1
|
Wang Q, Duan D, Luo C, Huang J, Wei J, Zhang Y, Zhang K, Zhou T, Wang W, Yang S, Ma L. Astilbin exerts anti-hypersensitivity by regulating metabolic demand and neuronal activity in rodent model of neuropathic pain. Ann Med 2024; 56:2396561. [PMID: 39624967 PMCID: PMC11616750 DOI: 10.1080/07853890.2024.2396561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 05/22/2024] [Accepted: 07/26/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVE Astilbe chinensis, is a traditional Chinese medicine commonly employed for pain management. However, its primary active ingredient remains a subject of debate. METHODS Spinal nerve ligation (SNL) and formalin-induced pain models were employed. Network pharmacology and bioinformatics were utilized to identify targets. Verification was performed through spinal cord double immunofluorescence staining, quantitative PCR and whole-cell recording techniques. RESULTS In experiments conducted on neuropathic rats, both systemic and intrathecal administration of astilbin, an essential constituent, exhibited a noteworthy and dose-dependently decrease in chronic and acute pain behaviours. The ED50 value, which represents the dose at which 50% effectiveness is achieved, was measure at 7.59 μg, while the Emax value, indicating the maximum attainable effect, was found to be 60% of the maximal possible effect (% MPE). Forty-two shared targets were identified, enriching the metabolic and synaptic pathways in the network pharmacology analysis, as confirmed by transcriptomic analysis. Weighted gene co-expression network analysis (WGCNA) revealed a strong correlation between the anti-nociceptive effects of astilbin and neuronal metabolic processes. Spinal functional ultrasound (FUS) analysis indicated increased spinal blood flow intensity and changes in metabolism-related enzyme activity, including stearoyl-CoA desaturase (Scd), 17beta-hydroxysteroid dehydrogenase (Hsd17b7) and sterol 14alpha-demethylase (Cyp51) in neuropathic rats, pretreatment with astilbin decreased formalin-induced blood flow in acute pain. Bath application of astilbin dose-dependently inhibited neuronal activity by reducing the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs) without affecting miniature inhibitory postsynaptic currents (mIPSCs). CONCLUSIONS In summary, this study provides evidence that astilbin alleviates pain by modulating neuronal metabolic processes and synaptic homeostasis.
Collapse
Affiliation(s)
- Qiru Wang
- Department of Pharmacy, Shanghai Cancer Center, Fudan University, Minhang Branch, Shanghai, China
| | - Dongxia Duan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Chao Luo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinlu Huang
- Department of Pharmacy, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinbao Wei
- Department of Pharmacy, Xiamen Haicang Hospital, Xiamen, China
| | - Yang Zhang
- Shanghai Jiao Tong University Hospital, Shanghai, China
| | - Ke Zhang
- Department of Pharmacy, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Zhou
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Shaoxin Yang
- Shanghai Jiao Tong University School of Medicine, Shanghai Ninth People’s Hospital, Shanghai, China
| | - Le Ma
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| |
Collapse
|
2
|
Ishii M, Yamaguchi Y, Takada K, Hamaya H, Ogawa S, Akishita M. Effect of decreased expression of latent TGF-β binding proteins 4 on the pathogenesis of emphysema as an age-related disease. Arch Gerontol Geriatr 2024; 127:105597. [PMID: 39121531 DOI: 10.1016/j.archger.2024.105597] [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: 05/08/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
PURPOSE Latent TGF-β binding protein 4 (LTBP4) is involved in the production of elastin fibers and has been implicated in LTBP4-related cutis laxa and its complication, emphysema-like changes. Various factors have been implicated in the pathogenesis of emphysema, including elastic degeneration, inflammation, cellular senescence, mitochondrial dysfunction, and decreased angiogenesis in the lungs. We investigated the association between LTBP4 and emphysema using human lung fibroblasts with silenced LTBP4 genes. METHODS Cell contraction, elastin expression, cellular senescence, inflammation, anti-inflammatory factors, and mitochondrial function were compared between the LTBP4 small interfering RNA (siRNA) and control siRNA. RESULTS Under the suppression of LTBP4, significant changes were observed in the following: decreased cell contractility, decreased elastin expression, increased expression of the p16 gene involved in cellular senescence, increased TNFα, decreased GSTM3 and SOD, decreased mitochondrial membrane potential, and decreased VEGF expression. Furthermore, the decreased cell contractility and increased GSTM3 expression observed under LTBP4 suppression were restored by the addition of N-acetyl-L-cysteine or recombinant LTBP4. CONCLUSION The decreased elastin expression, cellular senescence, inflammation, decreased antioxidant activity, mitochondrial dysfunction, and decreased VEGF expression under reduced LTBP4 expression may all be involved in the destruction of the alveolar wall in emphysema. Smoking is the most common cause of emphysema; however, genetic factors related to LTBP4 expression and other factors may also contribute to its pathogenesis.
