1
|
Feng L, Li B, Yong SS, Wen X, Tian Z. The emerging role of exercise in Alzheimer's disease: Focus on mitochondrial function. Ageing Res Rev 2024; 101:102486. [PMID: 39243893 DOI: 10.1016/j.arr.2024.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory impairment and cognitive dysfunction, which eventually leads to the disability and mortality of older adults. Although the precise mechanisms by which age promotes the development of AD remains poorly understood, mitochondrial dysfunction plays a central role in the development of AD. Currently, there is no effective treatment for this debilitating disease. It is well accepted that exercise exerts neuroprotective effects by ameliorating mitochondrial dysfunction in the neurons of AD, which involves multiple mechanisms, including mitochondrial dynamics, biogenesis, mitophagy, transport, and signal transduction. In addition, exercise promotes mitochondria communication with other organelles in AD neurons, which should receive more attentions in the future.
Collapse
Affiliation(s)
- Lili Feng
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Bowen Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Su Sean Yong
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Xu Wen
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
2
|
Li N, Qiu G, Xu X, Shen Y, Chen Y. TRIM11 Prevents Ferroptosis in model of asthma by UBE2N-TAX1BP1 signaling. BMC Pulm Med 2024; 24:542. [PMID: 39472837 PMCID: PMC11523820 DOI: 10.1186/s12890-024-03351-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Asthma is a complex chronic respiratory inflammatory disease affected by both genetic and environmental factors. Therefore, our study explored the influence of TRIM11 on asthma and its underlying mechanisms. Our research involved patients diagnosed with asthma and healthy volunteers recruited from our hospital. We observed a reduction in serum TRIM11 expression in asthma patients, which positively correlated with the levels of anti-IgE or IgE. Additionally, both TRIM11 mRNA and protein expression in lung tissue were diminished. The introduction of the TRIM11 gene resulted in a reduction in inflammation in an in vitro asthma model and prevented the development of asthma in a mouse model. Moreover, the TRIM11 gene exhibited a suppressive effect on Ferroptosis and mitigated ROS-induced mitochondrial damage in the asthma model. TRIM11 was found to stimulate UBE2N-TAX1BP1 signaling in the asthma model, with UBE2N being identified as the specific target for TRIM11's effects on Ferroptosis. Furthermore, TRIM11 protein interacted with UBE2N protein and facilitated the dissociation of UBE2N-UBE2N in the asthma model. In conclusion, TRIM11 plays a vital role in preventing Ferroptosis in the asthma model through UBE2N-TAX1BP1 signaling. This indicates that targeting the TRIM11 mechanism could serve as a promising strategy for anti-Ferroptosis immunotherapy in asthma treatment.
Collapse
Affiliation(s)
- Na Li
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China.
| | - Guoqing Qiu
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| | - Xiangqin Xu
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| | - Yan Shen
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| | - Yuming Chen
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| |
Collapse
|
3
|
Chang H, Zhang W, Xu L, Li Z, Lin C, Shen Y, Zhang G, Mao L, Ma C, Liu N, Lu H. Copper aggravated synaptic damage after traumatic brain injury by downregulating BNIP3-mediated mitophagy. Autophagy 2024:1-17. [PMID: 39415457 DOI: 10.1080/15548627.2024.2409613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
Synaptic damage is a crucial pathological process in traumatic brain injury. However, the mechanisms driving this process remain poorly understood. In this report, we demonstrate that the accumulation of damaged mitochondria, resulting from impaired mitphagy, plays a significant role in causing synaptic damage. Moreover, copper induced downregulation of BNIP3 is a key player in regulating mitophagy. DMSA alleviates synaptic damage and mitochondrial dysfunction by promoting urinary excretion of copper. Mechanistically, we find that copper downregulate BNIP3 by increasing the nuclear translocation of NFKB, which is triggered by TRIM25-mediated ubiquitination-dependent degradation of NFKBIA. Our study underscores the importance of copper accumulation in the regulation of BNIP3-mediated mitophagy and suggests that therapeutic targeting of the copper-TRIM25-NFKB-BNIP3 axis holds promise to attenuate synaptic damage after traumatic brain injury.
Collapse
Affiliation(s)
- Hanxiao Chang
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Weiwei Zhang
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Xu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Zheng Li
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Chao Lin
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Yuqi Shen
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Guangjian Zhang
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Lei Mao
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Chencheng Ma
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Ning Liu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Hua Lu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Shen M, Chen M, Chen Y, Yu Y. Mitophagy related diagnostic biomarkers for coronary in-stent restenosis identified using machine learning and bioinformatics. Sci Rep 2024; 14:24137. [PMID: 39406802 PMCID: PMC11480419 DOI: 10.1038/s41598-024-74862-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Percutaneous coronary intervention (PCI) combined with stent implantation is currently one of the most effective treatments for coronary artery disease (CAD). However, in-stent restenosis (ISR) significantly compromises its long-term efficacy. Mitophagy plays a crucial role in vascular homeostasis, yet its role in ISR remains unclear. This study aims to identify mitophagy-related biomarkers for ISR and explore their underlying molecular mechanisms. Through differential gene expression analysis between ISR and Control samples in the combined dataset, 169 differentially expressed genes (DEGs) were identified. Twenty-three differentially expressed mitophagy-related genes (DEMRGs) were identified by intersecting with mitophagy-related genes (MRGs) from the GeneCards, and functional enrichment analysis indicated their significant involvement in mitophagy-related biological processes. Using Weighted Gene Co-expression Network Analysis (WGCNA) and three machine learning algorithms (Logistic-LASSO, RF, and SVM-RFE), LRRK2, and ANKRD13A were identified as mitophagy-related biomarkers for ISR. The nomogram based on these two genes also exhibited promising diagnostic performance for ISR. Gene Set Enrichment Analysis (GSEA) as well as immune infiltration analyses showed that these two genes were closely associated with immune and inflammatory responses in ISR. Furthermore, potential small molecule compounds with therapeutic implications for ISR were predicted using the connectivity Map (cMAP) database. This study systematically investigated mitophagy-related biomarkers for ISR and their potential biological functions, providing new insights into early diagnosis and precision treatment strategies for ISR.
