。
    Diabetes mellitus (DM) is a progressive, chronic metabolic disorder characterized by high oxidative stress, which can lead to cardiac damage. Methionine sulfoxylation (MetO) of proteins by excessive reactive oxygen species (ROS) can impair the basic functionality of essential cellular proteins, contributing to heart failure. Methionine sulfoxide reductase B2 (MsrB2) can reverse oxidation induced MetO in mitochondrial proteins, so we investigated its role in diabetic cardiomyopathy. We observed that DM-induced heart damage in diabetic mice model is characterized by increased ROS, increased protein MetO with mitochondria structural pathology, and cardiac fibrosis. In addition, MsrB2 was significantly increased in mouse DM cardiomyocytes, supporting the induction of a protective process. Further, MsrB2 directly induces Parkin and LC3 activation (mitophagy markers) in cardiomyocytes. In MsrB2, knockout mice displayed abnormal electrophysiological function, as determined by ECG analysis. Histological analysis confirmed increased cardiac fibrosis and disrupted cardiac tissue in MsrB2 knockout DM mice. We then corroborated our findings in human DM heart samples. Our study demonstrates that increased MsrB2 expression in the heart protects against diabetic cardiomyopathy.
    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
    越来越多的证据支持细胞竞争,多细胞生物中至关重要的选择和质量控制机制,参与肿瘤发生和发展;然而,细胞竞争与肿瘤耐药性之间的相关机制贡献仍不明确.在我们的研究中,基于对列维替尼耐药的肝细胞癌(HCC)细胞通过重编程能量代谢显示出明显的竞争生长优势。机械上,Lenvatinib耐药HCC细胞中BCL2相互作用蛋白3(BNIP3)介导的线粒体自噬的过度激活通过将能量产生从线粒体氧化磷酸化转移到糖酵解来促进糖酵解通量,通过调节AMP激活的蛋白激酶(AMPK)-烯醇化酶2(ENO2)信号,永久保持耐乐替尼肝癌细胞相对于敏感肝癌细胞的竞争优势。值得注意的是,BNIP3抑制显著敏化乐伐替尼在HCC中的抗肿瘤功效。我们的发现强调了BNIP3-AMPK-ENO2信号传导在通过调节能量代谢重编程维持耐来维替尼肝癌细胞的竞争性结果中的重要作用;同时,这项工作认为BNIP3是克服HCC耐药性的有希望的靶标。
    An increasing evidence supports that cell competition, a vital selection and quality control mechanism in multicellular organisms, is involved in tumorigenesis and development; however, the mechanistic contributions to the association between cell competition and tumor drug resistance remain ill-defined. In our study, based on a contructed lenvitinib-resistant hepatocellular carcinoma (HCC) cells display obvious competitive growth dominance over sensitive cells through reprogramming energy metabolism. Mechanistically, the hyperactivation of BCL2 interacting protein3 (BNIP3) -mediated mitophagy in lenvatinib-resistant HCC cells promotes glycolytic flux via shifting energy production from mitochondrial oxidative phosphorylation to glycolysis, by regulating AMP-activated protein kinase (AMPK) -enolase 2 (ENO2) signaling, which perpetually maintaining lenvatinib-resistant HCC cells\' competitive advantage over sensitive HCC cells. Of note, BNIP3 inhibition significantly sensitized the anti-tumor efficacy of lenvatinib in HCC. Our findings emphasize a vital role for BNIP3-AMPK-ENO2 signaling in maintaining the competitive outcome of lenvitinib-resistant HCC cells via regulating energy metabolism reprogramming; meanwhile, this work recognizes BNIP3 as a promising target to overcome HCC drug resistance.