Collapse
Affiliation(s)
- Masaki Ishii
- The Department of Geriatric Medicine, The University of Tokyo, Japan.
| | - Yasuhiro Yamaguchi
- Division of Department of Respiratory Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Kazufumi Takada
- The Department of Geriatric Medicine, The University of Tokyo, Japan
| | - Hironobu Hamaya
- The Department of Geriatric Medicine, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| | - Sumito Ogawa
- The Department of Geriatric Medicine, The University of Tokyo, Japan
| | - Masahiro Akishita
- The Department of Geriatric Medicine, The University of Tokyo, Japan; The Department of Geriatric Medicine, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| |
Collapse
|
3
|
Wang J, Ding HK, Xu HJ, Hu DK, Hankey W, Chen L, Xiao J, Liang CZ, Zhao B, Xu LF. Single-cell analysis revealing the metabolic landscape of prostate cancer. Asian J Androl 2024; 26:451-463. [PMID: 38657119 PMCID: PMC11449408 DOI: 10.4103/aja20243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/29/2024] [Indexed: 04/26/2024] Open
Abstract
ABSTRACT Tumor metabolic reprogramming is a hallmark of cancer development, and targeting metabolic vulnerabilities has been proven to be an effective approach for castration-resistant prostate cancer (CRPC) treatment. Nevertheless, treatment failure inevitably occurs, largely due to cellular heterogeneity, which cannot be deciphered by traditional bulk sequencing techniques. By employing computational pipelines for single-cell RNA sequencing, we demonstrated that epithelial cells within the prostate are more metabolically active and plastic than stromal cells. Moreover, we identified that neuroendocrine (NE) cells tend to have high metabolic rates, which might explain the high demand for nutrients and energy exhibited by neuroendocrine prostate cancer (NEPC), one of the most lethal variants of prostate cancer (PCa). Additionally, we demonstrated through computational and experimental approaches that variation in mitochondrial activity is the greatest contributor to metabolic heterogeneity among both tumor cells and nontumor cells. These results establish a detailed metabolic landscape of PCa, highlight a potential mechanism of disease progression, and emphasize the importance of future studies on tumor heterogeneity and the tumor microenvironment from a metabolic perspective.