Collapse
Affiliation(s)
- Ming Shen
- Department of Cardiology, the 926th Hospital of the Joint Logistic Support Force of PLA, Affiliated Hospital of Kunming University of Science and Technology, Kaiyuan, 661600, Yunnan, China.
- Department of Cardiology, the 920th Hospital of the Joint Logistic Support Force of PLA, Kunming, 650032, Yunnan, China.
| | - Meixian Chen
- Department of Cardiology, Fuzong Clinical Medical College of Fujian Medical University (900th Hospital of the Joint Logistic Support Force of PLA), Fuzhou, 350025, Fujian, China
| | - Yu Chen
- Department of Cardiology, the 920th Hospital of the Joint Logistic Support Force of PLA, Kunming, 650032, Yunnan, China
| | - Yunhua Yu
- Department of Geriatric, Fuzong Clinical Medical College of Fujian Medical University (900th Hospital of the Joint Logistic Support Force of PLA), Fuzhou, 350025, Fujian, China.
| |
Collapse
|
5
|
Cong X, Zhang Z, Li H, Yang YG, Zhang Y, Sun T. Nanocarriers for targeted drug delivery in the vascular system: focus on endothelium. J Nanobiotechnology 2024; 22:620. [PMID: 39396002 PMCID: PMC11470712 DOI: 10.1186/s12951-024-02892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
Endothelial cells (ECs) are pivotal in maintaining vascular health, regulating hemodynamics, and modulating inflammatory responses. Nanocarriers hold transformative potential for precise drug delivery within the vascular system, particularly targeting ECs for therapeutic purposes. However, the complex interactions between vascular ECs and nanocarriers present significant challenges for the development and clinical translation of nanotherapeutics. This review assesses recent advancements and key strategies in employing nanocarriers for drug delivery to vascular ECs. It suggested that through precise physicochemical design and surface modifications, nanocarriers can enhance targeting specificity and improve drug internalization efficiency in ECs. Additionally, we elaborated on the applications of nanocarriers specifically designed for targeting ECs in the treatment of cardiovascular diseases, cancer metastasis, and inflammatory disorders. Despite these advancements, safety concerns, the complexity of in vivo processes, and the challenge of achieving subcellular drug delivery remain significant obstacles to the effective targeting of ECs with nanocarriers. A comprehensive understanding of endothelial cell biology and its interaction with nanocarriers is crucial for realizing the full potential of targeted drug delivery systems.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - Zebin Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, 130012, Jilin, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China.
| |
Collapse
|
6
|
Zdanowicz A, Grosicka-Maciąg E. The Interplay between Autophagy and Mitochondria in Cancer. Int J Mol Sci 2024; 25:9143. [PMID: 39273093 PMCID: PMC11395105 DOI: 10.3390/ijms25179143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Besides producing cellular energy, mitochondria are crucial in controlling oxidative stress and modulating cellular metabolism, particularly under stressful conditions. A key aspect of this regulatory role involves the recycling process of autophagy, which helps to sustain energy homeostasis. Autophagy, a lysosome-dependent degradation pathway, plays a fundamental role in maintaining cellular homeostasis by degrading damaged organelles and misfolded proteins. In the context of tumor formation, autophagy significantly influences cancer metabolism and chemotherapy resistance, contributing to both tumor suppression and surveillance. This review focuses on the relationship between mitochondria and autophagy, specifically in the context of cancer progression. Investigating the interaction between autophagy and mitochondria reveals new possibilities for cancer treatments and may result in the development of more effective therapies targeting mitochondria, which could have significant implications for cancer treatment. Additionally, this review highlights the increasing understanding of autophagy's role in tumor development, with a focus on modulating mitochondrial function and autophagy in both pre-clinical and clinical cancer research. It also explores the potential for developing more-targeted and personalized therapies by investigating autophagy-related biomarkers.
Collapse
Affiliation(s)
- Aleksandra Zdanowicz
- Department of Biochemistry, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Zwirki i Wigury 81 Str., 02-091 Warsaw, Poland
| | - Emilia Grosicka-Maciąg
- Department of Biochemistry and Laboratory Diagnostic, Collegium Medicum Cardinal Stefan Wyszyński University, Kazimierza Wóycickiego 1 Str., 01-938 Warsaw, Poland
| |
Collapse
|
7
|
Liu D, Qin H, Gao Y, Sun M, Wang M. Cardiovascular disease: Mitochondrial dynamics and mitophagy crosstalk mechanisms with novel programmed cell death and macrophage polarisation. Pharmacol Res 2024; 206:107258. [PMID: 38909638 DOI: 10.1016/j.phrs.2024.107258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024]
Abstract
Several cardiovascular illnesses are associated with aberrant activation of cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis, and macrophage polarisation as hallmarks contributing to vascular damage and abnormal cardiac function. Meanwhile, these three novel forms of cellular dysfunction are closely related to mitochondrial homeostasis. Mitochondria are the main organelles that supply energy and maintain cellular homeostasis. Mitochondrial stability is maintained through a series of regulatory pathways, such as mitochondrial fission, mitochondrial fusion and mitophagy. Studies have shown that mitochondrial dysfunction (e.g., impaired mitochondrial dynamics and mitophagy) promotes ROS production, leading to oxidative stress, which induces cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage M1 phenotypic polarisation. Therefore, an in-depth knowledge of the dynamic regulation of mitochondria during cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage polarisation is necessary to understand cardiovascular disease development. This paper systematically summarises the impact of changes in mitochondrial dynamics and mitophagy on regulating novel cellular dysfunctions and macrophage polarisation to promote an in-depth understanding of the pathogenesis of cardiovascular diseases and provide corresponding theoretical references for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Dandan Liu
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Hewei Qin
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450002, China.
