1
|
Gao L, Moreno-Domínguez A, Ortega-Sáenz P, López-Barneo J. The carotid body oxygen sensor. Curr Opin Neurobiol 2025; 92:103022. [PMID: 40267624 DOI: 10.1016/j.conb.2025.103022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025]
Abstract
Carotid body (CB) chemoreceptor glomus cells sense hypoxia through the inhibition of plasmalemmal K+ channels, which leads to the opening of Ca2+ channels, Ca2+ influx, and neurotransmitter release. The mechanism of O2 sensing and the regulation of membrane ion channels by O2 have remained undefined and a subject of debate. Here, we summarize the molecular pathway that underlies acute O2 sensing in the CB. This process does not rely on a single-molecule O2 sensor expressed in glomus cells but rather on HIF2α-dependent genetically specialized mitochondria that can detect changes in O2 tension, within physiological ranges, and generate biochemical signals that regulate membrane ion channels. The acute O2-sensing pathway in glomus cells could provide new targets for respiratory and cardiovascular pharmacology.
Collapse
Affiliation(s)
- Lin Gao
- Institute of Biomedicine of Seville (IBiS), University Hospital "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Department of Medical Physiology and Biophysics. School of Medicine, University of Seville, Seville, Spain; CIBERNED, Madrid, Spain
| | - Alejandro Moreno-Domínguez
- Institute of Biomedicine of Seville (IBiS), University Hospital "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Department of Medical Physiology and Biophysics. School of Medicine, University of Seville, Seville, Spain; CIBERNED, Madrid, Spain
| | - Patricia Ortega-Sáenz
- Institute of Biomedicine of Seville (IBiS), University Hospital "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Department of Medical Physiology and Biophysics. School of Medicine, University of Seville, Seville, Spain; CIBERNED, Madrid, Spain
| | - José López-Barneo
- Institute of Biomedicine of Seville (IBiS), University Hospital "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Department of Medical Physiology and Biophysics. School of Medicine, University of Seville, Seville, Spain; CIBERNED, Madrid, Spain.
| |
Collapse
|
2
|
Ma Z, Cen Y, Xun W, Mou C, Yu J, Hu Y, Liu C, Sun J, Bi R, Qiu Y, Ding M, Jin L. Exercise enhances cardiomyocyte mitochondrial homeostasis to alleviate left ventricular dysfunction in pressure overload induced remodelling. Sci Rep 2025; 15:11698. [PMID: 40188200 PMCID: PMC11972341 DOI: 10.1038/s41598-025-95637-z] [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: 05/23/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
This study aims to explore how exercise enhances mitochondrial regulation and mitigates pathological cardiac hypertrophy. Rat groups were assigned as the control group (CN, n = 8), sham group (sham, n = 8), model group (SC, n = 16) and exercise group (SE, n = 20). A bioinformatics analysis was conducted to uncover the underlying mechanisms.H9C2 cells were divided into: the Ang II 0 h group (CON), Ang II 48 h group (Ang II), Ang II 48 h + sh-control group (sh-GFP + Ang II), Ang II 48 h + sh-ndufb10 group (sh-ndufb10 + Ang II), Ang II 48 h + overexpressedndufb10 control group (Ad-GFP + Ang II) and Ang II 48 h + over-expressedndufb10group (Ad-ndufb10 + Ang II). Mitochondrial function was measured. mRNA and protein expression were assessed by qPCR or western blot analysis respectively. In the SC group, a significant increase was observed in cardiomyocyte diameter, cardiac function, autophagy, and apoptosis. After 8 weeks of swimming exercise, there was a substantial reduction in cardiomyocyte diameter, an improvement in cardiac function, a mitigation of mitochondrial fission and autophagy. Ndufb10 was markedly enriched in oxidative phosphorylation and downregulated in the SC group, while it was upregulated in the SE group. In the sh-ndufb10 group, mitochondrial fusion was suppressed; fission and autophagy were further facilitated; mitochondrial membrane potential, mPTP, and ROS levels increased; and TUNEL positive nuclei and apoptosis-related proteins showed significant upregulation. Overexpression of ndufb10 reversed pathological hypertrophy, mitochondrial autophagy, mitochondrial dysfunction, and cardiomyocyte apoptosis in vitro. Swimming exercise improves mitochondrial abnormalities and reduces cardiomyocyte hypertrophy through regulation of the ndufb10 in left ventricular hypertrophy.
Collapse
Affiliation(s)
- Zhichao Ma
- School of Physical Education, Wuhan Business University, Wuhan, 430056, China.
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
- Equine Science Research and Horse Doping Control Laboratory, Wuhan Business University, Wuhan, 430056, China.
| | - Yanling Cen
- School of Physical Education, Wuhan Business University, Wuhan, 430056, China
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Weiwei Xun
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Caiying Mou
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Junwen Yu
- Aquinas International Academy, Ontario, CA, 90623, USA
| | - Yarui Hu
- Chiko Sports Institute, Sichuan University of Science and Technology, Meishan, 620000, China
| | - Chen Liu
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Jun Sun
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Rui Bi
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Yanli Qiu
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Mingchao Ding
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Li Jin
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China.
| |
Collapse
|
3
|
Zhang J, Liu T, Wu H, Wei J, Qu Q. Target oxidative stress-induced disulfidptosis: novel therapeutic avenues in Parkinson's disease. Mol Brain 2025; 18:29. [PMID: 40186271 PMCID: PMC11971801 DOI: 10.1186/s13041-025-01200-2] [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: 12/02/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD), a globally prevalent neurodegenerative disorder, has been implicated with oxidative stress (OS) as a central pathomechanism. Excessive reactive oxygen species (ROS) trigger neuronal damage and may induce disulfidptosis-a novel cell death modality not yet characterized in PD pathogenesis. METHOD Integrated bioinformatics analyses were conducted using GEO datasets to identify PD-associated differentially expressed genes (DEGs). These datasets were subjected to: immune infiltration analysis, gene set enrichment analysis (GSEA), weighted gene co-expression network analysis (WGCNA), intersection analysis of oxidative stress-related genes (ORGs) and disulfidptosis-related genes (DRGs) for functional enrichment annotation. Following hub gene identification, diagnostic performance was validated using independent cohorts. LASSO regression was applied for feature selection, with subsequent experimental validation in MPTP-induced PD mouse models. Single-cell transcriptomic profiling and molecular docking studies were performed to map target gene expression and assess drug-target interactions. RESULT A total of 1615 PD DEGs and 200 WGCNA DEGs were obtained, and the intersection with ORGs and DRGs resulted in 202 DEORGs, 11 DEDRGs, and 5 DED-ORGs (NDUFS2, LRPPRC, NDUFS1, GLUD1, and MYH6). These genes are mainly associated with oxidative stress, the respiratory electron transport chain, the ATP metabolic process, oxidative phosphorylation, mitochondrial respiration, and the TCA cycle. 10 hub genes have good diagnostic value, including in the validation dataset (AUC ≥ 0.507). LASSO analysis of hub genes yielded a total of 6 target genes, ACO2, CYCS, HSPA9, SNCA, SDHA, and VDAC1. In the MPTP-induced PD mice model, the expression of ACO2, HSPA9, and SDHA was decreased while the expression of CYCS, SNCA, and VDAC1 was increased, and the expression of the 5 DED-ORGs was decreased. Additionally, it was discovered that N-Acetylcysteine (NAC) could inhibit the occurrence of disulfidptosis in the MPTP-induced PD model. Subsequently, the distribution of target genes with AUC > 0.7 in different cell types of the brain was analyzed. Finally, molecular docking was performed between the anti-PD drugs entering clinical phase IV and the target genes. LRPPRC has low binding energy and strong affinity with duloxetine and donepezil, with binding energies of -7.6 kcal/mol and - 8.7 kcal/mol, respectively. CONCLUSION This study elucidates the pathogenic role of OS-induced disulfidptosis in PD progression. By identifying novel diagnostic biomarkers (e.g., DED-ORGs) and therapeutic targets (e.g., LRPPRC), our findings provide a mechanistic framework for PD management and lay the groundwork for future therapeutic development.
Collapse
Affiliation(s)
- Junshi Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Neurology, Huaihe Hospital of Henan Universtiy, Kaifeng, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Haojie Wu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Jianshe Wei
- Department of Neurology, Huaihe Hospital of Henan Universtiy, Kaifeng, 475004, China.
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
4
|
Cai F, Dong J, Xie P, He H, Yao H, Guo J, Yan Z, Ma L, Chen T. Mn-Specific Recognition of Guanidine Drives Selective Inhibition of Complex I. J Med Chem 2025; 68:5641-5654. [PMID: 39977356 DOI: 10.1021/acs.jmedchem.4c02904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Developing structurally well-defined targeted drugs is an effective way to enhance the chemotherapy efficacy. Herein, a target mitochondrial complex I (complex I) inhibitor was developed for the key methylation site ARG-85 in the key subunit NDUFS2. Based on the unique :NH═C- group of guanidyl and the surrounding environment of ARG-85, the macrocyclic and bulky manganese porphyrin complex [MnIII(TTPPC2-)]+ was selected to insert into the gap of NDUFS2. Experimental and computational analyses revealed that the planar π system of the TTPPC2- ligand and the rotatable benzene ring stably bind between the :NH═C- group of ARG-85 and the manganese metal center, a medium-strong Lewis acid. The Mn-specific recognition of guanidine drives the selective inhibition of complex I activity. Further, MnIII(TTPPC2-)]+ was modified into targeted nanoformulation Mn NPs. In vitro and in vivo experiments confirmed the efficient and mechanism inhibition of complex I activity, offering a novel strategy for targeted drug development.
Collapse
Affiliation(s)
- Fei Cai
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Jinrong Dong
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Peng Xie
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Hanlong He
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Huiyi Yao
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Junxian Guo
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Zhibo Yan
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Li Ma
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Key Laboratory for Regenerative Medicine of Ministry of Education, Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, Jinan University, Guangzhou 510632, China
| |
Collapse
|
5
|
Jin Y, Liu Z, Yang Z, Fang L, Zhao FQ, Liu H. Effects of hypoxia stress on the milk synthesis in bovine mammary epithelial cells. J Anim Sci Biotechnol 2025; 16:37. [PMID: 40050971 PMCID: PMC11887346 DOI: 10.1186/s40104-025-01174-0] [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: 10/05/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Milk synthesis is an energy-intensive process influenced by oxygen availability. This study investigates how hypoxia affects milk synthesis in BMECs, focusing on key genes involved in lactation and energy metabolism. METHODS BMECs were cultured in a normoxic environment and then transferred to a hypoxia chamber with 1% O2 for specified durations. The study evaluated cellular responses through various molecular experiments and RNA sequencing. Small interfering RNA was employed to knock down HIF-1α to investigate whether the lactation-related phenotype alteration depends on HIF-1α. RESULTS Hypoxia disrupted milk protein production by reducing mTOR/P70S6K/4EBP1 signaling and downregulating genes critical for amino acid transport and protein synthesis. Triglyceride synthesis increased due to enhanced fatty acid uptake and the upregulation of regulatory proteins, including FASN and PPARγ. Although glucose uptake was elevated under hypoxia, key enzymes for lactose synthesis were downregulated, suggesting a redirection of glucose toward energy production. Mitochondrial function was impaired under hypoxia, with reduced gene expression in TCA cycle, ETC, cytosol-mitochondrial transport, decreased ATP levels, increased ROS levels, and structural alterations. Additionally, lipid synthesis and glucose uptake depend on HIF-1α, while milk protein synthesis alterations occurred independently of HIF-1α. CONCLUSIONS Hypoxia alters milk synthesis in BMECs by disrupting milk protein synthesis, enhancing lipid metabolism, and impairing energy production. These findings provide valuable insights into the molecular mechanisms underlying the effect of oxygen deprivation on lactation efficiency, offering potential targets for mitigating hypoxic stress in the mammary glands of dairy animals.