Collapse
Affiliation(s)
- Jing Wang
- Department of Urologic Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - He-Kang Ding
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, China
- Institute of Urology, Anhui Medical University, Hefei 230001, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230001, China
| | - Han-Jiang Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, China
- Institute of Urology, Anhui Medical University, Hefei 230001, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230001, China
| | - De-Kai Hu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, China
- Institute of Urology, Anhui Medical University, Hefei 230001, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230001, China
| | - William Hankey
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Chen
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Jun Xiao
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chao-Zhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, China
- Institute of Urology, Anhui Medical University, Hefei 230001, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230001, China
| | - Bing Zhao
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ling-Fan Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, China
- Institute of Urology, Anhui Medical University, Hefei 230001, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230001, China
| |
Collapse
|
4
|
Huan MJ, Fu PP, Chen X, Wang ZX, Ma ZR, Cai SZ, Jiang Q, Wang Q. Identification of the central role of RNA polymerase mitochondrial for angiogenesis. Cell Commun Signal 2024; 22:343. [PMID: 38907279 PMCID: PMC11191269 DOI: 10.1186/s12964-024-01712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024] Open
Abstract
Mitochondria are central to endothelial cell activation and angiogenesis, with the RNA polymerase mitochondrial (POLRMT) serving as a key protein in regulating mitochondrial transcription and oxidative phosphorylation. In our study, we examined the impact of POLRMT on angiogenesis and found that its silencing or knockout (KO) in human umbilical vein endothelial cells (HUVECs) and other endothelial cells resulted in robust anti-angiogenic effects, impeding cell proliferation, migration, and capillary tube formation. Depletion of POLRMT led to impaired mitochondrial function, characterized by mitochondrial depolarization, oxidative stress, lipid oxidation, DNA damage, and reduced ATP production, along with significant apoptosis activation. Conversely, overexpressing POLRMT promoted angiogenic activity in the endothelial cells. In vivo experiments demonstrated that endothelial knockdown of POLRMT, by intravitreous injection of endothelial specific POLRMT shRNA adeno-associated virus, inhibited retinal angiogenesis. In addition, inhibiting POLRMT with a first-in-class inhibitor IMT1 exerted significant anti-angiogenic impact in vitro and in vivo. Significantly elevated expression of POLRMT was observed in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. POLRMT endothelial knockdown inhibited pathological retinal angiogenesis and mitigated retinal ganglion cell (RGC) degeneration in DR mice. At last, POLRMT expression exhibited a substantial increase in the retinal proliferative membrane tissues of human DR patients. These findings collectively establish the indispensable role of POLRMT in angiogenesis, both in vitro and in vivo.
Collapse
Affiliation(s)
- Meng-Jia Huan
- Department of Ophthalmology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Ping-Ping Fu
- Department of Ophthalmology, Shanghai Eye Diseases Prevention & Treatment Center, Shanghai Eye Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xia Chen
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, 215025, China
| | - Zhao-Xia Wang
- Department of Endocrinology, Fengcheng Hospital of Fengxian Distric, Shanghai, China
| | - Zhou-Rui Ma
- Department of Burn and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Shi-Zhong Cai
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, China.
- Key Laboratory of Congenital Structural Malformations of Suzhou City, Suzhou, China.
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China.
| | - Qian Wang
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
5
|
Adamo KB, Goudreau AD, Corson AE, MacDonald ML, O'Rourke N, Tzaneva V. Physically active pregnancies: Insights from the placenta. Physiol Rep 2024; 12:e16104. [PMID: 38872466 PMCID: PMC11176744 DOI: 10.14814/phy2.16104] [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: 02/01/2024] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Physical activity (PA) positively influences pregnancy, a critical period for health promotion, and affects placental structure and function in ways previously overlooked. Here, we summarize the current body of literature examining the association between PA, placenta biology, and physiology while also highlighting areas where gaps in knowledge exist. PA during pregnancy induces metabolic changes, influencing nutrient availability and transporter expression in the placenta. Hormones and cytokines secreted during PA contribute to health benefits, with intricate interactions in pro- and anti-inflammatory markers. Extracellular vesicles and placental "-omics" data suggest that gestational PA can shape placental biology, affecting gene expression, DNA methylation, metabolite profiles, and protein regulation. However, whether cytokines that respond to PA alter placental proteomic profiles during pregnancy remains to be elucidated. The limited research on placenta mitochondria of physically active gestational parents (gesP), has shown improvements in mitochondrial DNA and antioxidant capacity, but the relationship between PA, placental mitochondrial dynamics, and lipid metabolism remains unexplored. Additionally, PA influences the placenta-immune microenvironment, angiogenesis, and may confer positive effects on neurodevelopment and mental health through placental changes, vascularization, and modulation of brain-derived neurotrophic factor. Ongoing exploration is crucial for unraveling the multifaceted impact of PA on the intricate placental environment.