| | - Yang Gao
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengyan Sun
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengnan Wang
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
8
|
Han Q, Yu Y, Liu X, Guo Y, Shi J, Xue Y, Li Y. The Role of Endothelial Cell Mitophagy in Age-Related Cardiovascular Diseases. Aging Dis 2024:AD.2024.0788. [PMID: 39122456 DOI: 10.14336/ad.2024.0788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Aging is a major risk factor for cardiovascular diseases (CVD), and mitochondrial autophagy impairment is considered a significant physiological change associated with aging. Endothelial cells play a crucial role in maintaining vascular homeostasis and function, participating in various physiological processes such as regulating vascular tone, coagulation, angiogenesis, and inflammatory responses. As aging progresses, mitochondrial autophagy impairment in endothelial cells worsens, leading to the development of numerous cardiovascular diseases. Therefore, regulating mitochondrial autophagy in endothelial cells is vital for preventing and treating age-related cardiovascular diseases. However, there is currently a lack of systematic reviews in this area. To address this gap, we have written this review to provide new research and therapeutic strategies for managing aging and age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Quancheng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghong Guo
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Liu N, Liang Y, Wei T, Huang X, Zhang T, Tang M. ROS-mediated NRF2/p-ERK1/2 signaling-involved mitophagy contributes to macrophages activation induced by CdTe quantum dots. Toxicology 2024; 505:153825. [PMID: 38710382 DOI: 10.1016/j.tox.2024.153825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/08/2024]
Abstract
Cadmium telluride (CdTe) quantum dots (QDs) have garnered significant attention for tumor imaging due to their exceptional properties. However, there remains a need for further investigation into their potential toxicity mechanisms and corresponding enhancements. Herein, CdTe QDs were observed to accumulate in mouse liver, leading to a remarkable overproduction of IL-1β and IL-6. Additionally, there was evidence of macrophage infiltration and activation following exposure to 12.5 μmol/kg body weight of QDs. To elucidate the underlying mechanism of macrophage activation, CdTe QDs functionalized with 3-mercaptopropionic acid (MPA) were utilized. In vitro experiments revealed that 1.0 μM MPA-CdTe QDs activated PINK1-dependent mitophagy in RAW264.7 macrophages. Critically, the autophagic flux remained unimpeded, as demonstrated by the absence of p62 accumulation, LC3 turnover assay results, and successful fusion of autophagosomes with lysosomes. Mechanically, QDs increased reactive oxygen species (ROS) and mitoROS by damaging both mitochondria and lysosomes. ROS, in turn, inhibited NRF2, resulting in the phosphorylation of ERK1/2 and subsequent activation of mitophagy. Notably, 1.0 μM QDs disrupted lysosomes but autophagic flux was not impaired. Eventually, the involvement of the ROS-NRF2-ERK1/2 pathway-mediated mitophagy in the increase of IL-1β and IL-6 in macrophages was confirmed using Trolox, MitoTEMPO, ML385, specific siRNAs, and lentivirus-based interventions. This study innovatively revealed the pro-inflammatory rather than anti-inflammatory role of mitophagy in nanotoxicology, shedding new light on the mechanisms of mitochondrial disorders induced by QDs and identifying several molecular targets to comprehend the toxicological mechanisms of CdTe QDs.
Collapse
Affiliation(s)
- Na Liu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - Ying Liang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Tingting Wei
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xiaoquan Huang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
10
|
Meng Y, Zhang J, Liu Y, Zhu Y, Lv H, Xia F, Guo Q, Shi Q, Qiu C, Wang J. The biomedical application of inorganic metal nanoparticles in aging and aging-associated diseases. J Adv Res 2024:S2090-1232(24)00213-3. [PMID: 38821357 DOI: 10.1016/j.jare.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Aging and aging-associated diseases (AAD), including neurodegenerative disease, cancer, cardiovascular diseases, and diabetes, are inevitable process. With the gradual improvement of life style, life expectancy is gradually extended. However, the extended lifespan has not reduced the incidence of disease, and most elderly people are in ill-health state in their later years. Hence, understanding aging and AAD are significant for reducing the burden of the elderly. Inorganic metal nanoparticles (IMNPs) predominantly include gold, silver, iron, zinc, titanium, thallium, platinum, cerium, copper NPs, which has been widely used to prevent and treat aging and AAD due to their superior properties (essential metal ions for human body, easily synthesis and modification, magnetism). Therefore, a systematic review of common morphological alternations of senescent cells, altered genes and signal pathways in aging and AAD, and biomedical applications of IMNPs in aging and AAD is crucial for the further research and development of IMNPs in aging and AAD. This review focus on the existing research on cellular senescence, aging and AAD, as well as the applications of IMNPs in aging and AAD in the past decade. This review aims to provide cutting-edge knowledge involved with aging and AAD, the application of IMNPs in aging and AAD to promote the biomedical application of IMNPs in aging and AAD.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haining Lv
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qianli Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jigang Wang
- Department of Urology, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
11
|
Huang M, Zhang W, Yang Y, Shao W, Wang J, Cao W, Zhu Z, Yang F, Zheng H. From homeostasis to defense: Exploring the role of selective autophagy in innate immunity and viral infections. Clin Immunol 2024; 262:110169. [PMID: 38479440 DOI: 10.1016/j.clim.2024.110169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
The process of autophagy, a conservative evolutionary mechanism, is responsible for the removal of surplus and undesirable cytoplasmic components, thereby ensuring cellular homeostasis. Autophagy exhibits a remarkable level of selectivity by employing a multitude of cargo receptors that possess the ability to bind both ubiquitinated cargoes and autophagosomes. In the context of viral infections, selective autophagy plays a crucial role in regulating the innate immune system. Notably, numerous viruses have developed strategies to counteract, evade, or exploit the antiviral effects of selective autophagy. This review encompasses the latest research progress of selective autophagy in regulating innate immunity and virus infectious.