Collapse
Affiliation(s)
- Yanshan Jin
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhuolin Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ziyan Yang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lizhu Fang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng-Qi Zhao
- Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
6
|
Yin KF, Chen T, Gu XJ, Jiang Z, Su WM, Duan QQ, Wen XJ, Cao B, Li JR, Chi LY, Chen YP. Identification of Potential Causal Genes for Neurodegenerative Diseases by Mitochondria-Related Genome-Wide Mendelian Randomization. Mol Neurobiol 2025; 62:3892-3902. [PMID: 39347895 DOI: 10.1007/s12035-024-04528-3] [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: 10/29/2023] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Current research lacks comprehensive investigations into the potential causal link between mitochondrial-related genes and the risk of neurodegenerative diseases (NDDs). We aimed to identify potential causative genes for five NDDs through an examination of mitochondrial-related gene expression levels. Through the integration of summary statistics from expression quantitative trait loci (eQTL) datasets (human blood and brain tissue), mitochondrial DNA copy number (mtDNA-CN), and genome-wide association studies (GWAS) datasets of five NDDs from European ancestry, we conducted a Mendelian randomization (MR) analysis to explore the potential causal relationship between mitochondrial-related genes and Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Lewy body dementia (LBD). Sensitivity analysis and Bayesian colocalization were employed to validate this causal relationship. Through MR analysis, we have identified potential causal relationships between 12 mitochondria-related genes and AD, PD, ALS, and FTD overlapping with motor neuron disease (FTD_MND) in human blood or brain tissue. Bayesian colocalization analysis further confirms 9 causal genes, including NDUFS2, EARS2, and MRPL41 for AD; NDUFAF2, MALSU1, and METTL8 for PD; MYO19 and MRM1 for ALS; and FASTKD1 for FTD_MND. Importantly, in both human blood and brain tissue, NDUFS2 exhibits a significant pathogenic effect on AD, while NDUFAF2 demonstrates a robust protective effect on PD. Additionally, the mtDNA-CN plays a protected role in LBD (OR = 0.62, p = 0.031). This study presents evidence establishing a causal relationship between mitochondrial dysfunction and NDDs. Furthermore, the identified candidate genes may serve as potential targets for drug development aimed at preventing NDDs.
Collapse
Affiliation(s)
- Kang-Fu Yin
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ting Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiao-Jing Gu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiang-Jin Wen
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ju-Rong Li
- Department of Geriatrics, Dazhou Central Hospital, Dazhou, 635000, Sichuan, China
| | - Li-Yi Chi
- Department of Neurology, First Affiliated Hospital of Air Force Military Medical University, Xi'an, 710072, Shaanxi, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Lu Y, Kobayashi Y, Niki Y, Moriyama K. Possible role of superoxide dismutase 3 in hypoxia-induced developmental defects in murine molars. J Oral Biosci 2025; 67:100611. [PMID: 39746510 DOI: 10.1016/j.job.2024.100611] [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: 08/01/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVES To investigate the effects of hypoxia on tooth germ development in mice and explore the underlying mechanisms. METHODS Tooth germs were extracted from E14.5 mouse embryos and divided into the control and hypoxia groups for organ culture. The hypoxia group was exposed to hypoxia (0% oxygen) for 3 h, followed by normoxia for 21 h. After 2 or 7 days, samples were collected for morphometric analysis, reverse transcription-quantitative polymerase chain reaction, immunohistochemistry (IHC), and immunofluorescent staining (IF). Additionally, superoxide dismutase 3 (SOD3) expression patterns in mandibular molar tooth germs from C57BL/6 mouse embryos were analyzed using IHC. The SOD inhibitor sodium N, N-diethyldithiocarbamate trihydrate (DETC; 400 μM) was applied under normoxia for 3 days, followed by morphometry, IHC, and IF. RESULTS After 7 days, the hypoxia group exhibited significantly smaller tooth size, fewer cusps, reduced cell proliferation, and increased apoptosis in the epithelium compared to the control group. Sod3 mRNA expression was higher than other Sod family member expressions in the control group. In the hypoxia group, Sod3 mRNA and SOD3 protein expression were significantly decreased, whereas hypoxia-inducible factor-1 expression and reactive oxygen species levels were increased. SOD3 was primarily expressed in the dental epithelium from E12.5 to E17.5. DETC impaired tooth germ development in the control group, resulting in a phenotype similar to that of the hypoxia group, and significantly reduced amelogenin and msh homeobox 2 expression in the epithelium. CONCLUSIONS Hypoxia impairs tooth germ development. SOD3 probably plays a protective role during this process.
Collapse
Affiliation(s)
- Yeming Lu
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Yukiho Kobayashi
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan.
| | - Yuki Niki
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| |
Collapse
|
8
|
Zou X, Huang Q, Kang T, Shen S, Cao C, Wu J. An integrated investigation of mitochondrial genes in COPD reveals the causal effect of NDUFS2 by regulating pulmonary macrophages. Biol Direct 2025; 20:4. [PMID: 39789601 PMCID: PMC11715544 DOI: 10.1186/s13062-025-00593-3] [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: 09/03/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Despite the increasing body of evidence that mitochondrial activities implicate in chronic obstructive pulmonary disease (COPD), we are still far from a causal-logical and mechanistic understanding of the mitochondrial malfunctions in COPD pathogenesis. RESULTS Differential expression genes (DEGs) from six publicly available bulk human lung tissue transcriptomic datasets of COPD patients were intersected with the known mitochondria-related genes from MitoCarta3.0 to obtain mitochondria-related DEGs associated with COPD (MitoDEGs). The 32 hub MitoDEGs identified from protein-protein interaction (PPI) networks demonstrated superior overall diagnostic efficacy to non-hub MitoDEGs. Random forest (RF) analysis, least absolute shrinkage and selection operator (LASSO) regression, and Mendelian Randomization (MR) analysis of hub MitoDEGs further nominated NDUFS2, CAT, and MRPL2 as causal MitoDEGs for COPD, whose predominate expressions in pulmonary macrophages were revealed by an independent single-cell transcriptomic dataset of COPD human lungs. Finally, NDUFS2 was evaluated as the top-ranked contributor to COPD in the nomogram model and its downregulation in pulmonary macrophages could result in pro-inflammatory secretion, enhanced intercellular communications, whereas depressed phagocytosis of macrophages as revealed by gene set variation analysis (GSVA) and cell-cell interaction (CCI) analysis of single-cell transcriptomic dataset of COPD human lungs, which was later confirmed in COPD mouse model and macrophage cell lines. CONCLUSIONS Our study established the causal linkage between mitochondrial malfunctions and COPD, providing a potential therapeutic avenue to alleviate pulmonary inflammation accounting for COPD by targeting mitochondria-related genes. NDUFS2, a canonical component of mitochondrial electron respiratory chain, was highlighted instrumental for the susceptibility of risk-exposed individuals to COPD.
Collapse
Affiliation(s)
- Xiaoli Zou
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Qiqing Huang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Tutu Kang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Shaoran Shen
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Chenxi Cao
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jianqing Wu
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
9
|
Liao C, O’Farrell E, Qalieh Y, Savalia NK, Girgenti MJ, Kwan KY, Kwan AC. Single-nucleus transcriptomics reveals time-dependent and cell-type-specific effects of psilocybin on gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.04.631335. [PMID: 39803502 PMCID: PMC11722411 DOI: 10.1101/2025.01.04.631335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
There is growing interest to investigate classic psychedelics as potential therapeutics for mental illnesses. Previous studies have demonstrated that one dose of psilocybin leads to persisting neural and behavioral changes. The durability of psilocybin's effects suggests that there are likely alterations of gene expression at the transcriptional level. In this study, we performed single-nucleus RNA sequencing of the dorsal medial frontal cortex of male and female mice. Samples were collected at 1, 2, 4, 24, or 72 hours after psilocybin or ketamine administration and from control animals. At baseline, major excitatory and GABAergic cell types selectively express particular serotonin receptor transcripts. The psilocybin-evoked differentially expressed genes in excitatory neurons were involved in synaptic plasticity, which were distinct from genes enriched in GABAergic neurons that contribute to mitochondrial function and cellular metabolism. The effect of psilocybin on gene expression was time-dependent, including an early phase at 1-2 hours followed by a late phase at 72 hours of transcriptional response after administration. Ketamine administration produced transcriptional changes that show a high degree of correlation to those induced by psilocybin. Collectively, the results reveal that psilocybin produces time-dependent and cell-type specific changes in gene expression in the medial frontal cortex, which may underpin the drug's long-term effects on neural circuits and behavior.
Collapse
Affiliation(s)
- Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Ethan O’Farrell
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yaman Qalieh
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Neil K. Savalia
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
| | - Matthew J. Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, USA
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Kenneth Y. Kwan
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alex C. Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
10
|
Zhang F, Ye Z, Xie Y, Liu M, Zhang L, Zhang J, Xu Z. Levodopa-induced dyskinesia: brain iron deposition as a new hypothesis. Biometals 2024; 37:1307-1323. [PMID: 39212870 DOI: 10.1007/s10534-024-00628-8] [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: 07/08/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in the older adults. The main pathological change in PD is the degenerative death of dopamine (DA) neurons in the midbrain substantia nigra, which causes a significant decrease in the DA content of the striatum. However, the exact etiology of this pathological change remains unclear. Genetic factors, environmental factors, aging, and oxidative stress may be involved in the degenerative death of dopaminergic neurons in PD. Pharmacological treatment using levodopa (L-DOPA) remains the main treatment for PD. Most patients with PD consuming L-DOPA for a long time usually develop levodopa-induced dyskinesia (LID) after 6.5 years of use, and LID seriously affects the quality of life and increases the risk of disability. Recently, studies have revealed that cerebral iron deposition may be involved in LID development and that iron deposition has neurotoxic effects and accelerates disease onset. However, the relationship between cerebral iron deposition and LID remains unclear. Herein, we reviewed the mechanisms by which iron deposition may be associated with LID development, which are mainly related to oxidative stress, neuroinflammation, and mitochondrial and lysosomal dysfunction. Using iron as an important target, the search and development of safe and effective brain iron scavengers, and thus the alleviation and treatment of LID, has a very important scientific and clinical value, as well as a good application prospect.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Zhuofan Ye
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
- Department of Neurology, Guizhou Pronvincial People's Hospital, Guiyang, China
| | - Yuanyang Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
11
|
Archer SL, Dunham-Snary KJ, Bentley R, Alizadeh E, Weir EK. Hypoxic Pulmonary Vasoconstriction: An Important Component of the Homeostatic Oxygen Sensing System. Physiol Res 2024; 73:S493-S510. [PMID: 39589299 PMCID: PMC11627260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 11/27/2024] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) rapidly and reversibly matches lung ventilation (V) and perfusion (Q), optimizing oxygen uptake and systemic oxygen delivery. HPV occurs in small pulmonary arteries (PA), which uniquely constrict to hypoxia. Although HPV is modulated by the endothelium the core mechanism of HPV resides in PA smooth muscle cells (PASMC). The PASMC's mitochondrial oxygen sensor lies within the electron transport chain (ETC) and includes NDUFS2 in ETC Complex-I. PASMC mitochondria respond to hypoxia by varying production of reactive oxygen species (ROS) and hydrogen peroxide in proportion to alveolar oxygen tension. Hypoxic ROS inhibition results in a state of reduction which triggers a redox-mediated inhibition of oxygen-sensitive, voltage-gated, potassium channels, including Kv1.5 and Kv2.1. Kv channel inhibition depolarizes the PASMC, opening of large-conductance calcium channels (CaL), elevating cytosolic calcium and activating the contractile apparatus. HPV is strongest in small PAs where sensors (hypoxia-responsive mitochondria) and effectors (oxygen-sensitive K+ channels) are enriched. Oxygenation at birth reverses fetal HPV, contributing to the rapid neonatal drop in pulmonary vascular resistance (PVR). A similar mitochon-drial-K+ channel sensor-effector mechanism exists in the ductus arteriosus (DA), however in DASMC it is oxygen-induced increases in mitochondrial ROS that inhibit DASMC K+ channels, causing DA constriction. Atelectasis and pneumonia elicit HPV, which optimises V/Q matching, increasing systemic oxygenation. Whilst HPV in response to localized hypoxia in a single lung lobe does not increase PA pressure; global airway hypoxia, as occurs with altitude or sleep apnea, causes pulmonary hypertension. HPV can be inhibited by drugs, including calcium channel blockers, or used to maintain a dry operative field during single lung anesthesia for lung surgery. HPV does not normally cause lung edema but excessive, heterogenous HPV contributes to high altitude pulmonary edema. HPV is suppressed in COVID-19 pneumonia by a SARS-CoV-2 mitochondriopathy. HPV is a component of the body's homeostatic oxygen sensing system. Keywords: Ductus arteriosus, Redox, NDUFS2, Oxygen sensitive potassium, Channels, High altitude pulmonary edema (HAPE), Mitochondrial electron transport chain, COVID-19 pneumonia, Atelectasis.