Collapse
Affiliation(s)
- Kristi B Adamo
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandra D Goudreau
- Department of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Abbey E Corson
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Meaghan L MacDonald
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Nicholas O'Rourke
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Velislava Tzaneva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Stige KE, Kverneng SU, Sharma S, Skeie GO, Sheard E, Søgnen M, Geijerstam SA, Vetås T, Wahlvåg AG, Berven H, Buch S, Reese D, Babiker D, Mahdi Y, Wade T, Miranda GP, Ganguly J, Tamilselvam YK, Chai JR, Bansal S, Aur D, Soltani S, Adams S, Dölle C, Dick F, Berntsen EM, Grüner R, Brekke N, Riemer F, Goa PE, Haugarvoll K, Haacke EM, Jog M, Tzoulis C. The STRAT-PARK cohort: A personalized initiative to stratify Parkinson's disease. Prog Neurobiol 2024; 236:102603. [PMID: 38604582 DOI: 10.1016/j.pneurobio.2024.102603] [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: 02/01/2024] [Revised: 03/15/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
The STRAT-PARK initiative aims to provide a platform for stratifying Parkinson's disease (PD) into biological subtypes, using a bottom-up, multidisciplinary biomarker-based and data-driven approach. PD is a heterogeneous entity, exhibiting high interindividual clinicopathological variability. This diversity suggests that PD may encompass multiple distinct biological entities, each driven by different molecular mechanisms. Molecular stratification and identification of disease subtypes is therefore a key priority for understanding and treating PD. STRAT-PARK is a multi-center longitudinal cohort aiming to recruit a total of 2000 individuals with PD and neurologically healthy controls from Norway and Canada, for the purpose of identifying molecular disease subtypes. Clinical assessment is performed annually, whereas biosampling, imaging, and digital and neurophysiological phenotyping occur every second year. The unique feature of STRAT-PARK is the diversity of collected biological material, including muscle biopsies and platelets, tissues particularly useful for mitochondrial biomarker research. Recruitment rate is ∼150 participants per year. By March 2023, 252 participants were included, comprising 204 cases and 48 controls. STRAT-PARK is a powerful stratification initiative anticipated to become a global research resource, contributing to personalized care in PD.
Collapse
Affiliation(s)
- Kjersti Eline Stige
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway; K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, Bergen 5020, Norway; The Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology, Trondheim 7491, Norway; Department of Neurology and Clinical Neurophysiology, St Olav's University Hospital, Trondheim 7006, Norway
| | - Simon Ulvenes Kverneng
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway; K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, Bergen 5020, Norway
| | - Soumya Sharma
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Geir-Olve Skeie
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway
| | - Erika Sheard
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Mona Søgnen
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Solveig Af Geijerstam
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Therese Vetås
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Anne Grete Wahlvåg
- Department of Neurology and Clinical Neurophysiology, St Olav's University Hospital, Trondheim 7006, Norway
| | - Haakon Berven
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway; K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, Bergen 5020, Norway
| | - Sagar Buch
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - David Reese
- Imaging Research Laboratories, Robarts Research Institute, Ontario, London N6A 5B7, Canada
| | - Dina Babiker
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Yekta Mahdi
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Trevor Wade
- Department of Medical Biophysics, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, Ontario, London N6A 6B7, Canada
| | - Gala Prado Miranda
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Jacky Ganguly
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Yokhesh Krishnasamy Tamilselvam
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada; Department of Electrical and Computer Engineering, Canadian Surgical Technologies and Advanced Robotics (CSTAR), University of Western Ontario (UWO), Ontario, London, Canada
| | - Jia Ren Chai
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Saurabh Bansal
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Dorian Aur
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Sima Soltani
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Scott Adams
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada; School of Communication Sciences & Disorders, Faculty of Health Sciences, Western University, Canada
| | - Christian Dölle
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway; K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, Bergen 5020, Norway
| | - Fiona Dick
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway; K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, Bergen 5020, Norway
| | - Erik Magnus Berntsen
- Department of Radiology and Nuclear Medicine, St. Olav's University Hospital, Trondheim 7006, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Renate Grüner
- Department of Physics and Technology, University of Bergen, Bergen 5007, Norway; Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Post Office Box 1400, Bergen 5021, Norway
| | - Njål Brekke
- Department of Physics and Technology, University of Bergen, Bergen 5007, Norway; Radiology Department, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Frank Riemer
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Post Office Box 1400, Bergen 5021, Norway
| | - Pål Erik Goa
- Department of Radiology and Nuclear Medicine, St. Olav's University Hospital, Trondheim 7006, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Kristoffer Haugarvoll
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - E Mark Haacke
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA; Department of Radiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mandar Jog
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON N6A 5A5, Canada
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, Bergen 5020, Norway; K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, Bergen 5020, Norway.