Collapse
Affiliation(s)
- Mengyao Huang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Wei Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| | - Yang Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Wenhua Shao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Jiali Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Weijun Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Zixiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Fan Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| |
Collapse
|
12
|
Zhang R, Yang H, Guo M, Niu S, Xue Y. Mitophagy and its regulatory mechanisms in the biological effects of nanomaterials. J Appl Toxicol 2024. [PMID: 38642013 DOI: 10.1002/jat.4609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/22/2024]
Abstract
Mitophagy is a selective cellular process critical for the removal of damaged mitochondria. It is essential in regulating mitochondrial number, ensuring mitochondrial functionality, and maintaining cellular equilibrium, ultimately influencing cell destiny. Numerous pathologies, such as neurodegenerative diseases, cardiovascular disorders, cancers, and various other conditions, are associated with mitochondrial dysfunctions. Thus, a detailed exploration of the regulatory mechanisms of mitophagy is pivotal for enhancing our understanding and for the discovery of novel preventive and therapeutic options for these diseases. Nanomaterials have become integral in biomedicine and various other sectors, offering advanced solutions for medical uses including biological imaging, drug delivery, and disease diagnostics and therapy. Mitophagy is vital in managing the cellular effects elicited by nanomaterials. This review provides a comprehensive analysis of the molecular mechanisms underpinning mitophagy, underscoring its significant influence on the biological responses of cells to nanomaterials. Nanoparticles can initiate mitophagy via various pathways, among which the PINK1-Parkin pathway is critical for cellular defense against nanomaterial-induced damage by promoting mitophagy. The role of mitophagy in biological effects was induced by nanomaterials, which are associated with alterations in Ca2+ levels, the production of reactive oxygen species, endoplasmic reticulum stress, and lysosomal damage.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Haitao Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Gwon JG, Lee SM. Role of PTEN-Induced Protein Kinase 1 as a Mitochondrial Dysfunction Regulator in Cardiovascular Disease Pathogenesis. Vasc Specialist Int 2024; 40:9. [PMID: 38486493 PMCID: PMC10940882 DOI: 10.5758/vsi.230116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
Cardiovascular disease (CVD) remains a global health challenge, primarily due to atherosclerosis, which leads to conditions such as coronary artery disease, cerebrovascular disease, and peripheral arterial disease. Mitochondrial dysfunction initiates endothelial dysfunction, a key contributor to CVD pathogenesis, as well as triggers the accumulation of reactive oxygen species (ROS), energy stress, and cell death in endothelial cells, which are crucial for atherosclerosis development. This review explores the role of PTEN-induced protein kinase 1 (PINK1) in mitochondrial quality control, focusing on its significance in cardiovascular health. PINK1 plays a pivotal role in mitophagy (selective removal of damaged mitochondria), contributing to the prevention of CVD progression. PINK1-mediated mitophagy also affects the maintenance of cardiomyocyte homeostasis in ischemic heart disease, thus mitigating mitochondrial dysfunction and oxidative stress, as well as regulates endothelial health in atherosclerosis through influencing ROS levels and inflammatory response. We also investigated the role of PINK1 in vascular smooth muscle cells, emphasizing on its role in apoptosis and atherosclerosis. Dysfunctional mitophagy in these cells accelerates cellular senescence and contributes to adverse effects including plaque rupture and inflammation. Mitophagy has also been explored as a potential therapeutic target for vascular calcification, a representative lesion in atherosclerosis, with a focus on lactate-induced mechanisms. Finally, we highlight the current research and clinical trials targeting mitophagy as a therapeutic avenue for CVD.
Collapse
Affiliation(s)
- Jun Gyo Gwon
- Division of Vascular Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Ying X, Zheng X, Zhang X, Yin Y, Wang X. Kynurenine in IDO1 high cancer cell-derived extracellular vesicles promotes angiogenesis by inducing endothelial mitophagy in ovarian cancer. J Transl Med 2024; 22:267. [PMID: 38468343 PMCID: PMC10929174 DOI: 10.1186/s12967-024-05054-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Mitophagy, a prominent cellular homeostasis process, has been implicated in modulating endothelial cell function. Emerging evidence suggests that extracellular vesicles (EVs) participate in intercellular communication, which could modulate tumor angiogenesis, a hallmark of ovarian cancer (OC) progression. However, the underlying mechanisms through how EVs regulate endothelial mitophagy associated with tumor angiogenesis during OC development remain obscure. METHODS The effect of cancer cell-derived EVs on endothelial mitophagy and its correlation with tumor angiogenesis and OC development were explored by in vitro and in vivo experiments. Multi-omics integration analysis was employed to identify potential regulatory mechanisms of cancer cell-derived EVs on endothelial mitophagy, which is involved in tumor angiogenesis associated with OC development. These insights were then further corroborated through additional experiments. An orthotopic OC mouse model was constructed to assess the antiangiogenic and therapeutic potential of the Indoleamine 2,3 dioxygenase-1 (IDO1) inhibitor. RESULTS Cancer cell-derived EVs promoted tumor angiogenesis via the activation of endothelial mitophagy, contributing to the growth and metastasis of OC. The aberrantly high expression of IDO1 mediated abnormal tryptophan metabolism in cancer cells and promoted the secretion of L-kynurenine (L-kyn)-enriched EVs, with associated high levels of L-kyn in EVs isolated from both the tumor tissues and patient plasma in OC. EVs derived from IDO1high ovarian cancer cells elevated nicotinamide adenine dinucleotide (NAD +) levels in endothelial cells via delivering L-kyn. Besides, IDO1high ovarian cancer cell-derived EVs upregulated sirt3 expression in endothelial cells by increasing acetylation modification. These findings are crucial for promoting endothelial mitophagy correlated with tumor angiogenesis. Notably, both endothelial mitophagy and tumor angiogenesis could be suppressed by the IDO1 inhibitor in the orthotopic OC mouse model. CONCLUSIONS Together, our findings unveil a mechanism of mitophagy in OC angiogenesis and indicate the clinical relevance of EV enriched L-kyn as a potential biomarker for tumorigenesis and progression. Additionally, IDO1 inhibitors might become an alternative option for OC adjuvant therapy.