Collapse
Affiliation(s)
- S L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| | | | | | | | | |
Collapse
|
12
|
Shi P, Sha Y, Wang X, Yang T, Wu J, Zhou J, Liu K, Guan X, Wang S, Liu Y, Gao J, Sun H, Ban T, Cao Y. Targeted Delivery and ROS-Responsive Release of Lutein Nanoassemblies Inhibit Myocardial Ischemia-Reperfusion Injury by Improving Mitochondrial Function. Int J Nanomedicine 2024; 19:11973-11996. [PMID: 39583319 PMCID: PMC11585303 DOI: 10.2147/ijn.s488532] [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: 09/11/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Purpose Myocardial ischemia-reperfusion injury (MI/RI) is associated with increased oxidative damage and mitochondrial dysfunction, resulting in an elevated risk of mortality. MI/RI may be alleviated by protecting cardiomyocytes from oxidative stress. Lutein, which belongs to a class of carotenoids, has proven to be effective in cardiovascular disease treatment due to its remarkable antioxidant properties, but its application is limited due to its poor stability and low bioavailability in vivo. Methods In this study, a delivery system was developed based on distearoyl phosphatidyl ethanolamine (DSPE)-thiol-ketone (TK)-PEG2K (polyethylene glycol 2000) (abbreviated as DTP) and PCM-SH (CWLSEAGPVVTVRALRGTGSW) to deliver lutein (abbreviated as lutein@DTPP) to damaged myocardium. First, lutein, lutein@DTP, or lutein@DTPP were injected through the tail vein once a day for 3 days and then MI/RI model rats were established by exposing rats to ischemia for 45 min and reperfusion for 6 h. We employed a range of experimental techniques including qRT-PCR, Western blotting, transmission electron microscopy, immunohistochemistry, immunofluorescence, flow cytometry, immunoprecipitation, molecular docking, and molecular dynamics simulations. Results Lutein@DTPP exhibited good myocardial targeting and ROS-responsive release. Our data suggested that lutein@DTPP effectively suppresses ferroptosis in cardiomyocytes. Mechanistically, we observed an upregulation of mouse double minute-2 (MDM2) in the hearts of MI/RI models and cardiomyocytes exposed to hypoxia/reoxygenation (H/R) conditions. In addition, NADH-ubiquinone oxidoreductase 75 kDa Fe-S protein 1 (NDUFS1) translocation from the cytosol to the mitochondria was inhibited by MDM2 upregulation. Notably, no significant variation in the total NDUFS1 expression was observed in H/R-exposed cardiomyocytes following treatment with siMDM2. Further study indicated that lutein facilitates the translocation of NDUFS1 from the cytosol to mitochondria by directly binding and sequestering MDM2, thereby improving mitochondrial function and inhibiting ferroptosis. Conclusion Lutein@DTPP promoted the mitochondrial translocation of NDUFS1 to restore mitochondrial function and inhibited the ferroptosis of cardiomyocytes by directly binding and sequestering MDM2.
Collapse
Affiliation(s)
- Pilong Shi
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yuetong Sha
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Xinran Wang
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Tao Yang
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jiawei Wu
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jiajun Zhou
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Kai Liu
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Xue Guan
- Morphological Experiment Center, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Song Wang
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yongsheng Liu
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jingquan Gao
- Department of Nursing, School of Medicine, Lishui University, Lishui, People’s Republic of China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Tao Ban
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| |
Collapse
|
13
|
Maya-López M, Monsalvo-Maraver LA, Delgado-Arzate AL, Olivera-Pérez CI, El-Hafidi M, Silva-Palacios A, Medina-Campos O, Pedraza-Chaverri J, Aschner M, Tinkov AA, Túnez I, Retana-Márquez S, Zazueta C, Santamaría A. Anandamide and WIN 55212-2 Afford Protection in Rat Brain Mitochondria in a Toxic Model Induced by 3-Nitropropionic Acid: an In Vitro Study. Mol Neurobiol 2024; 61:6435-6452. [PMID: 38307967 PMCID: PMC11338978 DOI: 10.1007/s12035-024-03967-2] [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: 07/13/2023] [Accepted: 01/16/2024] [Indexed: 02/04/2024]
Abstract
Mitochondrial dysfunction plays a key role in the development of neurodegenerative disorders. In contrast, the regulation of the endocannabinoid system has been shown to promote neuroprotection in different neurotoxic paradigms. The existence of an active form of the cannabinoid receptor 1 (CB1R) in mitochondrial membranes (mitCB1R), which might exert its effects through the same signaling mechanisms as the cell membrane CB1R, has been shown to regulate mitochondrial activity. Although there is evidence suggesting that some cannabinoids may induce protective effects on isolated mitochondria, substantial evidence on the role of cannabinoids in mitochondria remains to be explored. In this work, we developed a toxic model of mitochondrial dysfunction induced by exposure of brain mitochondria to the succinate dehydrogenase inhibitor 3-nitropropionic acid (3-NP). Mitochondria were also pre-incubated with the endogenous agonist anandamide (AEA) and the synthetic CB1R agonist WIN 55212-2 to evaluate their protective effects. Mitochondrial reduction capacity, reactive oxygen species (ROS) formation, and mitochondrial swelling were assessed as toxic markers. While 3-NP decreased the mitochondrial reduction capacity and augmented mitochondrial ROS formation and swelling, both AEA and WIN 55212-2 ameliorated these toxic effects. To explore the possible involvement of mitCB1R activation on the protective effects of AEA and WIN 55212-2, mitochondria were also pre-incubated in the presence of the selective CB1R antagonist AM281, which completely reverted the protective effects of the cannabinoids to levels similar to those evoked by 3-NP. These results show partial protective effects of cannabinoids, suggesting that mitCB1R activation may be involved in the recovery of compromised mitochondrial activity, related to reduction of ROS formation and further prevention of mitochondrial swelling.
Collapse
Affiliation(s)
- Marisol Maya-López
- Doctorado en Ciencias Biológicas y de La Salud, Universidad Autónoma Metropolitana, 09310, Mexico City, Mexico.
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| | | | | | | | - Mohammed El-Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, SSA, 14080, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, SSA, 14080, Mexico City, Mexico
| | - Omar Medina-Campos
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Autónoma de México, 04510, Mexico City, Mexico
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Autónoma de México, 04510, Mexico City, Mexico
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
- Department of Human Ecology and Bioelementology, and Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Isaac Túnez
- Instituto de Investigaciones Biomedicas Maimónides de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Córdoba, Spain
- Red Española de Excelencia en Estimulación Cerebral (REDESTIM), 14071, Córdoba, Spain
| | - Socorro Retana-Márquez
- Departamento de Biología de La Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, 09310, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, SSA, 14080, Mexico City, Mexico.
| | - Abel Santamaría
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
14
|
Alva R, Wiebe JE, Stuart JA. Revisiting reactive oxygen species production in hypoxia. Pflugers Arch 2024; 476:1423-1444. [PMID: 38955833 DOI: 10.1007/s00424-024-02986-1] [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: 05/02/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
Cellular responses to hypoxia are crucial in various physiological and pathophysiological contexts and have thus been extensively studied. This has led to a comprehensive understanding of the transcriptional response to hypoxia, which is regulated by hypoxia-inducible factors (HIFs). However, the detailed molecular mechanisms of HIF regulation in hypoxia remain incompletely understood. In particular, there is controversy surrounding the production of mitochondrial reactive oxygen species (ROS) in hypoxia and how this affects the stabilization and activity of HIFs. This review examines this controversy and attempts to shed light on its origin. We discuss the role of physioxia versus normoxia as baseline conditions that can affect the subsequent cellular response to hypoxia and highlight the paucity of data on pericellular oxygen levels in most experiments, leading to variable levels of hypoxia that might progress to anoxia over time. We analyze the different outcomes reported in isolated mitochondria, versus intact cells or whole organisms, and evaluate the reliability of various ROS-detecting tools. Finally, we examine the cell-type and context specificity of oxygen's various effects. We conclude that while recent evidence suggests that the effect of hypoxia on ROS production is highly dependent on the cell type and the duration of exposure, efforts should be made to conduct experiments under carefully controlled, physiological microenvironmental conditions in order to rule out potential artifacts and improve reproducibility in research.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| | - Jacob E Wiebe
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
15
|
Ricardo PC, Arias MC, de Souza Araujo N. Decoding bee cleptoparasitism through comparative transcriptomics of Coelioxoides waltheriae and its host Tetrapedia diversipes. Sci Rep 2024; 14:12361. [PMID: 38811580 PMCID: PMC11137135 DOI: 10.1038/s41598-024-56261-5] [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: 08/23/2023] [Accepted: 03/04/2024] [Indexed: 05/31/2024] Open
Abstract
Cleptoparasitism, also known as brood parasitism, is a widespread strategy among bee species in which the parasite lays eggs into the nests of the host species. Even though this behavior has significant ecological implications for the dynamics of several species, little is known about the molecular pathways associated with cleptoparasitism. To shed some light on this issue, we used gene expression data to perform a comparative analysis between two solitary neotropical bees: Coelioxoides waltheriae, an obligate parasite, and their specific host Tetrapedia diversipes. We found that ortholog genes involved in signal transduction, sensory perception, learning, and memory formation were differentially expressed between the cleptoparasite and the host. We hypothesize that these genes and their associated molecular pathways are engaged in cleptoparasitism-related processes and, hence, are appealing subjects for further investigation into functional and evolutionary aspects of cleptoparasitism in bees.
Collapse
Affiliation(s)
- Paulo Cseri Ricardo
- Departamento de Genética e Biologia Evolutiva - Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| | - Maria Cristina Arias
- Departamento de Genética e Biologia Evolutiva - Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
16
|
Vlachakis D, Tsilafakis K, Kostavasili I, Kossida S, Mavroidis M. Unraveling Desmin's Head Domain Structure and Function. Cells 2024; 13:603. [PMID: 38607042 PMCID: PMC11012097 DOI: 10.3390/cells13070603] [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: 01/09/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Understanding the structure and function of intermediate filaments (IFs) is necessary in order to explain why more than 70 related IF genes have evolved in vertebrates while maintaining such dramatically tissue-specific expression. Desmin is a member of the large multigene family of IF proteins and is specifically expressed in myocytes. In an effort to elucidate its muscle-specific behavior, we have used a yeast two-hybrid system in order to identify desmin's head binding partners. We described a mitochondrial and a lysosomal protein, NADH ubiquinone oxidoreductase core subunit S2 (NDUFS2), and saposin D, respectively, as direct desmin binding partners. In silico analysis indicated that both interactions at the atomic level occur in a very similar way, by the formation of a three-helix bundle with hydrophobic interactions in the interdomain space and hydrogen bonds at R16 and S32 of the desmin head domain. The interactions, confirmed also by GST pull-down assays, indicating the necessity of the desmin head domain and, furthermore, point out its role in function of mitochondria and lysosomes, organelles which are disrupted in myopathies due to desmin head domain mutations.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Biotechnology Department, Agricultural University of Athens, 11855 Athens, Greece;
| | - Konstantinos Tsilafakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
- Biochemistry & Biotechnology Department, University of Thessaly, 41500 Larisa, Greece
| | - Ioanna Kostavasili
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| | - Sophia Kossida
- IMGT, The International ImMunoGeneTics Information System, National Center for Scientific Research (CNRS), Institute of Human Genetics (IGH), University of Montpellier (UM), 34090 Montpellier, France;
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| |
Collapse
|
17
|
Wu Q, Yuan C, Wang J, Li G, Zhu C, Li L, Wang Z, Lv Q, Mei W. Uridine-Modified Ruthenium(II) Complex as Lysosomal LIMP-2 Targeting Photodynamic Therapy Photosensitizer for the Treatment of Triple-Negative Breast Cancer. JACS AU 2024; 4:1081-1096. [PMID: 38559730 PMCID: PMC10976599 DOI: 10.1021/jacsau.3c00808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 04/04/2024]
Abstract
Lysosome-targeted photodynamic therapy, which enhances reactive oxygen species (ROS)-responsive tumor cell death, has emerged as a promising strategy for cancer treatment. Herein, a uridine (dU)-modified Ru(II) complex (RdU) was synthesized by click chemistry. It was found that RdU exhibits impressive photo-induced inhibition against the growth of triple-negative breast cancer (TNBC) cells in normoxic and hypoxic microenvironments through ROS production. It was further revealed that RdU induces ferroptosis of MDA-MB-231 cells under light irradiation (650 nm, 300 mW/cm2). Additional experiments showed that RdU binds to lysosomal integral membrane protein 2 (LIMP-2), which was confirmed by the fact that RdU selectively localizes in the lysosomes of MDA-MB-231 cells and significantly augments the levels of LIMP-2. Molecular docking simulations and an isothermal titration calorimetry assay also showed that RdU has a high affinity to LIMP-2. Finally, in vivo studies in tumor-bearing (MDA-MB-231 cells) nude mice showed that RdU exerts promising photodynamic therapeutic effects on TNBC tumors. In summary, the uridine-modified Ru(II) complex has been developed as a potential LIMP-2 targeting agent for TNBC treatment through enhancing ROS production and promoting ferroptosis.