    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: Retraction of Publication
    上述文章发表后,一位有关的读者提请编辑注意,图中某些免疫荧光数据。1H,图中的TUNEL测定数据。2A,图中的细胞基因组c泄漏测定数据。2H,图中心磷脂图像的染色。2H,无花果中的薄片染色数据。3A,和图中的免疫荧光测定数据。3F和5D与由不同研究机构的不同作者撰写的其他文章中以不同形式出现的数据惊人地相似,这些文章在本论文提交给肿瘤学报告之前已经在其他地方发表。或者大约在同一时间正在考虑出版(其中一些已经被撤回)。此外,图中的数据面板中注意到数据的重叠部分。3D和F,这样,旨在表示来自不同进行的实验的结果的数据显然来自相同的原始来源。鉴于这些数据中的某些数据在提交本文发表之前显然已经发表,鉴于对所提供的数据总体上缺乏信心,《肿瘤学报告》的编辑已经决定,这篇论文应该从期刊上撤回。作者被要求解释这些担忧,但编辑部没有收到回复。编辑对读者造成的不便表示歉意。[肿瘤学报告39:1671-1681,2018;DOI:10.3892/or.2018.6252]。
    Following the publication of the above article, a concerned reader drew to the Editor\'s attention that certain of the immunofluorescence data featured in Fig. 1H, TUNEL assay data in Fig. 2A, cytochome c leakage assay data in Fig. 2H, staining of cardiolipin images in Fig. 2H, lamellipodia‑stained data in Fig. 3A, and immunofluorescence assay data in Figs. 3F and 5D were strikingly similar to data appearing in different form in other articles written by different authors at different research institutes that had either already been published elsewhere prior to the submission of this paper to Oncology Reports, or were under consideration for publication at around the same time (several of which have now been retracted). In addition, overlapping sections of data were noted within the data panels in Fig. 3D and F, such that data which were intended to represent the results from differently performed experiments had apparently been derived from the same original source(s). In view of the fact that certain of these data had already apparently been published prior to the submission of this article for publication, and in view of an overall lack of confidence in the presented data, the Editor of Oncology Reports has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Oncology Reports 39: 1671‑1681, 2018; DOI: 10.3892/or.2018.6252].
    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
    肺癌,全球公认的恶性肿瘤相关发病率和死亡率的主要原因,以其高流行率和高杀伤力为标志,引起医学界的广泛关注。线粒体自噬是一个关键的细胞过程,在调节代谢和确保细胞内的质量控制中起着至关重要的作用。它与肺癌的相关性引起了研究人员和科学家的极大关注。线粒体自噬参与肺癌包括其启动,programming,转移性播散和治疗。线粒体自噬的调控景观是复杂的,涉及许多可能在肿瘤环境中表现出异常改变或突变的信号蛋白和途径。在治疗领域,线粒体自噬的调节被认为是决定癌症化疗的关键,放射治疗,其他治疗方案,和抗药性。当代研究旨在利用线粒体自噬调节剂,抑制剂和活化剂,在治疗策略中,重点是实现特异性,以最大程度地减少对健康细胞群体的附带损害。此外,与线粒体自噬相关的分子成分和途径,作为潜在的生物标志物,为提高诊断准确性提供有希望的途径,预后评估,和预测肺癌的治疗反应。未来的努力还将涉及研究线粒体自噬对肿瘤微环境中免疫细胞的组成和功能的影响。旨在增强我们对线粒体自噬如何调节对肺癌的免疫反应的理解。本综述旨在全面概述线粒体自噬在肿瘤发生中的作用的最新进展。子代和转移,以及线粒体自噬对肺癌治疗的影响。我们还讨论了线粒体自噬在肺癌领域的未来研究方向。
    Lung cancer, recognized globally as a leading cause of malignancy-associated morbidity and mortality, is marked by its high prevalence and lethality, garnering extensive attention within the medical community. Mitophagy is a critical cellular process that plays a crucial role in regulating metabolism and ensuring quality control within cells. Its relevance to lung cancer has garnered significant attention among researchers and scientists. Mitophagy\'s involvement in lung cancer encompasses its initiation, progression, metastatic dissemination and treatment. The regulatory landscape of mitophagy is complex, involving numerous signaling proteins and pathways that may exhibit aberrant alterations or mutations within the tumor environment. In the field of treatment, the regulation of mitophagy is considered key to determining cancer chemotherapy, radiation therapy, other treatment options, and drug resistance. Contemporary investigations are directed towards harnessing mitophagy modulators, both inhibitors and activators, in therapeutic strategies, with an emphasis on achieving specificity to minimize collateral damage to healthy cellular populations. Furthermore, molecular constituents and pathways affiliated with mitophagy, serving as potential biomarkers, offer promising avenues for enhancing diagnostic accuracy, prognostic assessment, and prediction of therapeutic responses in lung cancer. Future endeavors will also involve investigating the impact of mitophagy on the composition and function of immune cells within the tumor microenvironment, aiming to enhance our understanding of how mitophagy modulates the immune response to lung cancer. This review aims to comprehensively overview recent advancements about the role of mitophagy in the tumor genesis, progenesis and metastasis, and the impact of mitophagy on the treatment of lung cancer. We also discussed the future research direction of mitophagy in the field of lung cancer.