Collapse
Affiliation(s)
- Xiang Ying
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaoqian Zhang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
15
|
Tian T, Pang H, Li X, Ma K, Liu T, Li J, Luo Z, Li M, Hou Q, Hao H, Dong J, Du H, Liu X, Sun Z, Zhao C, Song X, Jin M. The role of DRP1 mediated mitophagy in HT22 cells apoptosis induced by silica nanoparticles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116050. [PMID: 38325272 DOI: 10.1016/j.ecoenv.2024.116050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/19/2024] [Accepted: 01/27/2024] [Indexed: 02/09/2024]
Abstract
Silica nanoparticles (SiNPs) are widely used in the biomedical field and can enter the central nervous system through the blood-brain barrier, causing damage to hippocampal neurons. However, the specific mechanism remains unclear. In this experiment, HT22 cells were selected as the experimental model in vitro, and the survival rate of cells under the action of SiNPs was detected by MTT method, reactive oxygen species (ROS), lactate dehydrogenase (LDH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px) and adenosine triphosphate (ATP) were tested by the kit, the ultrastructure of the cells was observed by transmission electron microscope, membrane potential (MMP), calcium ion (Ca2+) and apoptosis rate were measured by flow cytometry, and the expressions of mitochondrial functional protein, mitochondrial dynein, mitochondrial autophagy protein as well as apoptosis related protein were detected by Western blot. The results showed that cell survival rate, SOD, CAT, GSH-Px, ATP and MMP gradually decreased with the increase of SiNPs concentration, while intracellular ROS, Ca2+, LDH and apoptosis rate increased with the increase of SiNPs concentration. In total cellular proteins,the expressions of mitochondrial functional proteins VDAC and UCP2 gradually increased, the expression of mitochondrial dynamic related protein DRP1 increased while the expressions of OPA1 and Mfn2 decreased. The expressions of mitophagy related proteins PINK1, Parkin and LC3Ⅱ/LC3Ⅰ increased and P62 gradually decreased, as well as the expressions of apoptosis related proteins Apaf-1, Cleaved-Caspase-3, Caspase-3, Caspase-9, Bax and Cyt-C. In mitochondrial proteins, the expressions of mitochondrial dynamic related proteins DRP1 and p-DRP1 were increased, while the expressions of OPA1 and Mfn2 were decreased. Expressions of mitochondrial autophagy associated proteins PINK1, Parkin, LC3II/LC3I increased, P62 decreased gradually, as well as the expressions of apoptosis related proteins Cleaved-Caspase-3, Caspase-3, and Caspase-9 increased, and Cyt-C expressions decreased. To further demonstrate the role of ROS and DRP1 in HT22 cell apoptosis induced by SiNPs, we selected the ROS inhibitor N-Acetylcysteine (NAC) and Dynamin-related protein 1 (DRP1) inhibitor Mdivi-1. The experimental results indicated that the above effects were remarkably improved after the use of inhibitors, further confirming that SiNPs induce the production of ROS in cells, activate DRP1, cause excessive mitochondrial division, induce mitophagy, destroy mitochondrial function and eventually lead to apoptosis.
Collapse
Affiliation(s)
- Tiantian Tian
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Huan Pang
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Xinyue Li
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Kai Ma
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Tianxiang Liu
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Jiali Li
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Zhixuan Luo
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Meng Li
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Qiaohong Hou
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Huifang Hao
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Jianfei Dong
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Haiying Du
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Xiaomei Liu
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China
| | - Zhiwei Sun
- School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Chao Zhao
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China.
| | - Xiuling Song
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China.
| | - Minghua Jin
- School of Public Health Jilin University, Changchun, Jilin 130021, PR China.
| |
Collapse
|
16
|
Yao Y, Zhang T, Tang M. Toxicity mechanism of engineered nanomaterials: Focus on mitochondria. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 343:123231. [PMID: 38154775 DOI: 10.1016/j.envpol.2023.123231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
With the rapid development of nanotechnology, engineered nanomaterials (ENMs) are widely used in various fields. This has exacerbated the environmental pollution and human exposure of ENMs. The study of toxicity of ENMs and its mechanism has become a hot research topic in recent years. Mitochondrial damage plays an important role in the toxicity of ENMs. This paper reviews the structural damage, dysfunction, and molecular level perturbations caused by different ENMs to mitochondria, including ZnO NPs, Ag NPs, TiO2 NPs, iron oxide NPs, cadmium-based quantum dots, CuO NPs, silica NPs, carbon-based nanomaterials. Among them, mitochondrial quality control plays an important role in mitochondrial damage. We further summarize the cellular level outcomes caused by mitochondrial damage, mainly including, apoptosis, ferroptosis, pyroptosis and inflammation response. In addition, we concluded that reducing mitochondrial damage at source as well as accelerating recovery from mitochondrial damage through ENMs modification and pharmacological intervention are two feasible strategies. This review further provides new insights into the mitochondrial toxicity mechanisms of ENMs and provides a new foothold for predicting human health and environmental risks of ENMs.
Collapse
Affiliation(s)
- Yongshuai Yao
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, PR China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, PR China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
17
|
Zhang Z, Miao G, Lu L, Yin H, Wang Y, Wang B, Pan R, Zheng C, Jin X. Crucial physicochemical factors mediating mitochondrial toxicity of nanoparticles at noncytotoxic concentration. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168211. [PMID: 37918742 DOI: 10.1016/j.scitotenv.2023.168211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Nanomaterials have been extensively applied in multiple industries, among which silver nanoparticles (AgNPs), silicon dioxide nanoparticles (SiNPs), and gold nanoparticles (AuNPs) have become representative of widely consumed NPs. Limited knowledge is available regarding the subcellular responses of NPs with different physicochemical properties, i.e. material type and size, under the noncytotoxic concentrations. Macrophages are important sensitive cells exposed to NPs, and mitochondria are sensitive organelles that respond at the subcellular level. Herein, we found that sublethal concentrations of AgNPs and SiNPs, not AuNPs, decreased the mitochondrial membrane potential (MMP) and tubular mitochondria, and further resulted in an increase of ROS level and a decrease of ATP generation. AgNPs and SiNPs can also disturb mitochondrial dynamics manifested as increasing Mfn2 expression and decreasing Drp1 expression. Further assessments for mitochondrial function showed that AgNPs and SiNPs exposure led to a decrease in the gene expressions related to complex I (Ndufa8 and Ndufs2), complex III (Uqcrc2 and Uqcrfs1), complex IV (Cox6b1), and activity of complex I, suggesting their potential roles in impairing cellular respiration. In terms of the effects of NPs with different sizes, stronger toxicity was observed in smaller-sized nanoparticles. Among the above mitochondrial changes, we identified that ROS, ATP, MMP, tubular mitochondria, and expression of Drp1 were relatively sensitive indicators in subcellular response to NPs. With the above sensitive indicators, the comparison of heterogeneity between material type and size of the NPs showed that material type occupied a main influence on subcellular mitochondrial effects. Our finding provided important data on the potential subcellular risks of NPs, and indicated the vital role of material type for a better understanding of the nanomaterial biological safety.