Collapse
Affiliation(s)
- Qiong Wu
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, Guangzhou 530316, China
| | - Chanling Yuan
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Jiacheng Wang
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Guohu Li
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Chunguang Zhu
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Li Li
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Zongtao Wang
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Qingshuang Lv
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Wenjie Mei
- School
of Pharmacy, Guangdong Engineering Technology Research Centre of Molecular
Probe and Biomedicine Imaging, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
18
|
Yang J, Xu J, Xu S, Fan Z, Zhu C, Wan J, Yang J, Xing X. Oxidative stress in acute pulmonary embolism: emerging roles and therapeutic implications. Thromb J 2024; 22:9. [PMID: 38216919 PMCID: PMC10785361 DOI: 10.1186/s12959-023-00577-1] [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: 08/24/2023] [Accepted: 12/25/2023] [Indexed: 01/14/2024] Open
Abstract
Oxidative stress is an imbalance between the body's reactive oxygen species and antioxidant defense mechanisms. Oxidative stress is involved in the development of several cardiovascular diseases, such as pulmonary hypertension, atherosclerosis, and diabetes mellitus. A growing number of studies have suggested the potential role of oxidative stress in the pathogenesis of pulmonary embolism. Biomarkers of oxidative stress in pulmonary embolism have also been explored, such as matrix metalloproteinases, asymmetric dimethylarginine, and neutrophil/lymphocyte ratio. Here, we comprehensively summarize some oxidative stress mechanisms and biomarkers in the development of acute pulmonary embolism and summarize related treatments based on antioxidant stress to explore effective treatment strategies for acute pulmonary embolism.
Collapse
Affiliation(s)
- Jingchao Yang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, China
| | - Jinzhu Xu
- Department of Pulmonary and Critical Care Medicine, Yuxi Municipal Hospital of T.C. M, 653100, Yuxi, China
| | - Shuanglan Xu
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Yunnan University, 650021, Kunming, China
| | - Zeqin Fan
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Yunnan University, 650021, Kunming, China
| | - Chenshao Zhu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, China
| | - Jianyuan Wan
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, China
| | - Jiao Yang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, China.
| | - Xiqian Xing
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Yunnan University, 650021, Kunming, China.
| |
Collapse
|
19
|
Xiong S, Liu Q, Zhou S, Xiao Y. Identification of key genes and regulatory networks involved in the Comorbidity of atrial fibrillation and chronic obstructive pulmonary disease. Heliyon 2023; 9:e22430. [PMID: 39811093 PMCID: PMC11731475 DOI: 10.1016/j.heliyon.2023.e22430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 01/16/2025] Open
Abstract
Background The underlying molecular processes of atrial fibrillation (AF) and chronic obstructive pulmonary disease (COPD) are frequently linked to increased morbidity and mortality when they co-occur. However, their underlying molecular mechanisms are questioned due to their incomplete analysis. Objective This study aimed to identify common differentially expressed genes (DEGs) in AF and COPD patients and investigate their potential biological functions and pathways. We hope to complement and update previous research through clearer figure presentation and different bioinformatic analysis methods with different datasets. Methods We used statistical analysis to identify DEGs in the expression profiles of AF and COPD patients using datasets from the Gene Expression Omnibus database. To ascertain whether the common DEGs were functionally enriched, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were used. In addition, we generated protein‒protein interaction networks and identified significant hub genes. Furthermore, the hub genes were used to analyze transcription factor (TF)-gene interactions and TF-miRNA coregulatory networks, and their expression levels were validated in additional datasets. Results We identified a total of 15 DEGs that were upregulated, whereas 36 were downregulated in AF and COPD patients. The DEGs were commonly expressed in both AF and COPD patients, with functional enrichment analysis revealing their involvement in metabolic processes and neuron-to-neuron synapses. We identified significant hub genes, including TGM2, ITPR1, CHL1, ALDOC, RPS3, FBLN2, NDUFS2, ITGA5, CTNNB1, RBP1, CLSTN2, FABP5, EPHA4, LDHA, and HNRNPL, and analyzed their coexpression and biological functions. TF-gene interaction and TF-miRNA coregulatory network analyses revealed the regulatory relationships of the hub genes. Additional datasets were analyzed to validate hub gene expression, and ALDOC, HNRNPL, and NDUFS2 displayed similar processes in AF and COPD patients. Conclusions In our study, we demonstrate that metabolic processes and neuron-to-neuron synaptic connections may contribute to the cooccurrence of AF and COPD. The identified hub genes and regulatory networks may act as potential biomarkers and therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Shan Xiong
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Okoye CN, Koren SA, Wojtovich AP. Mitochondrial complex I ROS production and redox signaling in hypoxia. Redox Biol 2023; 67:102926. [PMID: 37871533 PMCID: PMC10598411 DOI: 10.1016/j.redox.2023.102926] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023] Open
Abstract
Mitochondria are a main source of cellular energy. Oxidative phosphorylation (OXPHOS) is the major process of aerobic respiration. Enzyme complexes of the electron transport chain (ETC) pump protons to generate a protonmotive force (Δp) that drives OXPHOS. Complex I is an electron entry point into the ETC. Complex I oxidizes nicotinamide adenine dinucleotide (NADH) and transfers electrons to ubiquinone in a reaction coupled with proton pumping. Complex I also produces reactive oxygen species (ROS) under various conditions. The enzymatic activities of complex I can be regulated by metabolic conditions and serves as a regulatory node of the ETC. Complex I ROS plays diverse roles in cell metabolism ranging from physiologic to pathologic conditions. Progress in our understanding indicates that ROS release from complex I serves important signaling functions. Increasing evidence suggests that complex I ROS is important in signaling a mismatch in energy production and demand. In this article, we review the role of ROS from complex I in sensing acute hypoxia.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Shon A Koren
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew P Wojtovich
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA; Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
21
|
Grayson C, Mailloux RJ. Coenzyme Q 10 and nicotinamide nucleotide transhydrogenase: Sentinels for mitochondrial hydrogen peroxide signaling. Free Radic Biol Med 2023; 208:260-271. [PMID: 37573896 DOI: 10.1016/j.freeradbiomed.2023.08.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/21/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Mitochondria use hydrogen peroxide (H2O2) as a mitokine for cell communication. H2O2 output for signaling depends on its rate of production and degradation, both of which are strongly affected by the redox state of the coenzyme Q10 (CoQ) pool and NADPH availability. Here, we propose the CoQ pool and nicotinamide nucleotide transhydrogenase (NNT) have evolved to be central modalities for mitochondrial H2O2 signaling. Both factors play opposing yet equally important roles in dictating H2O2 availability because they are connected to one another by two central parameters in bioenergetics: electron supply and Δp. The CoQ pool is the central point of convergence for electrons from various dehydrogenases and the electron transport chain (ETC). The increase in Δp creates a significant amount of protonic backpressure on mitochondria to promote H2O2 genesis through CoQ pool reduction. These same factors also drive the activity of NNT, which uses electrons and the Δp to eliminate H2O2. In this way, electron supply and the magnitude of the Δp manifests as a redox connection between the two sentinels, CoQ and NNT, which serve as opposing yet equally important forces required for budgeting H2O2. Taken together, CoQ and NNT are sentinels linked through mitochondrial bioenergetics to manage H2O2 availability for interorganelle and intercellular redox signaling.
Collapse
Affiliation(s)
- Cathryn Grayson
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
22
|
Feng J, Zhu W, Shi H, Peng D, Zang L, Wang Y, ZhaXi L, BaiMa J, Amevor FK, Wang X, Ma X, Zhao X. Analysis of the Selection Signal of the Tibetan Black Chicken Genome Based on Whole-Genome Sequencing. Genes (Basel) 2023; 14:1672. [PMID: 37761812 PMCID: PMC10531317 DOI: 10.3390/genes14091672] [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: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The Tibetan chicken has adapted well to high altitudes genetically after its long-term habitation in the plateau. In this study, we analyzed the selection signal of Tibetan black chickens (TBCs) and discovered genes associated with the characteristics of germplasm. METHODS Whole-genome sequencing (WGS) was used to identify the single-nucleotide polymorphism (SNP) markers and genetic structures in the genome of Tibetan black chickens. Further, we performed a comparative population genomics analysis between the genomic data obtained in this present study and the genomic data for five wild red jungle fowls (RJFs) accessed from the NCBI database (GenBank accession number PRJNA241474). Thereafter, the Fst and Pi selections were used to identify genes under positive selection in the Tibetan black chicken genome. RESULTS A total of 9,490,690 SNPs were identified in the Tibetan black chickens. In addition, the results from the gene ontology (GO) analysis showed that 732 genes of TBCs were enriched in a total of 210 GO terms with specific molecular functions such as regulation of cellular catabolic process, the MAPK signaling pathway, regulation of ion transport, growth, morphogenesis and lung alveolus development which may provide a better mechanism to facilitate oxygen transport and utilization in TBCs. Moreover, the results from the KEGG analysis showed that 732 genes of the TBCs were significantly enriched in the calcium signaling pathway, circadian entrainment (ADCY1, GNG7 and PER3), oxytocin signaling pathway and pathways of multiple neurodegeneration diseases. In addition, the CD86 antigen (CD86) was identified as a gene associated with the immune response in chickens. It was also revealed that genes such as TRIT1, HPCAL4, NT5C1A and HEYL were discovered under selection in Tibetan black chickens on chromosome 23. These genes may be related to the local adaptive characteristics of Tibetan black chickens, for instance, NT5C1A and HEYL may be involved in the high-altitude adaption of oxygen delivery in Tibetan black chickens. CONCLUSIONS In summary, we found that selection mainly affects the disease resistance and cold acclimatization of Tibetan black chickens. Hence, these results may provide important genetic information for the evolution and breeding of Tibetan black chickens.
Collapse
Affiliation(s)
- Jing Feng
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, China; (H.S.); (D.P.); (Y.W.); (X.M.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850009, China
| | - Wei Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (W.Z.); (F.K.A.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hairen Shi
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, China; (H.S.); (D.P.); (Y.W.); (X.M.)
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - Da Peng
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, China; (H.S.); (D.P.); (Y.W.); (X.M.)
| | - Lei Zang
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, China; (H.S.); (D.P.); (Y.W.); (X.M.)
| | - Yan Wang
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, China; (H.S.); (D.P.); (Y.W.); (X.M.)
| | - Luobu ZhaXi
- Shannan Longzi County Agriculture and Animal Husbandry Comprehensive Service Center, Shannan 856600, China (J.B.)
| | - Jiancai BaiMa
- Shannan Longzi County Agriculture and Animal Husbandry Comprehensive Service Center, Shannan 856600, China (J.B.)
| | - Felix Kwame Amevor
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (W.Z.); (F.K.A.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoqi Wang
- Agriculture and Animal Husbandry Comprehensive Service Center of Lazi County, Shigatse 858100, China;
| | - Xueying Ma
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, China; (H.S.); (D.P.); (Y.W.); (X.M.)
| | - Xiaoling Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (W.Z.); (F.K.A.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
23
|
Geng Y, Hu Y, Zhang F, Tuo Y, Ge R, Bai Z. Mitochondria in hypoxic pulmonary hypertension, roles and the potential targets. Front Physiol 2023; 14:1239643. [PMID: 37645564 PMCID: PMC10461481 DOI: 10.3389/fphys.2023.1239643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023] Open
Abstract
Mitochondria are the centrol hub for cellular energy metabolisms. They regulate fuel metabolism by oxygen levels, participate in physiological signaling pathways, and act as oxygen sensors. Once oxygen deprived, the fuel utilizations can be switched from mitochondrial oxidative phosphorylation to glycolysis for ATP production. Notably, mitochondria can also adapt to hypoxia by making various functional and phenotypes changes to meet the demanding of oxygen levels. Hypoxic pulmonary hypertension is a life-threatening disease, but its exact pathgenesis mechanism is still unclear and there is no effective treatment available until now. Ample of evidence indicated that mitochondria play key factor in the development of hypoxic pulmonary hypertension. By hypoxia-inducible factors, multiple cells sense and transmit hypoxia signals, which then control the expression of various metabolic genes. This activation of hypoxia-inducible factors considered associations with crosstalk between hypoxia and altered mitochondrial metabolism, which plays an important role in the development of hypoxic pulmonary hypertension. Here, we review the molecular mechanisms of how hypoxia affects mitochondrial function, including mitochondrial biosynthesis, reactive oxygen homeostasis, and mitochondrial dynamics, to explore the potential of improving mitochondrial function as a strategy for treating hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Yumei Geng
- Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Yu Hu
- Department of Pharmacy, Qinghai Provincial Traffic Hospital, Xining, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Yajun Tuo
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Rili Ge
- Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Research Center for High Altitude Medicine, Qinghai University, Xining, China
| | - Zhenzhong Bai
- Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Research Center for High Altitude Medicine, Qinghai University, Xining, China
| |
Collapse
|
24
|
Du J, Sudlow LC, Shahverdi K, Zhou H, Michie M, Schindler TH, Mitchell JD, Mollah S, Berezin MY. Oxaliplatin-induced cardiotoxicity in mice is connected to the changes in energy metabolism in the heart tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542198. [PMID: 37292714 PMCID: PMC10245950 DOI: 10.1101/2023.05.24.542198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oxaliplatin is a platinum-based alkylating chemotherapeutic agent used for cancer treatment. At high cumulative dosage, the negative effect of oxaliplatin on the heart becomes evident and is linked to a growing number of clinical reports. The aim of this study was to determine how chronic oxaliplatin treatment causes the changes in energy-related metabolic activity in the heart that leads to cardiotoxicity and heart damage in mice. C57BL/6 male mice were treated with a human equivalent dosage of intraperitoneal oxaliplatin (0 and 10 mg/kg) once a week for eight weeks. During the treatment, mice were followed for physiological parameters, ECG, histology and RNA sequencing of the heart. We identified that oxaliplatin induces strong changes in the heart and affects the heart's energy-related metabolic profile. Histological post-mortem evaluation identified focal myocardial necrosis infiltrated with a small number of associated neutrophils. Accumulated doses of oxaliplatin led to significant changes in gene expression related to energy related metabolic pathways including fatty acid (FA) oxidation, amino acid metabolism, glycolysis, electron transport chain, and NAD synthesis pathway. At high accumulative doses of oxaliplatin, the heart shifts its metabolism from FAs to glycolysis and increases lactate production. It also leads to strong overexpression of genes in NAD synthesis pathways such as Nmrk2. Changes in gene expression associated with energy metabolic pathways can be used to develop diagnostic methods to detect oxaliplatin-induced cardiotoxicity early on as well as therapy to compensate for the energy deficit in the heart to prevent heart damage.