    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
    肾小管上皮细胞(RTEC)损伤标志着脓毒症相关急性肾损伤(SA-AKI)的发生和进展。最近对线粒体的研究表明,线粒体自噬在减轻RTEC损伤中起着至关重要的生理作用,并且在SA-AKI中受到炎症反应的逐渐抑制。然而,炎症影响线粒体自噬的机制仍然知之甚少。我们检查了巨噬细胞移动抑制因子(MIF)一种促炎蛋白,通过研究MIF被抑制或过表达时的蛋白质-蛋白质相互作用来影响线粒体自噬的PINK1-Parkin途径。令人惊讶的是,发现高水平的MIF直接与PINK1结合,破坏了其与Parkin的相互作用。这种干扰阻碍了Parkin向线粒体的募集,并阻碍了线粒体自噬的启动。此外,这一结果导致了RTECs的显著凋亡,这可能,然而,可以通过MIF抑制剂ISO-1和/或新的线粒体自噬激活剂T0467逆转。这些发现强调了MIF通过破坏PINK1和Parkin之间的相互作用对肾脏损害的有害影响。以及ISO-1和T0467在缓解SA-AKI方面的治疗潜力。这项研究为通过靶向MIF和线粒体自噬治疗SA-AKI提供了新的视角。
    Damage to renal tubular epithelial cells (RTECs) signaled the onset and progression of sepsis-associated acute kidney injury (SA-AKI). Recent research on mitochondria has revealed that mitophagy plays a crucial physiological role in alleviating injury to RTECs and it is suppressed progressively by the inflammation response in SA-AKI. However, the mechanism by which inflammation influences mitophagy remains poorly understood. We examined how macrophage migration inhibitory factor (MIF), a pro-inflammatory protein, influences the PINK1-Parkin pathway of mitophagy by studying protein-protein interactions when MIF was inhibited or overexpressed. Surprisingly, elevated levels of MIF were found to directly bind to PINK1, disrupting its interaction with Parkin. This interference hindered the recruitment of Parkin to mitochondria and impeded the initiation of mitophagy. Furthermore, this outcome led to significant apoptosis of RTECs, which could, however, be reversed by an MIF inhibitor ISO-1 and/or a new mitophagy activator T0467. These findings highlight the detrimental impact of MIF on renal damage through its disruption of the interaction between PINK1 and Parkin, and the therapeutic potential of ISO-1 and T0467 in mitigating SA-AKI. This study offers a fresh perspective on treating SA-AKI by targeting MIF and mitophagy.