Collapse
Affiliation(s)
- Ze Zhang
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Gan Miao
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Lin Lu
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Hao Yin
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Yingzhu Wang
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Baoqiang Wang
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Ruonan Pan
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Chuer Zheng
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Xiaoting Jin
- School of Public Health, Qingdao University, Qingdao 266071, PR China.
| |
Collapse
|
18
|
Mavil-Guerrero E, Vazquez-Duhalt R, Juarez-Moreno K. Exploring the cytotoxicity mechanisms of copper ions and copper oxide nanoparticles in cells from the excretory system. CHEMOSPHERE 2024; 347:140713. [PMID: 37981015 DOI: 10.1016/j.chemosphere.2023.140713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
Copper oxide nanoparticles (CuO NPs) are widely applied in various products, including food, cosmetic, biomedical, and environmental goods. Despite their broad use, potential risks are still associated with these NPs, therefore, the aim of this study is to delve deeper into the cytotoxic effects of 85 nm CuO NPs on kidney MDCK and liver AML-12 cells, representing cell models from the excretory system. Our findings pointed out that the viability of both cell lines decreased in a concentration-dependent manner when exposed to CuO NPs. Additionally, CuO NPs induced the overproduction of reactive oxygen species (ROS) and caused depolarization of the mitochondrial membrane, thereby arresting the cell cycle at the G2/M phase in MDCK and AML-12 cells. Importantly, unlike others our study uncovered distinctive forms of cellular death induced by CuO NPs in these cell lines. MDCK cells exhibited a combination of apoptosis and autophagy while early apoptosis was predominant in AML-12 cells. Moreover, the role of Cu2+ ions and CuO NPs in exerting cytotoxic effects was investigated, revealing that MDCK cells were affected by both copper ions and NPs. In contrast, AML-12 cells experienced toxic effects solely from CuO NPs. These findings provide crucial insights into the different cell death mechanisms caused either by CuO NPs or Cu2+ ions in excretory system cells in vitro. Nevertheless, further research is needed to explore the underlying mechanisms at the in vivo level, ensuring the safe use of CuO NPs. The results suggest that specific concentrations of metal oxide NPs can impact the physiology of cells within the excretory system of various mammals, including humans, and pave the way for comparing the toxic effects between ions and nanoparticles for further nanotoxicological studies.
Collapse
Affiliation(s)
- Elizabeth Mavil-Guerrero
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, (CFATA-UNAM), Blvd. Juriquilla #3001, Querétaro 76230, Mexico; Posgrado en Nanociencias, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada B.C. 22860, Mexico
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología (CNyN), Universidad Nacional Autónoma de México (UNAM), Ensenada B.C. 22860, Mexico
| | - Karla Juarez-Moreno
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, (CFATA-UNAM), Blvd. Juriquilla #3001, Querétaro 76230, Mexico.
| |
Collapse
|
19
|
Kanithi M, Kumari L, Yalakaturi K, Munjal K, Jimitreddy S, Kandamuri M, Veeramachineni P, Chopra H, Junapudi S. Nanoparticle Polymers Influence on Cardiac Health: Good or Bad for Cardiac Physiology? Curr Probl Cardiol 2024; 49:102145. [PMID: 37852559 DOI: 10.1016/j.cpcardiol.2023.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Cardiovascular diseases (CVD) are one of the leading causes of death and morbidity worldwide. Lifestyle modifications, medications, and addressing epidemiological factors have long been at the forefront of targeting therapeutics for CVD. Treatments can be further complicated given the intersection of gender, age, unique comorbidities, and healthcare access, among many other factors. Therefore, expanding treatment and diagnostic modalities for CVD is absolutely necessary. Nanoparticles and nanomaterials are increasingly being used as therapeutic and diagnostic modalities in various disciplines of biomedicine. Nanoparticles have multiple ways of interacting with the cardiovascular system. Some of them alter cardiac physiology by impacting ion channels, whereas others influence ions directly or indirectly, improving cellular death via decreasing oxidative stress. While embedding nanoparticles into therapeutics can help enhance healthy cardiovascular function in other scenarios, they can also impair physiology by increasing reactive oxidative species and leading to cardiotoxicity. This review explores different types of nanoparticles, their effects, and the applicable dosages to create a better foundation for understanding the current research findings.
Collapse
Affiliation(s)
- Manasa Kanithi
- Michigan State University College of Osteopathic Medicine, East Lansing, MI
| | - Lata Kumari
- People University of Medical and Health Sciences, Nawab Shah, Sindh, Pakistan
| | | | - Kavita Munjal
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | | | | | | | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sunil Junapudi
- Geethanjali College of Pharmacy, Hyderabad, Telangana, India.
| |
Collapse
|
20
|
Wang M, Zheng L, Ma S, Lin R, Li J, Yang S. Cuproptosis: emerging biomarkers and potential therapeutics in cancers. Front Oncol 2023; 13:1288504. [PMID: 38023234 PMCID: PMC10662309 DOI: 10.3389/fonc.2023.1288504] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
The sustenance of human life activities depends on copper, which also serves as a crucial factor for vital enzymes. Under typical circumstances, active homeostatic mechanisms keep the intracellular copper ion concentration low. Excess copper ions cause excessive cellular respiration, which causes cytotoxicity and cell death as levels steadily rise above a threshold. It is a novel cell death that depends on mitochondrial respiration, copper ions, and regulation. Cuproptosis is now understood to play a role in several pathogenic processes, including inflammation, oxidative stress, and apoptosis. Copper death is a type of regulatory cell death(RCD).Numerous diseases are correlated with the development of copper homeostasis imbalances. One of the most popular areas of study in the field of cancer is cuproptosis. It has been discovered that cancer angiogenesis, proliferation, growth, and metastasis are all correlated with accumulation of copper ions. Copper ion concentrations can serve as a crucial marker for cancer development. In order to serve as a reference for clinical research on the product, diagnosis, and treatment of cancer, this paper covers the function of copper ion homeostasis imbalance in malignant cancers and related molecular pathways.