Collapse
Affiliation(s)
- Junwei Du
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
- Institute of Materials Science & Engineering Washington University, St. Louis, MO 63130, USA
| | - Leland C Sudlow
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Kiana Shahverdi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Haiying Zhou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Megan Michie
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Thomas H Schindler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Joshua D Mitchell
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamim Mollah
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Mikhail Y Berezin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
- Institute of Materials Science & Engineering Washington University, St. Louis, MO 63130, USA
| |
Collapse
|
25
|
Transgenic NADH dehydrogenase restores oxygen regulation of breathing in mitochondrial complex I-deficient mice. Nat Commun 2023; 14:1172. [PMID: 36859533 PMCID: PMC9977773 DOI: 10.1038/s41467-023-36894-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
The hypoxic ventilatory response (HVR) is a life-saving reflex, triggered by the activation of chemoreceptor glomus cells in the carotid body (CB) connected with the brainstem respiratory center. The molecular mechanisms underlying glomus cell acute oxygen (O2) sensing are unclear. Genetic disruption of mitochondrial complex I (MCI) selectively abolishes the HVR and glomus cell responsiveness to hypoxia. However, it is unknown what functions of MCI (metabolic, proton transport, or signaling) are essential for O2 sensing. Here we show that transgenic mitochondrial expression of NDI1, a single-molecule yeast NADH/quinone oxidoreductase that does not directly contribute to proton pumping, fully recovers the HVR and glomus cell sensitivity to hypoxia in MCI-deficient mice. Therefore, maintenance of mitochondrial NADH dehydrogenase activity and the electron transport chain are absolutely necessary for O2-dependent regulation of breathing. NDI1 expression also rescues other systemic defects caused by MCI deficiency. These data explain the role of MCI in acute O2 sensing by arterial chemoreceptors and demonstrate the optimal recovery of complex organismal functions by gene therapy.
Collapse
|
26
|
Breault NM, Wu D, Dasgupta A, Chen KH, Archer SL. Acquired disorders of mitochondrial metabolism and dynamics in pulmonary arterial hypertension. Front Cell Dev Biol 2023; 11:1105565. [PMID: 36819102 PMCID: PMC9933518 DOI: 10.3389/fcell.2023.1105565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is an orphan disease of the cardiopulmonary unit that reflects an obstructive pulmonary vasculopathy and presents with hypertrophy, inflammation, fibrosis, and ultimately failure of the right ventricle (RVF). Despite treatment using pulmonary hypertension (PH)-targeted therapies, persistent functional impairment reduces the quality of life for people with PAH and death from RVF occurs in approximately 40% of patients within 5 years of diagnosis. PH-targeted therapeutics are primarily vasodilators and none, alone or in combination, are curative. This highlights a need to therapeutically explore molecular targets in other pathways that are involved in the pathogenesis of PAH. Several candidate pathways in PAH involve acquired mitochondrial dysfunction. These mitochondrial disorders include: 1) a shift in metabolism related to increased expression of pyruvate dehydrogenase kinase and pyruvate kinase, which together increase uncoupled glycolysis (Warburg metabolism); 2) disruption of oxygen-sensing related to increased expression of hypoxia-inducible factor 1α, resulting in a state of pseudohypoxia; 3) altered mitochondrial calcium homeostasis related to impaired function of the mitochondrial calcium uniporter complex, which elevates cytosolic calcium and reduces intramitochondrial calcium; and 4) abnormal mitochondrial dynamics related to increased expression of dynamin-related protein 1 and its binding partners, such as mitochondrial dynamics proteins of 49 kDa and 51 kDa, and depressed expression of mitofusin 2, resulting in increased mitotic fission. These acquired mitochondrial abnormalities increase proliferation and impair apoptosis in most pulmonary vascular cells (including endothelial cells, smooth muscle cells and fibroblasts). In the RV, Warburg metabolism and induction of glutaminolysis impairs bioenergetics and promotes hypokinesis, hypertrophy, and fibrosis. This review will explore our current knowledge of the causes and consequences of disordered mitochondrial function in PAH.
Collapse
Affiliation(s)
- Nolan M. Breault
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Danchen Wu
- Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Danchen Wu, ; Stephen L. Archer,
| | - Asish Dasgupta
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Stephen L. Archer
- Department of Medicine, Queen’s University, Kingston, ON, Canada,Queen’s Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Danchen Wu, ; Stephen L. Archer,
| |
Collapse
|
27
|
Tryphena KP, Anuradha U, Kumar R, Rajan S, Srivastava S, Singh SB, Khatri DK. Understanding the Involvement of microRNAs in Mitochondrial Dysfunction and Their Role as Potential Biomarkers and Therapeutic Targets in Parkinson's Disease. J Alzheimers Dis 2023; 94:S187-S202. [PMID: 35848027 PMCID: PMC10473154 DOI: 10.3233/jad-220449] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting the elderly worldwide and causing significant movement impairments. The goal of PD treatment is to restore dopamine levels in the striatum and regulate movement symptoms. The lack of specific biomarkers for early diagnosis, as well as medication aimed at addressing the pathogenic mechanisms to decelerate the progression of dopaminergic neurodegeneration, are key roadblocks in the management of PD. Various pathogenic processes have been identified to be involved in the progression of PD, with mitochondrial dysfunction being a major contributor to the disease's pathogenesis. The regulation of mitochondrial functions is influenced by a variety of factors, including epigenetics. microRNAs (miRNAs) are epigenetic modulators involved in the regulation of gene expression and regulate a variety of proteins that essential for proper mitochondrial functioning. They are found to be dysregulated in PD, as evidenced by biological samples from PD patients and in vitro and in vivo research. In this article, we attempt to provide an overview of several miRNAs linked to mitochondrial dysfunction and their potential as diagnostic biomarkers and therapeutic targets in PD.
Collapse
Affiliation(s)
- Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Urati Anuradha
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Rohith Kumar
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shruti Rajan
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shashi Bala Singh
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| |
Collapse
|
28
|
Basu K, Dey A, Kiran M. Inefficient splicing of long non-coding RNAs is associated with higher transcript complexity in human and mouse. RNA Biol 2023; 20:563-572. [PMID: 37543950 PMCID: PMC10405767 DOI: 10.1080/15476286.2023.2242649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023] Open
Abstract
Recent reports show that long non-coding RNAs (lncRNAs) have inefficient splicing and fewer alternative splice variants than mRNAs. Here, we have explored the efficiency of lncRNAs and mRNAs in producing various splice variants, given the number of exons in humans and mice. Intriguingly, lncRNAs produce more splice variants per exon, referred to as Transcript Complexity, than mRNAs. Most lncRNA splice variants are the product of the alternative last exon and exon skipping. LncRNAs and mRNAs with higher transcript complexity have shorter intron lengths. Longer exon length and GC/AG at 5'/3' splice sites are associated with higher transcript complexity in lncRNAs. Lastly, our results indicate that inefficient splicing of lncRNAs may facilitate multiple introns splicing and, thus, more spliced products per exon.
Collapse
Affiliation(s)
- Koushiki Basu
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Anubha Dey
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Manjari Kiran
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
29
|
Moreno-Domínguez A, Colinas O, Smani T, Ureña J, López-Barneo J. Acute oxygen sensing by vascular smooth muscle cells. Front Physiol 2023; 14:1142354. [PMID: 36935756 PMCID: PMC10020353 DOI: 10.3389/fphys.2023.1142354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
An adequate supply of oxygen (O2) is essential for most life forms on earth, making the delivery of appropriate levels of O2 to tissues a fundamental physiological challenge. When O2 levels in the alveoli and/or blood are low, compensatory adaptive reflexes are produced that increase the uptake of O2 and its distribution to tissues within a few seconds. This paper analyzes the most important acute vasomotor responses to lack of O2 (hypoxia): hypoxic pulmonary vasoconstriction (HPV) and hypoxic vasodilation (HVD). HPV affects distal pulmonary (resistance) arteries, with its homeostatic role being to divert blood to well ventilated alveoli to thereby optimize the ventilation/perfusion ratio. HVD is produced in most systemic arteries, in particular in the skeletal muscle, coronary, and cerebral circulations, to increase blood supply to poorly oxygenated tissues. Although vasomotor responses to hypoxia are modulated by endothelial factors and autonomic innervation, it is well established that arterial smooth muscle cells contain an acute O2 sensing system capable of detecting changes in O2 tension and to signal membrane ion channels, which in turn regulate cytosolic Ca2+ levels and myocyte contraction. Here, we summarize current knowledge on the nature of O2 sensing and signaling systems underlying acute vasomotor responses to hypoxia. We also discuss similarities and differences existing in O2 sensors and effectors in the various arterial territories.
Collapse
Affiliation(s)
- Alejandro Moreno-Domínguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Olaia Colinas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Tarik Smani
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Juan Ureña
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- *Correspondence: José López-Barneo,
| |
Collapse
|
30
|
Archer SL, Dasgupta A, Chen KH, Wu D, Baid K, Mamatis JE, Gonzalez V, Read A, Bentley RE, Martin AY, Mewburn JD, Dunham-Snary KJ, Evans GA, Levy G, Jones O, Al-Qazazi R, Ring B, Alizadeh E, Hindmarch CC, Rossi J, Lima PDA, Falzarano D, Banerjee A, Colpitts CC. SARS-CoV-2 mitochondriopathy in COVID-19 pneumonia exacerbates hypoxemia. Redox Biol 2022; 58:102508. [PMID: 36334378 PMCID: PMC9558649 DOI: 10.1016/j.redox.2022.102508] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022] Open
Abstract
RATIONALE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19 pneumonia. We hypothesize that SARS-CoV-2 causes alveolar injury and hypoxemia by damaging mitochondria in airway epithelial cells (AEC) and pulmonary artery smooth muscle cells (PASMC), triggering apoptosis and bioenergetic impairment, and impairing hypoxic pulmonary vasoconstriction (HPV), respectively. OBJECTIVES We examined the effects of: A) human betacoronaviruses, SARS-CoV-2 and HCoV-OC43, and individual SARS-CoV-2 proteins on apoptosis, mitochondrial fission, and bioenergetics in AEC; and B) SARS-CoV-2 proteins and mouse hepatitis virus (MHV-1) infection on HPV. METHODS We used transcriptomic data to identify temporal changes in mitochondrial-relevant gene ontology (GO) pathways post-SARS-CoV-2 infection. We also transduced AECs with SARS-CoV-2 proteins (M, Nsp7 or Nsp9) and determined effects on mitochondrial permeability transition pore (mPTP) activity, relative membrane potential, apoptosis, mitochondrial fission, and oxygen consumption rates (OCR). In human PASMC, we assessed the effects of SARS-CoV-2 proteins on hypoxic increases in cytosolic calcium, an HPV proxy. In MHV-1 pneumonia, we assessed HPV via cardiac catheterization and apoptosis using the TUNEL assay. RESULTS SARS-CoV-2 regulated mitochondrial apoptosis, mitochondrial membrane permeabilization and electron transport chain (ETC) GO pathways within 2 hours of infection. SARS-CoV-2 downregulated ETC Complex I and ATP synthase genes, and upregulated apoptosis-inducing genes. SARS-CoV-2 and HCoV-OC43 upregulated and activated dynamin-related protein 1 (Drp1) and increased mitochondrial fission. SARS-CoV-2 and transduced SARS-CoV-2 proteins increased apoptosis inducing factor (AIF) expression and activated caspase 7, resulting in apoptosis. Coronaviruses also reduced OCR, decreased ETC Complex I activity and lowered ATP levels in AEC. M protein transduction also increased mPTP opening. In human PASMC, M and Nsp9 proteins inhibited HPV. In MHV-1 pneumonia, infected AEC displayed apoptosis and HPV was suppressed. BAY K8644, a calcium channel agonist, increased HPV and improved SpO2. CONCLUSIONS Coronaviruses, including SARS-CoV-2, cause AEC apoptosis, mitochondrial fission, and bioenergetic impairment. SARS-CoV-2 also suppresses HPV by targeting mitochondria. This mitochondriopathy is replicated by transduction with SARS-CoV-2 proteins, indicating a mechanistic role for viral-host mitochondrial protein interactions. Mitochondriopathy is a conserved feature of coronaviral pneumonia that may exacerbate hypoxemia and constitutes a therapeutic target.