    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
    肝癌是世界范围内癌症相关死亡的主要原因之一。癌症干细胞(CSC)是促进肿瘤生长的癌细胞的独特子集,维护,和治疗抗性,导致复发。在目前的工作中,含有1,3-噻唑烷-2-硫酮(RCT)的钌络合物的能力,用化学式[Ru(tzdt)(bipy)(dppb)]PF6,在人肝癌HepG2细胞中探索抑制肝CSC。RCT对固体和血液癌细胞系表现出有效的细胞毒性,并降低了克隆形成潜力,CD133+和CD44高细胞百分比和HepG2细胞的肿瘤球体生长。RCT还抑制细胞运动,如在伤口愈合测定和transwell细胞迁移测定中观察到的。RCT降低了Akt1,磷酸-Akt(Ser473)的水平,磷酸化Akt(Thr308),磷酸-mTOR(Ser2448),和磷酸-S6(Ser235/Ser236)在HepG2细胞,指示干扰Akt/mTOR信令是RCT的作用机制。在RCT处理的HepG2细胞中,活性caspase-3和裂解的PARP(Asp214)的水平增加,表明凋亡细胞死亡的诱导。此外,在mt-Keima转染的小鼠胚胎成纤维细胞(MEF)细胞模型中,RCT调节HepG2细胞中的自噬标志物LC3B和p62/SQSTM1,并增加线粒体自噬,自噬抑制剂部分阻止了RCT诱导的细胞毒性。此外,突变型Atg5-/-MEFs和PentaKOHeLa细胞(具有五个自噬受体敲除的人宫颈腺癌)对RCT细胞毒性的敏感性低于其亲本细胞系,表明RCT诱导自噬介导的细胞死亡。一起来看,这些数据表明RCT是一种对CSC具有抑制作用的新型潜在抗肝癌药物.
    Hepatic cancer is one of the main causes of cancer-related death worldwide. Cancer stem cells (CSCs) are a unique subset of cancer cells that promote tumour growth, maintenance, and therapeutic resistance, leading to recurrence. In the present work, the ability of a ruthenium complex containing 1,3-thiazolidine-2-thione (RCT), with the chemical formula [Ru(tzdt)(bipy)(dppb)]PF6, to inhibit hepatic CSCs was explored in human hepatocellular carcinoma HepG2 cells. RCT exhibited potent cytotoxicity to solid and haematological cancer cell lines and reduced the clonogenic potential, CD133+ and CD44high cell percentages and tumour spheroid growth of HepG2 cells. RCT also inhibited cell motility, as observed in the wound healing assay and transwell cell migration assay. RCT reduced the levels of Akt1, phospho-Akt (Ser473), phospho-Akt (Thr308), phospho-mTOR (Ser2448), and phospho-S6 (Ser235/Ser236) in HepG2 cells, indicating that interfering with Akt/mTOR signalling is a mechanism of action of RCT. The levels of active caspase-3 and cleaved PARP (Asp214) were increased in RCT-treated HepG2 cells, indicating the induction of apoptotic cell death. In addition, RCT modulated the autophagy markers LC3B and p62/SQSTM1 in HepG2 cells and increased mitophagy in a mt-Keima-transfected mouse embryonic fibroblast (MEF) cell model, and RCT-induced cytotoxicity was partially prevented by autophagy inhibitors. Furthermore, mutant Atg5-/- MEFs and PentaKO HeLa cells (human cervical adenocarcinoma with five autophagy receptor knockouts) were less sensitive to RCT cytotoxicity than their parental cell lines, indicating that RCT induces autophagy-mediated cell death. Taken together, these data indicate that RCT is a novel potential anti-liver cancer drug with a suppressive effect on CSCs.