| |
Collapse
|
7
|
Sadeghsoltani F, Hassanpour P, Safari MM, Haiaty S, Rahbarghazi R, Rahmati M, Mota A. Angiogenic activity of mitochondria; beyond the sole bioenergetic organelle. J Cell Physiol 2024; 239:e31185. [PMID: 38219050 DOI: 10.1002/jcp.31185] [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: 10/09/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/15/2024]
Abstract
Angiogenesis is a complex process that involves the expansion of the pre-existing vascular plexus to enhance oxygen and nutrient delivery and is stimulated by various factors, including hypoxia. Since the process of angiogenesis requires a lot of energy, mitochondria play an important role in regulating and promoting this phenomenon. Besides their roles as an oxidative metabolism base, mitochondria are potential bioenergetics organelles to maintain cellular homeostasis via sensing alteration in oxygen levels. Under hypoxic conditions, mitochondria can regulate angiogenesis through different factors. It has been indicated that unidirectional and bidirectional exchange of mitochondria or their related byproducts between the cells is orchestrated via different intercellular mechanisms such as tunneling nanotubes, extracellular vesicles, and gap junctions to maintain the cell homeostasis. Even though, the transfer of mitochondria is one possible mechanism by which cells can promote and regulate the process of angiogenesis under reperfusion/ischemia injury. Despite the existence of a close relationship between mitochondrial donation and angiogenic response in different cell types, the precise molecular mechanisms associated with this phenomenon remain unclear. Here, we aimed to highlight the possible role of mitochondria concerning angiogenesis, especially the role of mitochondrial transport and the possible relation of this transfer with autophagy, the housekeeping phenomenon of cells, and angiogenesis.
Collapse
Affiliation(s)
- Fatemeh Sadeghsoltani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hassanpour
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mir-Meghdad Safari
- Open Heart ICU of Shahid Madani Cardiovascular Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamad Rahmati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mota
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Laplace J, Bonneau B. [FUNDC1 promotes the formation of MAMs involved in angiogenesis]. Med Sci (Paris) 2024; 40:206-208. [PMID: 38411432 DOI: 10.1051/medsci/2023212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Affiliation(s)
- Julie Laplace
- M1 Biologie-Santé, Université Paris-Saclay, 91405 Orsay, France - DER de Biologie, École Normale Supérieure Paris-Saclay, Gif-sur-Yvette
| | | |
Collapse
|
9
|
Wei J, Xie J, He J, Li D, Wei D, Li Y, Li X, Fang W, Wei G, Lai K. Active fraction of Polyrhachis vicina (Roger) alleviated cerebral ischemia/reperfusion injury by targeting SIRT3-mediated mitophagy and angiogenesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155104. [PMID: 37797433 DOI: 10.1016/j.phymed.2023.155104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/01/2023] [Accepted: 09/17/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Damaged mitophagy and impaired angiogenesis involve in the pathogenic development of ischemic stroke. Active fraction of Polyrhachis vicina (Roger) (AFPR) showed great potential on neurological disease with it's remarkable anti-inflammatory and anti-oxidative effects. PURPOSE This study designed to clarify the correlation between Pink1/Parkin-mediated mitophagy and angiogenesis after stroke, and to elucidate the role of SIRT3 in regulating mitophagy and angiogenesis, and to address the mechanism of AFPR on promoting mitophagy and angiogenesis in microvessels endothelium of ischemic brain. STUDY DESIGN A cerebral ischemia/reperfusion (CIR) rat model was developed by middle cerebral artery occlusion procedure. bEnd.3 cells were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) to mimic CIR process. Neurological function, mitophagy and