Collapse
Affiliation(s)
- Stephen L Archer
- Department of Medicine, Queen's University, Kingston, ON, Canada; Queen's Cardiopulmonary Unit (QCPU), Queen's University, Kingston, ON, Canada.
| | - Asish Dasgupta
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Kaushal Baid
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Victoria Gonzalez
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada; Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan; Saskatoon, SK, Canada
| | - Austin Read
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | | | - Ashley Y Martin
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | | | - Kimberly J Dunham-Snary
- Department of Medicine, Queen's University, Kingston, ON, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Gerald A Evans
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Gary Levy
- University of Toronto, Toronto, ON, Canada
| | - Oliver Jones
- Queen's Cardiopulmonary Unit (QCPU), Queen's University, Kingston, ON, Canada
| | - Ruaa Al-Qazazi
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Brooke Ring
- Queen's Cardiopulmonary Unit (QCPU), Queen's University, Kingston, ON, Canada
| | - Elahe Alizadeh
- Queen's Cardiopulmonary Unit (QCPU), Queen's University, Kingston, ON, Canada
| | | | - Jenna Rossi
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Patricia DA Lima
- Queen's Cardiopulmonary Unit (QCPU), Queen's University, Kingston, ON, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada; Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan; Saskatoon, SK, Canada
| | - Arinjay Banerjee
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada; Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan; Saskatoon, SK, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Biology, University of Waterloo; Waterloo, ON, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
31
|
Pak O, Nolte A, Knoepp F, Giordano L, Pecina P, Hüttemann M, Grossman LI, Weissmann N, Sommer N. Mitochondrial oxygen sensing of acute hypoxia in specialized cells - Is there a unifying mechanism? BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148911. [PMID: 35988811 DOI: 10.1016/j.bbabio.2022.148911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Acclimation to acute hypoxia through cardiorespiratory responses is mediated by specialized cells in the carotid body and pulmonary vasculature to optimize systemic arterial oxygenation and thus oxygen supply to the tissues. Acute oxygen sensing by these cells triggers hyperventilation and hypoxic pulmonary vasoconstriction which limits pulmonary blood flow through areas of low alveolar oxygen content. Oxygen sensing of acute hypoxia by specialized cells thus is a fundamental pre-requisite for aerobic life and maintains systemic oxygen supply. However, the primary oxygen sensing mechanism and the question of a common mechanism in different specialized oxygen sensing cells remains unresolved. Recent studies unraveled basic oxygen sensing mechanisms involving the mitochondrial cytochrome c oxidase subunit 4 isoform 2 that is essential for the hypoxia-induced release of mitochondrial reactive oxygen species and subsequent acute hypoxic responses in both, the carotid body and pulmonary vasculature. This review compares basic mitochondrial oxygen sensing mechanisms in the pulmonary vasculature and the carotid body.
Collapse
Affiliation(s)
- Oleg Pak
- Justus Liebig University, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Anika Nolte
- Justus Liebig University, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Fenja Knoepp
- Justus Liebig University, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Luca Giordano
- Justus Liebig University, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Petr Pecina
- Laboratory of Bioenergetics, Institute of Physiology CAS, Prague, Czech Republic
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Norbert Weissmann
- Justus Liebig University, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Natascha Sommer
- Justus Liebig University, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
32
|
Yang W, Cui H, Chai Z, Zou P, Shi F, Yang B, Zhang G, Yang H, Chen Q, Liu J, Cao J, Ling X, Ao L. Benzo[a]pyrene inhibits testosterone biosynthesis via NDUFA10-mediated mitochondrial compromise in mouse Leydig cells: Integrating experimental and in silico toxicological approaches. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114075. [PMID: 36108438 DOI: 10.1016/j.ecoenv.2022.114075] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 06/15/2023]
Abstract
Benzo[a]pyrene (B[a]P), a representative of polycyclic aromatic hydrocarbons (PAHs), is ubiquitously spread in the environment and showing deleterious impacts on male steroidogenesis, including testosterone synthesis disorder. However, the precise mechanisms involved in B[a]P-induced steroidogenesis perturbation remains obscure. In the present study, we integrated in vivo tests, transcriptome profiling, in vitro assays, and conjoint in silico toxicological approaches to delineate the detailed mechanisms. In mouse models, we observed that B[a]P administration remarkably inhibited testosterone synthesis accompanied by ultrastructural impairments of mitochondria and mitophagosome formation in mouse Leydig cells. Transcriptome profiling showed that B[a]P down-regulated the expression of Ndufa9, Ndufa6, Ndufa10, and Ndufa5 in mouse testes, which are identified as critical genes involved in the assembly and functionality of mitochondrial complex I. In the in vitro tests, the bioactive B[a]P metabolite BPDE induced perturbation of testosterone synthesis by NDUFA10-mediated mitochondrial impairment, which was further exacerbated by mitophagy in TM3 Leydig cells. The findings of in silico toxicological analyses were highly consistent with the experimental observations and further unveiled that B[a]P/BPDE-involved PPARα activation could serve as a molecular initiating event to trigger the decline in Ndufa10 expression and testosterone synthesis. Overall, we have shown the first evidence that mitochondrial compromise in Leydig cells is the extremely crucial target in B[a]P-induced steroidogenesis perturbation.
Collapse
Affiliation(s)
- Wang Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Haonan Cui
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Zili Chai
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Peng Zou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Fuquan Shi
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Binwei Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Guowei Zhang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Huan Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Jinyi Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.
| | - Xi Ling
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.
| | - Lin Ao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
33
|
Oxygen regulation of breathing is abolished in mitochondrial complex III-deficient arterial chemoreceptors. Proc Natl Acad Sci U S A 2022; 119:e2202178119. [PMID: 36122208 PMCID: PMC9522341 DOI: 10.1073/pnas.2202178119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Oxygen sensing by chemoreceptor glomus cells in the carotid body plays an essential adaptive function in health and disease; however, the underlying mechanisms are not fully understood. Glomus cells survive genetic disruption of mitochondrial complex III, although this results in a functional disconnection between the distal and proximal components of the mitochondrial electron transport chain (ETC). These cells exhibit selective abolition of mitochondrial and cellular responsiveness to hypoxia, as well as altered systemic hyperventilation and acclimatization to hypoxia, indicating that acute oxygen-sensing and -signaling during hypoxia result from the integrated action of mitochondrial ETC components. The mitochondrial ETC emerges as a complex oxygen-sensing and -signaling system of potential pathophysiological relevance in maladaptive responses to hypoxia. Acute oxygen (O2) sensing is essential for adaptation of organisms to hypoxic environments or medical conditions with restricted exchange of gases in the lung. The main acute O2-sensing organ is the carotid body (CB), which contains neurosecretory chemoreceptor (glomus) cells innervated by sensory fibers whose activation by hypoxia elicits hyperventilation and increased cardiac output. Glomus cells have mitochondria with specialized metabolic and electron transport chain (ETC) properties. Reduced mitochondrial complex (MC) IV activity by hypoxia leads to production of signaling molecules (NADH and reactive O2 species) in MCI and MCIII that modulate membrane ion channel activity. We studied mice with conditional genetic ablation of MCIII that disrupts the ETC in the CB and other catecholaminergic tissues. Glomus cells survived MCIII dysfunction but showed selective abolition of responsiveness to hypoxia (increased [Ca2+] and transmitter release) with normal responses to other stimuli. Mitochondrial hypoxic NADH and reactive O2 species signals were also suppressed. MCIII-deficient mice exhibited strong inhibition of the hypoxic ventilatory response and altered acclimatization to sustained hypoxia. These data indicate that a functional ETC, with coupling between MCI and MCIV, is required for acute O2 sensing. O2 regulation of breathing results from the integrated action of mitochondrial ETC complexes in arterial chemoreceptors.
Collapse
|
34
|
Islam MZ, Shen X, Pardue S, Kevil CG, Shackelford RE. The ataxia-telangiectasia mutated gene product regulates the cellular acid-labile sulfide fraction. DNA Repair (Amst) 2022; 116:103344. [PMID: 35696854 PMCID: PMC11118069 DOI: 10.1016/j.dnarep.2022.103344] [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: 03/08/2022] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 11/03/2022]
Abstract
The ataxia-telangiectasia mutated (ATM) protein regulates cell cycle checkpoints, the cellular redox state, and double-stranded DNA break repair. ATM loss causes the disorder ataxia-telangiectasia (A-T), distinguished by ataxia, telangiectasias, dysregulated cellular redox and iron responses, and an increased cancer risk. We examined the sulfur pool in A-T cells, with and without an ATM expression vector. While free and bound sulfide levels were not changed with ATM expression, the acid-labile sulfide faction was significantly increased. ATM expression also increased cysteine desulfurase (NFS1), NFU1 iron-sulfur cluster scaffold homolog protein, and several mitochondrial complex I proteins' expression. Additionally, ATM expression suppressed cystathionine β-synthase and cystathionine γ-synthase protein expression, cystathionine γ-synthase enzymatic activity, and increased the reduced to oxidized glutathione ratio. This last observation is interesting, as dysregulated glutathione is implicated in A-T pathology. As ATM expression increases the expression of proteins central in initiating 2Fe-2S and 4Fe-4S cluster formation (NFS1 and NFU1, respectively), and the acid-labile sulfide faction is composed of sulfur incorporated into Fe-S clusters, our data indicates that ATM regulates aspects of Fe-S cluster biosynthesis, the transsulfuration pathway, and glutathione redox cycling. Thus, our data may explain some of the redox- and iron-related pathologies seen in A-T.
Collapse
Affiliation(s)
- Mohammad Z Islam
- Department of Pathology & Translational Pathobiology, LSU Health Sciences Center Shreveport, Shreveport, LA 71130, United States
| | - Xinggui Shen
- Department of Pathology & Translational Pathobiology, LSU Health Sciences Center Shreveport, Shreveport, LA 71130, United States
| | - Sibile Pardue
- Department of Pathology & Translational Pathobiology, LSU Health Sciences Center Shreveport, Shreveport, LA 71130, United States
| | - Christopher G Kevil
- Department of Pathology & Translational Pathobiology, LSU Health Sciences Center Shreveport, Shreveport, LA 71130, United States
| | - Rodney E Shackelford
- Department of Pathology & Translational Pathobiology, LSU Health Sciences Center Shreveport, Shreveport, LA 71130, United States.
| |
Collapse
|
35
|
Gao L, Ortega-Sáenz P, Moreno-Domínguez A, López-Barneo J. Mitochondrial Redox Signaling in O 2-Sensing Chemoreceptor Cells. Antioxid Redox Signal 2022; 37:274-289. [PMID: 35044243 DOI: 10.1089/ars.2021.0255] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Acute responses to hypoxia are essential for the survival of mammals. The carotid body (CB), the main arterial chemoreceptor, contains glomus cells with oxygen (O2)-sensitive K+ channels, which are inhibited during hypoxia to trigger adaptive cardiorespiratory reflexes. Recent Advances: In this review, recent advances in molecular mechanisms of acute O2 sensing in CB glomus cells are discussed, with a special focus on the signaling role of mitochondria through regulating cellular redox status. These advances have been achieved thanks to the use of genetically engineered redox-sensitive green fluorescent protein (roGFP) probes, which allowed us to monitor rapid changes in ROS production in real time in different subcellular compartments during hypoxia. This methodology was used in combination with conditional knockout mice models, pharmacological approaches, and transcriptomic studies. We have proposed a mitochondria-to-membrane signaling model of acute O2 sensing in which H2O2 released in the mitochondrial intermembrane space serves as a signaling molecule to inhibit K+ channels on the plasma membrane. Critical Issues: Changes in mitochondrial reactive oxygen species (ROS) production during acute hypoxia are highly compartmentalized in the submitochondrial regions. The use of redox-sensitive probes targeted to specific compartments is essential to fully understand the role of mitochondrial ROS in acute O2 sensing. Future Directions: Further studies are needed to specify the ROS and to characterize the target(s) of ROS in chemoreceptor cells during acute hypoxia. These data may also contribute to a more complete understanding of the implication of ROS in acute responses to hypoxia in O2-sensing cells in other organs. Antioxid. Redox Signal. 37, 274-289.