    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
    卵巢衰老是以卵母细胞数量和质量下降为特征的复杂过程,直接影响生育率和整体福祉。最近的研究已经确定线粒体是卵巢衰老的关键参与者,影响控制这个复杂过程的各种标志和途径。在这次审查中,我们讨论了线粒体在决定卵巢命运中的多方面作用,并概述了线粒体促进卵巢衰老的关键机制。具体来说,我们强调通过创新的治疗方法靶向线粒体功能障碍的潜力,包括抗氧化剂,代谢改善,促进生物发生,线粒体自噬增强,线粒体转移,和传统中药。这些策略有望成为减轻与年龄相关的生育能力下降和保护卵巢健康的有效手段。从该领域的先进研究中汲取见解,这篇综述提供了一个更深入的了解线粒体功能和卵巢衰老之间的复杂的相互作用,为开发旨在保持生育能力和增强整体生殖健康的新型治疗干预措施提供有价值的观点。
    Ovarian aging is a complex process characterized by a decline in oocyte quantity and quality, directly impacting fertility and overall well-being. Recent researches have identified mitochondria as pivotal players in the aging of ovaries, influencing various hallmarks and pathways governing this intricate process. In this review, we discuss the multifaceted role of mitochondria in determining ovarian fate, and outline the pivotal mechanisms through which mitochondria contribute to ovarian aging. Specifically, we emphasize the potential of targeting mitochondrial dysfunction through innovative therapeutic approaches, including antioxidants, metabolic improvement, biogenesis promotion, mitophagy enhancement, mitochondrial transfer, and traditional Chinese medicine. These strategies hold promise as effective means to mitigate age-related fertility decline and preserve ovarian health. Drawing insights from advanced researches in the field, this review provides a deeper understanding of the intricate interplay between mitochondrial function and ovarian aging, offering valuable perspectives for the development of novel therapeutic interventions aimed at preserving fertility and enhancing overall reproductive health.
    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
    这篇综述严格调查了与阿尔茨海默病相关的早期大脑变化,早在临床症状出现之前就显现出来了。它提供了钙(Ca2+)稳态失调的证据,伴随着线粒体功能障碍和异常的自噬过程,在无症状期间可能会推动疾病的进展,临床前阶段。了解在这个关键时期展开的复杂的分子相互作用为识别新的治疗靶点提供了一个窗口。从而推进神经退行性疾病的治疗。该评论深入研究了由于Ca2平衡的破坏而导致的分子变化的既定和新兴见解,为认知衰退和神经退化奠定基础。
    This review rigorously investigates the early cerebral changes associated with Alzheimer\'s disease, which manifest long before clinical symptoms arise. It presents evidence that the dysregulation of calcium (Ca2+) homeostasis, along with mitochondrial dysfunction and aberrant autophagic processes, may drive the disease\'s progression during its asymptomatic, preclinical stage. Understanding the intricate molecular interplay that unfolds during this critical period offers a window into identifying novel therapeutic targets, thereby advancing the treatment of neurodegenerative disorders. The review delves into both established and emerging insights into the molecular alterations precipitated by the disruption of Ca2+ balance, setting the stage for cognitive decline and neurodegeneration.
    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:脓毒症相关急性肾损伤(SA-AKI)是危重患者常见的并发症。本研究的目的是阐明Micheliolide(MCL)的潜在保护活性及其对SA-AKI的作用机制。
    方法:在脂多糖(LPS)处理的HK2细胞和SA-AKI小鼠模型中研究MCL对SA-AKI的保护潜力。通过检测活性氧和膜电位来确定线粒体损伤。通过在HK2细胞中转染Nrf2-shRNA来实现Nrf2沉默,和Nrf2抑制剂,在SA-AKI小鼠中采用ML385。通过检测与炎症相关的标志来评估MCL抗SA-AKI的机制。通过蛋白质印迹的线粒体自噬和Nrf2途径,免疫组织化学,和酶联免疫吸附测定。
    结果:MCL增强了生存能力,抑制细胞凋亡,在LPS处理的HK2细胞中,炎症细胞因子水平降低,线粒体损伤改善,并改善SA-AKI小鼠的肾损伤。此外,MCL可以通过增强线粒体自噬来降低NLRP3炎性体的活化。此外,Nrf2缺陷降低了MCL对NLRP3炎性体激活的抑制作用,阻断了MCL对LPS处理的HK2细胞线粒体自噬的促进作用,对于SA-AKI小鼠中的ML385治疗是一致的。
    结论:MCL可能靶向Nrf2,并通过增强线粒体自噬进一步降低NLRP3炎性体的激活,这减轻了SA-AKI。
    BACKGROUND: Sepsis-associated acute kidney injury (SA-AKI) represents a frequent complication of in critically ill patients. The objective of this study is to illuminate the potential protective activity of Micheliolide (MCL) and its behind mechanism against SA-AKI.