Collapse
Affiliation(s)
- Min Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Shuai Ma
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Ruixin Lin
- Department of Hepato-Biliary-Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiahui Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Shuli Yang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Zhu S, Wu H, Cui H, Guo H, Ouyang Y, Ren Z, Deng Y, Geng Y, Ouyang P, Wu A, Deng J, Deng H. Induction of mitophagy via ROS-dependent pathway protects copper-induced hypothalamic nerve cell injury. Food Chem Toxicol 2023; 181:114097. [PMID: 37839787 DOI: 10.1016/j.fct.2023.114097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
Copper (Cu) is one of the essential trace elements in the body, but excessive amounts of Cu harm multiple organs and tissues such as liver, kidneys, testis, ovaries, and brain. However, the mechanism of hypothalamic neurotoxicity induced by Cu is still unknown. This study examined the relationship between reactive oxygen species (ROS) and mitophagy in mouse hypothalamus treated with high Cu. The results demonstrated that high levels of copper sulfate (CuSO4) could cause histopathological and neuronal changes in the mouse hypothalamus, produce a large amount of ROS, induce mitophagy, and lead to an imbalance of mitochondrial fusion/fission. The main manifestations are an increase in the expression levels of LC3-II/LC3-I, p62, DRP1, and FIS1, and a decrease in the expression levels of MFN1 and MFN2. Cu can induce mitophagy also was confirmed by LC3 co-localization with TOMM20 (mitochondrial marker). Next, the effect of oxidative stress on CuSO4-induced mitophagy was demonstrated. The results showed that ROS inhibitor N-acetylcysteine (NAC) diminished CuSO4-induced mitophagy and reversed the disturbance of mitochondrial dynamics. Additionally, a study was carried out to evaluate the role of mitophagy in CuSO4-induced hypothalamic injury. The inhibition of mitophagy using mitophagy inhibitor (Mdivi-1) decreased cell viability and promoted CuSO4-inhibited mitochondrial fusion. The aforementioned results suggested that CuSO4 induced mitophagy via oxidative stress in N38 cells and mouse hypothalamus, and that the activation of mitophagy might generate protective mechanisms by alleviating Cu-induced mitochondrial dynamics disorder. This study provided a novel approach and theoretical basis for studying and preventing Cu neurotoxicity.
Collapse
Affiliation(s)
- Song Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Hongbin Wu
- The Experimental Animal Center of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Agricultural Information Engineering of Sichuan Province, Sichuan Agriculture University, Yaan, Sichuan, 625014, China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Yujuan Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Youtian Deng
- College of Food Science, Sichuan Agriculture University, Yaan, Sichuan, 625014, China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Aimin Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China.
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China.
| |
Collapse
|
22
|
Zhang Y, Weng J, Huan L, Sheng S, Xu F. Mitophagy in atherosclerosis: from mechanism to therapy. Front Immunol 2023; 14:1165507. [PMID: 37261351 PMCID: PMC10228545 DOI: 10.3389/fimmu.2023.1165507] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
Mitophagy is a type of autophagy that can selectively eliminate damaged and depolarized mitochondria to maintain mitochondrial activity and cellular homeostasis. Several pathways have been found to participate in different steps of mitophagy. Mitophagy plays a significant role in the homeostasis and physiological function of vascular endothelial cells, vascular smooth muscle cells, and macrophages, and is involved in the development of atherosclerosis (AS). At present, many medications and natural chemicals have been shown to alter mitophagy and slow the progression of AS. This review serves as an introduction to the field of mitophagy for researchers interested in targeting this pathway as part of a potential AS management strategy.
Collapse
Affiliation(s)
- Yanhong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiajun Weng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| | - Luyao Huan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Song Sheng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
23
|
Song M, Zhang J, Huo S, Zhang X, Cui Y, Li Y. Mitophagy alleviates AIF-mediated spleen apoptosis induced by AlCl3 through Parkin stabilization in mice. Food Chem Toxicol 2023; 176:113762. [PMID: 37028746 DOI: 10.1016/j.fct.2023.113762] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
Aluminium (Al) accumulates in the spleen and causes spleen apoptosis. Mitochondrial dyshomeostasis represents primary mechanisms of spleen apoptosis induced by Al. Apoptosis-inducing factor (AIF) is located in the gap of the mitochondrial membrane and can be released into the nucleus, leading to apoptosis. Phosphatase and tensin homolog (PTEN)-induced putative kinase1 (PINK1)/E3 ubiquitin ligase PARK2 (Parkin)-mediated mitophagy maintains mitochondrial homeostasis by removing damaged mitochondria, but its function in AIF-mediated spleen apoptosis induced by Al is not clear. In our study, aluminium trichloride (AlCl3) was diluted in water for 90 d and administered to 75 male C57BL/6N mice at 0, 44.8, 59.8, 89.7, and 179.3 mg/kg body weight. AlCl3 triggered PINK1/Parkin pathway-mediated mitophagy, induced AIF release and AIF-mediated spleen apoptosis. AlCl3 was administered to sixty male C57BL/6N mice of wild type and Parkin knockout for 90 d at 0 and 179.3 mg/kg body weight. The results indicated that Parkin deficiency decreased mitophagy, aggravated mitochondrial damage, AIF release and AIF-mediated spleen apoptosis induced by AlCl3. According to our results, PINK1/Parkin-mediated mitophagy and AIF-mediated spleen apoptosis are caused by AlCl3, whereas mitophagy is protective in AIF-mediated apoptosis induced by AlCl3.