Collapse
Affiliation(s)
- Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Patricia Ortega-Sáenz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alejandro Moreno-Domínguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
36
|
Altered Transcriptional Regulation of Glycolysis in Circulating CD8+ T Cells of Rheumatoid Arthritis Patients. Genes (Basel) 2022; 13:genes13071216. [PMID: 35886000 PMCID: PMC9323564 DOI: 10.3390/genes13071216] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Peripheral T lymphocytes of rheumatoid arthritis (RA) patients show pathological changes in their metabolic pathways, especially glycolysis. These changes may drive the increased proliferation and tissue invasiveness of RA T cells. In order to study the transcriptional regulation underlying these alterations, we analysed publicly available RNA sequencing data from circulating T lymphocyte subsets of healthy individuals, untreated RA patients, and patients undergoing treatment for RA. Differential co-expression networks were created using sample-wise edge weights from an analysis called “linear interpolation to obtain network estimates for single sample” (lionessR), and annotated using the Gene Transcription Regulation Database (GTRD). Genes with high centrality scores were identified. CD8+ effector memory cells (Tem) and CD8+CD45RA+ effector memory cells (Temra) showed large changes in the transcriptional regulation of glycolysis in untreated RA. PFKFB3 and GAPDH were differentially regulated and had high centrality scores in CD8+ Tem cells. PFKFB3 downregulation may be due to HIF1A post transcriptional inhibition. Tocilizumab treatment partially reversed the RA-associated differential expression of several metabolic and regulatory genes. MYC was upregulated and had high centrality scores in RA CD8+ Temra cells; however, its glycolysis targets were unaltered. The upregulation of the PI3K-AKT and mTOR pathways may explain MYC upregulation.
Collapse
|
37
|
Qi B, Song L, Hu L, Guo D, Ren G, Peng T, Liu M, Fang Y, Li C, Zhang M, Li Y. Cardiac-specific overexpression of Ndufs1 ameliorates cardiac dysfunction after myocardial infarction by alleviating mitochondrial dysfunction and apoptosis. Exp Mol Med 2022; 54:946-960. [PMID: 35817848 PMCID: PMC9355970 DOI: 10.1038/s12276-022-00800-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 06/15/2023] Open
Abstract
Myocardial infarction (MI) is the leading cause of premature death among adults. Cardiomyocyte death and dysfunction of the remaining viable cardiomyocytes are the main pathological factors of heart failure after MI. Mitochondrial complexes are emerging as critical mediators for the regulation of cardiomyocyte function. However, the precise roles of mitochondrial complex subunits in heart failure after MI remain unclear. Here, we show that NADH:ubiquinone oxidoreductase core subunit S1 (Ndufs1) expression is decreased in the hearts of heart failure patients and mice with myocardial infarction. Furthermore, we found that cardiac-specific Ndufs1 overexpression alleviates cardiac dysfunction and myocardial fibrosis in the healing phase of MI. Our results demonstrated that Ndufs1 overexpression alleviates MI/hypoxia-induced ROS production and ROS-related apoptosis. Moreover, upregulation of Ndufs1 expression improved the reduced activity of complex I and impaired mitochondrial respiratory function caused by MI/hypoxia. Given that mitochondrial function and cardiomyocyte apoptosis are closely related to heart failure after MI, the results of this study suggest that targeting Ndufs1 may be a potential therapeutic strategy to improve cardiac function in patients with heart failure.
Collapse
Affiliation(s)
- Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Liqiang Song
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Gaotong Ren
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Tingwei Peng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Mingchuan Liu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Chunyu Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China.
| |
Collapse
|
38
|
Zuo J, Zhang Z, Luo M, Zhou L, Nice EC, Zhang W, Wang C, Huang C. Redox signaling at the crossroads of human health and disease. MedComm (Beijing) 2022; 3:e127. [PMID: 35386842 PMCID: PMC8971743 DOI: 10.1002/mco2.127] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Redox biology is at the core of life sciences, accompanied by the close correlation of redox processes with biological activities. Redox homeostasis is a prerequisite for human health, in which the physiological levels of nonradical reactive oxygen species (ROS) function as the primary second messengers to modulate physiological redox signaling by orchestrating multiple redox sensors. However, excessive ROS accumulation, termed oxidative stress (OS), leads to biomolecule damage and subsequent occurrence of various diseases such as type 2 diabetes, atherosclerosis, and cancer. Herein, starting with the evolution of redox biology, we reveal the roles of ROS as multifaceted physiological modulators to mediate redox signaling and sustain redox homeostasis. In addition, we also emphasize the detailed OS mechanisms involved in the initiation and development of several important diseases. ROS as a double-edged sword in disease progression suggest two different therapeutic strategies to treat redox-relevant diseases, in which targeting ROS sources and redox-related effectors to manipulate redox homeostasis will largely promote precision medicine. Therefore, a comprehensive understanding of the redox signaling networks under physiological and pathological conditions will facilitate the development of redox medicine and benefit patients with redox-relevant diseases.
Collapse
Affiliation(s)
- Jing Zuo
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for BiotherapyChengduP. R. China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for BiotherapyChengduP. R. China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for BiotherapyChengduP. R. China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for BiotherapyChengduP. R. China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVictoriaAustralia
| | - Wei Zhang
- West China Biomedical Big Data CenterWest China HospitalSichuan UniversityChengduP. R. China
- Mental Health Center and Psychiatric LaboratoryThe State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduP. R. China
| | - Chuang Wang
- Department of PharmacologyProvincial Key Laboratory of Pathophysiology, Ningbo University School of MedicineNingboZhejiangP. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for BiotherapyChengduP. R. China
- Department of PharmacologyProvincial Key Laboratory of Pathophysiology, Ningbo University School of MedicineNingboZhejiangP. R. China
| |
Collapse
|
39
|
A Novel Mitochondrial-Related Gene Signature for the Tumor Immune Microenvironment Evaluation and Prognosis Prediction in Lung Adenocarcinoma. J Immunol Res 2022; 2022:5366185. [PMID: 35664356 PMCID: PMC9159837 DOI: 10.1155/2022/5366185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022] Open
Abstract
Lung adenocarcinoma (LUAD) remains the most common deadly disease and has a poor prognosis. More and more studies have reported that mitochondrial-related genes (MTRGs) were associated with the clinical outcomes of multiple tumors solely. In this study, we aimed to develop a novel prognostic model based on MTRGs. Differentially expressed MTRGs were identified from TCGA-LUAD and GSE31210 cohorts. Univariate Cox regression analysis was utilized to screen differentially expressed MTRGs that were related to prognosis of LUAD. Then, LASSO Cox regression analysis was used to develop a prognostic signature. ESTIMATE was used for estimating the fractions of immune cell types. In this study, we identified 44 overlapping differentially expressed MTRGs in TCGA-LUAD and GSE31210 cohorts. Among 44 overlapping differentially expressed MTRGs, nine genes were associated with prognosis of LUAD. When the penalty parameter lambda was the minimum, there were six genes meeting the conditions of constructing the signature, including SERPINB5, CCNB1, FGR MAOB, SH3BP5, and CYP24A1. The survival analysis suggested that prognosis of patients in the high-risk group was significantly worse than that in the low-risk group. Cox regression analyses showed that the risk score was an independent predictor of LUAD prognosis. As with the results of ESTIMATE score, the degree of immune cell infiltration in the low-risk group was higher than that in the high-risk group, such as TIL, Treg, and B cells. In addition, TMB and cancer stem cell infiltration were higher in the low-risk group than the high-risk group. In conclusion, we developed a novel MTRG signature acting as a negative independent prognostic factor. In the future, individualized treatments and medical decision-making may benefit from using the predicted model.
Collapse
|
40
|
A reversible mitochondrial complex I thiol switch mediates hypoxic avoidance behavior in C. elegans. Nat Commun 2022; 13:2403. [PMID: 35504873 PMCID: PMC9064984 DOI: 10.1038/s41467-022-30169-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 04/18/2022] [Indexed: 01/28/2023] Open
Abstract
C. elegans react to metabolic distress caused by mismatches in oxygen and energy status via distinct behavioral responses. At the molecular level, these responses are coordinated by under-characterized, redox-sensitive processes, thought to initiate in mitochondria. Complex I of the electron transport chain is a major site of reactive oxygen species (ROS) production and is canonically associated with oxidative damage following hypoxic exposure. Here, we use a combination of optogenetics and CRISPR/Cas9-mediated genome editing to exert spatiotemporal control over ROS production. We demonstrate a photo-locomotory remodeling of avoidance behavior by local ROS production due to the reversible oxidation of a single thiol on the complex I subunit NDUF-2.1. Reversible thiol oxidation at this site is necessary and sufficient for the behavioral response to hypoxia, does not respond to ROS produced at more distal sites, and protects against lethal hypoxic exposure. Molecular modeling suggests that oxidation at this thiol residue alters the ability for NDUF-2.1 to coordinate electron transfer to coenzyme Q by destabilizing the Q-binding pocket, causing decreased complex I activity. Overall, site-specific ROS production regulates behavioral responses and these findings provide a mechanistic target to suppress the detrimental effects of hypoxia.
Collapse
|
41
|
Ex Vivo Lung Perfusion with β-Nicotinamide Adenine Dinucleotide (NAD+) Improves Ischemic Lung Function. Antioxidants (Basel) 2022; 11:antiox11050843. [PMID: 35624707 PMCID: PMC9137530 DOI: 10.3390/antiox11050843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 12/31/2022] Open
Abstract
Ischemia-reperfusion injury compromises short- and long-term outcomes after lung transplantation. The scarce existing data on NAD+ suggest effects on hypoxia-induced vasoconstriction, on reactive oxygen species and on tampering inflammation. We exposed rat lungs to 14 h of cold ischemic storage and perfused them in a rat ex vivo lung perfusion (EVLP) system for 4 h. A control group (n = 6) was compared to groups receiving 100 µM (n = 6) or 200 µM NAD+ (n = 6) in the preservation solution and groups receiving 200 µM (n = 4) or 2000 µM (n = 6) NAD+ every 30 min in the perfusate, starting at 1 h of EVLP. Compared to the control, significant effects were only achieved in the 2000 µM NAD+ group. During the 4 h of EVLP, we monitored higher vascular flow, lower mean pulmonary arterial pressure and increased oxygenation capacity. Tissue inflammation estimated with the myeloperoxidase assay was lower in the 2000 µM NAD+ group. We observed higher levels of anti-inflammatory IL-10, higher anti-inflammatory IL-6/IL-10 ratios and lower levels of pro-inflammatory IL-12 and IL-18 as well as a trend of more anti-inflammatory IFNy in the 2000 µM NAD+ perfusate. In the bronchoalveolar lavage, the pro-inflammatory levels of IL-1α and IL-1β were lower in the 2000 µM NAD+ group. NAD+ administered during EVLP is a promising agent with both anti-inflammatory properties and the ability to improve ischemic lung function.
Collapse
|
42
|
Maarman GJ. Reviewing the suitability of mitochondrial transplantation as therapeutic approach for pulmonary hypertension in the era of personalised medicine. Am J Physiol Lung Cell Mol Physiol 2022; 322:L641-L646. [PMID: 35318860 DOI: 10.1152/ajplung.00484.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a fatal disease, defined as a mean pulmonary artery pressure ≥ 25 mm Hg. It is caused, in part, by mitochondrial dysfunction. Among the various biological therapies proposed to rescue mitochondrial dysfunction, evidence going back as far as 2009, suggests that mitochondrial transplantation is an alternative. Although scant, recent PH findings and other literature supports a role for mitochondrial transplantation as a therapeutic approach in the context of PH. In experimental models of PH, it confers beneficial effects that include reduced pulmonary vasoconstriction, reduced pulmonary vascular remodelling, and improved right ventricular function. It also reduces the proliferation of pulmonary artery smooth muscle cells. However, first, we must understand that more research is needed before mitochondrial transplantation can be considered an effective therapy in the clinical setting, as many of the mechanisms or potential long-term risks are still unknown. Second, the current challenges of mitochondrial transplantation are surmountable and should not deter researchers from further investigating its effectiveness and trying to overcome these challenges in creative ways.