    METHODS: The protective potential of MCL on SA-AKI was investigated in lipopolysaccharide (LPS) treated HK2 cells and SA-AKI mice model. The mitochondrial damage was determined by detection of reactive oxygen species and membrane potential. The Nrf2 silencing was achieved by transfection of Nrf2-shRNA in HK2 cells, and Nrf2 inhibitor, ML385 was employed in SA-AKI mice. The mechanism of MCL against SA-AKI was evaluated through detecting hallmarks related to inflammation, mitophagy and Nrf2 pathway via western blotting, immunohistochemistry, and enzyme linked immunosorbent assay.
    RESULTS: MCL enhanced viability, suppressed apoptosis, decreased inflammatory cytokine levels and improved mitochondrial damage in LPS-treated HK2 cells, and ameliorated renal injury in SA-AKI mice. Moreover, MCL could reduce the activation of NLRP3 inflammasome via enhancing mitophagy. Additionally, Nrf2 deficiency reduced the suppression effect of MCL on NLRP3 inflammasome activation and blocked the facilitation effect of MCL on mitophagy in LPS-treated HK2 cells, the consistent is true for ML385 treatment in SA-AKI mice.
    CONCLUSIONS: MCL might target Nrf2 and further reduce the NLRP3 inflammasome activation via enhancing mitophagy, which alleviated SA-AKI.
    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
    自噬和线粒体自噬在理解糖尿病心脏病(DHC)的病理学方面提出了尚未解决的挑战,其中包括与糖尿病和相关心肌病相关的一系列复杂的心血管问题。尽管在降低心血管疾病(CVD)死亡率方面取得了重大进展,心力衰竭仍然是糖尿病患者发病率增加的主要原因.这些细胞过程对于维持细胞平衡和去除受损或功能失调的组件至关重要。它们参与糖尿病性心脏病的发展使它们成为诊断和治疗的有吸引力的目标。虽然有多种常规诊断和治疗策略可用,DHC继续提出重大挑战。即时诊断,在纳米生物传感技术的支持下,为这些复杂的场景提供了一个有希望的替代方案。尽管常规药物已广泛用于DHC患者,他们对各种生理方面提出了一些担忧。现代医学非常重视纳米技术在DHC中靶向自噬和线粒体自噬的应用,提供了一种有希望的方法来提供超越传统疗法限制的药物。本文旨在探讨自噬之间的潜在联系,线粒体自噬和DHC,同时还讨论了基于纳米技术的专门针对这些分子途径的theranostic干预措施的前景。
    Autophagy and mitophagy pose unresolved challenges in understanding the pathology of diabetic heart condition (DHC), which encompasses a complex range of cardiovascular issues linked to diabetes and associated cardiomyopathies. Despite significant progress in reducing mortality rates from cardiovascular diseases (CVDs), heart failure remains a major cause of increased morbidity among diabetic patients. These cellular processes are essential for maintaining cellular balance and removing damaged or dysfunctional components, and their involvement in the development of diabetic heart disease makes them attractive targets for diagnosis and treatment. While a variety of conventional diagnostic and therapeutic strategies are available, DHC continues to present a significant challenge. Point-of-care diagnostics, supported by nanobiosensing techniques, offer a promising alternative for these complex scenarios. Although conventional medications have been widely used in DHC patients, they raise several concerns regarding various physiological aspects. Modern medicine places great emphasis on the application of nanotechnology to target autophagy and mitophagy in DHC, offering a promising approach to deliver drugs beyond the limitations of traditional therapies. This article aims to explore the potential connections between autophagy, mitophagy and DHC, while also discussing the promise of nanotechnology-based theranostic interventions that specifically target these molecular pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号