Collapse
|
24
|
Qu K, Yan F, Qin X, Zhang K, He W, Dong M, Wu G. Mitochondrial dysfunction in vascular endothelial cells and its role in atherosclerosis. Front Physiol 2022; 13:1084604. [PMID: 36605901 PMCID: PMC9807884 DOI: 10.3389/fphys.2022.1084604] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
The mitochondria are essential organelles that generate large amounts of ATP via the electron transport chain (ECT). Mitochondrial dysfunction causes reactive oxygen species accumulation, energy stress, and cell death. Endothelial mitochondrial dysfunction is an important factor causing abnormal function of the endothelium, which plays a central role during atherosclerosis development. Atherosclerosis-related risk factors, including high glucose levels, hypertension, ischemia, hypoxia, and diabetes, promote mitochondrial dysfunction in endothelial cells. This review summarizes the physiological and pathophysiological roles of endothelial mitochondria in endothelial function and atherosclerosis.
Collapse
Affiliation(s)
- Kai Qu
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Fang Yan
- Department of Geriatrics, Geriatric Diseases Institute of Chengdu, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China,Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Xian Qin
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Kun Zhang
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Wen He
- Department of Geriatrics, Clinical trial center, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China,*Correspondence: Mingqing Dong, ; Guicheng Wu,
| | - Guicheng Wu
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,*Correspondence: Mingqing Dong, ; Guicheng Wu,
| |
Collapse
|
25
|
Tracy EP, Nair R, Rowe G, Beare JE, Beyer A, LeBlanc AJ. Adipose stromal vascular fraction reverses mitochondrial dysfunction and hyperfission in aging-induced coronary microvascular disease. Am J Physiol Heart Circ Physiol 2022; 323:H749-H762. [PMID: 36018760 PMCID: PMC9529257 DOI: 10.1152/ajpheart.00311.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 01/28/2023]
Abstract
Aging is associated with blunted coronary microvascular vasodilatory function. Previously, systemically administered adipose stromal vascular fraction (SVF) therapy reversed aging-induced attenuation of β1-adrenergic- and flow-mediated dilation dependent on reducing mitochondrial reactive oxygen species. We hypothesized that SVF-mediated recovery of microvascular dilatory function is dependent on recovery of mitochondrial function, specifically by reducing mitochondrial hyperfission. Female Fischer-344 rats were allocated into young control, old control, and old + SVF therapy groups. Pressure myography, immunofluorescent staining, Western blot analysis, and RNA sequencing were performed to determine coronary microvascular mitochondrial dynamics and function. Gene and protein expression of fission-mediator DRP-1 was enhanced with aging but reversed by SVF therapy. SVF facilitated an increase in fusion-mediator MFN-1 gene and protein expression. Mitochondrial morphology was characterized as rod-like and densely networked in young controls, isolated circular and punctate with aging, and less circularity with partially restored mitochondrial branch density with SVF therapy. Decreased mitochondrial membrane potential and ATP bioavailability in aged animals at baseline and during flow-mediated dilation were reversed by SVF and accompanied with enhanced oxygen consumption. Dilation to norepinephrine and flow in young controls were dependent on uninhibited mitochondrial fusion, whereas inhibiting fission did not restore aged microvessel response to norepinephrine or flow. SVF-mediated recovery of β-adrenergic function was dependent on uninhibited mitochondrial fusion, whereas recovery of flow-mediated dilation was dependent on maintained mitochondrial fission. Impaired dilation in aging is mitigated by SVF therapy, which recovers mitochondrial function and fission/fusion balance.NEW & NOTEWORTHY We elucidated the consequences of aging on coronary microvascular mitochondrial health as well as SVF's ability to reverse these effects. Aging shifts gene/protein expression and mitochondrial morphology indicating hyperfission, alongside attenuated mitochondrial membrane potential and ATP bioavailability, all reversed using SVF therapy. Mitochondrial membrane potential and ATP levels correlated with vasodilatory efficiency. Mitochondrial dysfunction is a contributing pathological factor in aging that can be targeted by therapeutic SVF to preserve microvascular dilative function.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Rajeev Nair
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Jason E Beare
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky
| | - Andreas Beyer
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Amanda Jo LeBlanc
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky
| |
Collapse
|
26
|
Zhen Y, Ge L, Chen Q, Xu J, Duan Z, Loor JJ, Wang M. Latent Benefits and Toxicity Risks Transmission Chain of High Dietary Copper along the Livestock-Environment-Plant-Human Health Axis and Microbial Homeostasis: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6943-6962. [PMID: 35666880 DOI: 10.1021/acs.jafc.2c01367] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The extensive use of high-concentration copper (Cu) in feed additives, fertilizers, pesticides, and nanoparticles (NPs) inevitably causes significant pollution in the ecological environment. This type of chain pollution begins with animal husbandry: first, Cu accumulation in animals poisons them; second, high Cu enters the soil and water sources with the feces and urine to cause toxicity, which may further lead to crop and plant pollution; third, this process ultimately endangers human health through consumption of livestock products, aquatic foods, plants, and even drinking water. High Cu potentially alters the antibiotic resistance of soil and water sources and further aggravates human disease risks. Thus, it is necessary to formulate reasonable Cu emission regulations because the benefits of Cu for livestock and plants cannot be ignored. The present review evaluates the potential hazards and benefits of high Cu in livestock, the environment, the plant industry, and human health. We also discuss aspects related to bacterial and fungal resistance and homeostasis and perspectives on the application of Cu-NPs and microbial high-Cu removal technology to reduce the spread of toxicity risks to humans.
Collapse
Affiliation(s)
- Yongkang Zhen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, Xinjiang 832000, China
| | - Ling Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Qiaoqing Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jun Xu
- Institute for Quality and Safety and Standards of Agricultural Products Research, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330000, China
| | - Zhenyu Duan
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, Xinjiang 832000, China
| | - Juan J Loor
- Mammalian Nutrition Physiology Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Mengzhi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, Xinjiang 832000, China
| |
Collapse
|