Collapse
Affiliation(s)
- Gerald J Maarman
- CARMA: Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
43
|
Important Functions and Molecular Mechanisms of Mitochondrial Redox Signaling in Pulmonary Hypertension. Antioxidants (Basel) 2022; 11:antiox11030473. [PMID: 35326123 PMCID: PMC8944689 DOI: 10.3390/antiox11030473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are important organelles that act as a primary site to produce reactive oxygen species (ROS). Additionally, mitochondria play a pivotal role in the regulation of Ca2+ signaling, fatty acid oxidation, and ketone synthesis. Dysfunction of these signaling molecules leads to the development of pulmonary hypertension (PH), atherosclerosis, and other vascular diseases. Features of PH include vasoconstriction and pulmonary artery (PA) remodeling, which can result from abnormal proliferation, apoptosis, and migration of PA smooth muscle cells (PASMCs). These responses are mediated by increased Rieske iron–sulfur protein (RISP)-dependent mitochondrial ROS production and increased mitochondrial Ca2+ levels. Mitochondrial ROS and Ca2+ can both synergistically activate nuclear factor κB (NF-κB) to trigger inflammatory responses leading to PH, right ventricular failure, and death. Evidence suggests that increased mitochondrial ROS and Ca2+ signaling leads to abnormal synthesis of ketones, which play a critical role in the development of PH. In this review, we discuss some of the recent findings on the important interactive role and molecular mechanisms of mitochondrial ROS and Ca2+ in the development and progression of PH. We also address the contributions of NF-κB-dependent inflammatory responses and ketone-mediated oxidative stress due to abnormal regulation of mitochondrial ROS and Ca2+ signaling in PH.
Collapse
|
44
|
Hannemann J, Böger R. Dysregulation of the Nitric Oxide/Dimethylarginine Pathway in Hypoxic Pulmonary Vasoconstriction—Molecular Mechanisms and Clinical Significance. Front Med (Lausanne) 2022; 9:835481. [PMID: 35252268 PMCID: PMC8891573 DOI: 10.3389/fmed.2022.835481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/27/2022] [Indexed: 12/21/2022] Open
Abstract
The pulmonary circulation responds to hypoxia with vasoconstriction, a mechanism that helps to adapt to short-lived hypoxic episodes. When sustained, hypoxic pulmonary vasoconstriction (HPV) may become deleterious, causing right ventricular hypertrophy and failure, and contributing to morbidity and mortality in the late stages of several chronic pulmonary diseases. Nitric oxide (NO) is an important endothelial vasodilator. Its release is regulated, amongst other mechanisms, by the presence of endogenous inhibitors like asymmetric dimethylarginine (ADMA). Evidence has accumulated in recent years that elevated ADMA may be implicated in the pathogenesis of HPV and in its clinical sequelae, like pulmonary arterial hypertension (PAH). PAH is one phenotypic trait in experimental models with disrupted ADMA metabolism. In high altitude, elevation of ADMA occurs during long-term exposure to chronic or chronic intermittent hypobaric hypoxia; ADMA is significantly associated with high altitude pulmonary hypertension. High ADMA concentration was also reported in patients with chronic obstructive lung disease, obstructive sleep apnoea syndrome, and overlap syndrome, suggesting a pathophysiological role for ADMA-mediated impairment of endothelium-dependent, NO-mediated pulmonary vasodilation in these clinically relevant conditions. Improved understanding of the molecular (dys-)regulation of pathways controlling ADMA concentration may help to dissect the pathophysiology and find novel therapeutic options for these diseases.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany
- *Correspondence: Rainer Böger
| |
Collapse
|
45
|
Kaludercic N, Di Lisa F. Cyclophilin D and p66Shc contribute to KCl-induced Ca2+ increase in pulmonary artery smooth muscle cells: a potentially relevant phenomenon awaiting a definite mechanism. Cardiovasc Res 2022; 118:16-17. [PMID: 34343269 DOI: 10.1093/cvr/cvab261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
- Institute for Pediatric Research Città della Speranza, Padova, Italy
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| |
Collapse
|
46
|
Tuo B, Xu J, Zhang W, Li X, Peng L, Zou Q, Deng Y, Lei J, Li H. Upregulation of miR-140-5p uncouples mitochondria by targeting Bcl-xL in vascular smooth muscle cells in angiotensin II-induced hypertension. Bioengineered 2022; 13:1137-1148. [PMID: 35258391 PMCID: PMC8805896 DOI: 10.1080/21655979.2021.2017696] [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: 10/14/2021] [Accepted: 12/08/2021] [Indexed: 11/10/2022] Open
Abstract
Angiotensin II-induced vascular smooth muscle cell (VSMC) remodeling and dysfunction is a major contributor to the development of hypertension. In spite of the low content of mitochondria and their low contribution to bioenergetics in VSMCs, recent studies have suggested that mitochondria play an important role in the regulation of VSMC function. However, the role of mitochondria in angiotensin II-induced VSMC dysfunction remains unknown. Here, we found that angiotensin II decreased the expression of Bcl-2-like protein 1 (Bcl-xL), a newly identified protein in inhibition of uncoupled proton flux in mitochondria through interaction with the β-subunit of ATP synthase, and uncoupled mitochondria in VSMCs both in vivo and in vitro. Overexpression of Bcl-xL restored the mitochondrial and VSMC function in response to angiotensin II treatment in vitro, suggesting that angiotensin II uncouples mitochondria through downregulation of Bcl-xL. Mechanistically, angiotensin II increased the expression of miR-140-5p, which targeted and downregulated Bcl-xL in VSMCs. Inhibition of miR-140-5p using antagomir-140-5p in vivo attenuated mitochondrial uncoupling and hypertension in angiotensin II-treated mice. These results suggested that upregulation of miR-140-5p uncouples mitochondria by targeting Bcl-xL in VSMCs in angiotensin II-induced hypertension, and miR-140-5p and Bcl-xL are potential targets for treatment of vascular dysfunction.
Collapse
Affiliation(s)
- Buxiong Tuo
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Jie Xu
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Wenqiang Zhang
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaomiao Li
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lijing Peng
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Qian Zou
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Ying Deng
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Junning Lei
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| | - Hui Li
- Department of Cardiology, 986 Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
47
|
Abstract
Oxygen (O2) is essential for life and therefore the supply of sufficient O2 to the tissues is a major physiological challenge. In mammals, a deficit of O2 (hypoxia) triggers rapid cardiorespiratory reflexes (e.g. hyperventilation and increased heart output) that within a few seconds increase the uptake of O2 by the lungs and its distribution throughout the body. The prototypical acute O2-sensing organ is the carotid body (CB), which contains sensory glomus cells expressing O2-regulated ion channels. In response to hypoxia, glomus cells depolarize and release transmitters which activate afferent fibers terminating at the brainstem respiratory and autonomic centers. In this review, we summarize the basic properties of CB chemoreceptor cells and the essential role played by their specialized mitochondria in acute O2 sensing and signaling. We focus on recent data supporting a "mitochondria-to-membrane signaling" model of CB chemosensory transduction. The possibility that the differential expression of specific subunit isoforms and enzymes could allow mitochondria to play a generalized adaptive O2-sensing and signaling role in a wide variety of cells is also discussed.
Collapse
Affiliation(s)
- José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Patricia Ortega-Sáenz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
48
|
Zhang X, Gao F. Exercise improves vascular health: Role of mitochondria. Free Radic Biol Med 2021; 177:347-359. [PMID: 34748911 DOI: 10.1016/j.freeradbiomed.2021.11.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 01/10/2023]
Abstract
Vascular mitochondria constantly integrate signals from environment and respond accordingly to match vascular function to metabolic requirements of the organ tissues, while mitochondrial dysfunction contributes to vascular aging and pathologies such as atherosclerosis, stenosis, and hypertension. As an effective lifestyle intervention, exercise induces extensive mitochondrial adaptations through vascular mechanical stress and the increased production and release of reactive oxygen species and nitric oxide that activate multiple intracellular signaling pathways, among which peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) plays a critical role. PGC-1α coordinates mitochondrial quality control mechanisms to maintain a healthy mitochondrial pool and promote endothelial nitric oxide synthase activity in vasculature. The mitochondrial adaptations to exercise improve bioenergetics, balance redox status, protect endothelial cells against detrimental insults, increase vascular plasticity, and ameliorate aging-related vascular dysfunction, thus benefiting vascular health. This review highlights recent findings of mitochondria as a central hub integrating exercise-afforded vascular benefits and its underlying mechanisms. A better understanding of the mitochondrial adaptations to exercise will not only shed light on the mechanisms of exercise-induced cardiovascular protection, but may also provide new clues to mitochondria-oriented precise exercise prescriptions for cardiovascular health.
Collapse
Affiliation(s)
- Xing Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
49
|
Zhang X, Dong W, Zhang J, Liu W, Yin J, Shi D, Ma W. A Novel Mitochondrial-Related Nuclear Gene Signature Predicts Overall Survival of Lung Adenocarcinoma Patients. Front Cell Dev Biol 2021; 9:740487. [PMID: 34760888 PMCID: PMC8573348 DOI: 10.3389/fcell.2021.740487] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/28/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Lung cancer is the leading cause of cancer-related death worldwide, of which lung adenocarcinoma (LUAD) is one of the main histological subtypes. Mitochondria are vital for maintaining the physiological function, and their dysfunction has been found to be correlated with tumorigenesis and disease progression. Although, some mitochondrial-related genes have been found to correlate with the clinical outcomes of multiple tumors solely. The integrated relationship between nuclear mitochondrial genes (NMGs) and the prognosis of LUAD remains unclear. Methods: The list of NMGs, gene expression data, and related clinical information of LUAD were downloaded from public databases. Bioinformatics methods were used and obtained 18 prognostic related NMGs to construct a risk signature. Results: There were 18 NMGs (NDUFS2, ATP8A2, SCO1, COX14, COA6, RRM2B, TFAM, DARS2, GARS, YARS2, EFG1, GFM1, MRPL3, MRPL44, ISCU, CABC1, HSPD1, and ETHE1) identified by LASSO regression analysis. The mRNA expression of these 18 genes was positively correlated with their relative linear copy number alteration (CNA). Meanwhile, the established risk signature could effectively distinguish high- and low-risk patients, and its predictive capacity was validated in three independent gene expression omnibus (GEO) cohorts. Notably, a significantly lower prevalence of actionable EGFR alterations was presented in patients with high-risk NMGs signature but accompanied with a more inflame immune tumor microenvironment. Additionally, multicomponent Cox regression analysis showed that the model was stable when risk score, tumor stage, and lymph node stage were considered, and the 1-, 3-, and 5-year AUC were 0.74, 0.75, and 0.70, respectively. Conclusion: Together, this study established a signature based on NMGs that is a prognostic biomarker for LUAD patients and has the potential to be widely applied in future clinical settings.
Collapse
Affiliation(s)
- Xiangwei Zhang
- Department of General Thoracic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Dong
- Department of General Thoracic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jishuai Zhang
- Department of General Thoracic, Feicheng Hospital Affiliated to Shandong First Medical University, Feicheng, China
| | - Wenqiang Liu
- Department of General Thoracic, Shenxian County People's Hospital of Shandong Provincial Group, Liaocheng, China
| | - Jingjing Yin
- Department of General Thoracic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Duozhi Shi
- Lifehealthcare Clinical Laboratories, Hangzhou, China
| | - Wei Ma
- Department of General Thoracic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
50
|
Mitochondrial iron-sulfur clusters: Structure, function, and an emerging role in vascular biology. Redox Biol 2021; 47:102164. [PMID: 34656823 PMCID: PMC8577454 DOI: 10.1016/j.redox.2021.102164] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 12/31/2022] Open
Abstract
Iron-sulfur (Fe-S) clusters are essential cofactors most commonly known for their role mediating electron transfer within the mitochondrial respiratory chain. The Fe-S cluster pathways that function within the respiratory complexes are highly conserved between bacteria and the mitochondria of eukaryotic cells. Within the electron transport chain, Fe-S clusters play a critical role in transporting electrons through Complexes I, II and III to cytochrome c, before subsequent transfer to molecular oxygen. Fe-S clusters are also among the binding sites of classical mitochondrial inhibitors, such as rotenone, and play an important role in the production of mitochondrial reactive oxygen species (ROS). Mitochondrial Fe-S clusters also play a critical role in the pathogenesis of disease. High levels of ROS produced at these sites can cause cell injury or death, however, when produced at low levels can serve as signaling molecules. For example, Ndufs2, a Complex I subunit containing an Fe-S center, N2, has recently been identified as a redox-sensitive oxygen sensor, mediating homeostatic oxygen-sensing in the pulmonary vasculature and carotid body. Fe-S clusters are emerging as transcriptionally-regulated mediators in disease and play a crucial role in normal physiology, offering potential new therapeutic targets for diseases including malaria, diabetes, and cancer.
Collapse
|