angiogenesis related indicators were measured. SIRT3 siRNA and 3-MA were used to verify the interaction between SIRT3-mediated mitophagy and angiogenesis. METHODS CIR rats were orally treated with AFPR (8 and 4 g raw drug /kg) and Nimodipine (10.8 mg/kg) for 12 days to mimic the recovery phase post-stroke. The neurological function assessment, TTC staining, HE staining, TUNEL staining and Nissl staining were performed to assess neuroprotective effects of AFPR against CIR. Then CD31-labeled microvessel density in brain was visualized and quantified by immunofluorescence staining. Mitochondrial ultrastructure was assessed by transmission electron microscope scanning. Expressions of relative proteins,e.g. SIRT3, Pink1, Parkin, LC3-II, p62, VEGFA, involving in mitophagy and angiogenesis, were detected by Western blotting analysis. In vitro, bEnd.3 cells were cultured with AFPR or in combination of autophagy inhibitor 3-MA during the reoxygenation. Then cell viability, and LDH releasing were measured. Angiogenic indicators,such as migration and tube formation activity, VEGFA level were determined. To assess effects of AFPR on mitophagy, mitophagy-related proteins were detected, as well as the autophagosome engulfment and lysosome degradation of mitochondria. To address the role of SIRT3, deacetylation activity of SIRT3 was validated by detecting acetylated FOXO3A level with co-immunoprecipitation (Co-IP) assay. Pre-treatment of siRNA or combination use of 3-MA were used to verify the detailed mechanism. RESULTS AFPR remarkably reduced neurological scores and infarct size, alleviated neuron apoptosis in cortex, and increased Nissl density in hippocampus of CIR rats. In addition, AFPR significantly promoted angiogenesis by increasing microvessels density and VEGFA expressions, increased SIRT3 expression, and activated Pink1/Parkin mediated mitophagy. In bEnd.3 cells, the combination use of 3-MA and AFPR further demonstrated that AFPR might promote angiogenesis after OGD/R injury through activating Pink1/Parkin mediated mitophagy. Co-IP assay suggested AFPR reduced acetylated FOXO3A level. This might be correlated with an elevation of SIRT3 expression and it's deacetylation activity. SIRT3 siRNA pretreatment significantly abolished the activation of mitophagy through Pink1/Parkin axis, eventually inhibited angiogenesis. CONCLUSION AFPR promoted angiogenesis through activating mitophagy after cerebral ischemia reperfusion, which might partially involved in the amelioration of SIRT3-mediated regulation on Pink1/Parkin axis. Our study will shed new light on the role of SIRT3 in ischemic brain, especially in regulating mitophagy and angiogenesis after stroke.
Collapse
Affiliation(s)
- Jie Wei
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China
| | - Jiaxiu Xie
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China
| | - Junhui He
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China
| | - Dongmei Li
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Nanning, 530022, China
| | - Dongmei Wei
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China
| | - Yi Li
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guining Wei
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China.
| | - Kedao Lai
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, China.
| |
Collapse
|
10
|
Wu S, Zhang J, Peng C, Ma Y, Tian X. SIRT6 mediated histone H3K9ac deacetylation involves myocardial remodelling through regulating myocardial energy metabolism in TAC mice. J Cell Mol Med 2023; 27:3451-3464. [PMID: 37603612 PMCID: PMC10660608 DOI: 10.1111/jcmm.17915] [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: 03/19/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023] Open
Abstract
Pathological myocardial remodelling is the initial factor of chronic heart failure (CHF) and is induced by multiple factors. We previously demonstrated that histone acetylation is involved in CHF in transverse aortic constriction (TAC) mice, a model for pressure overload-induced heart failure. In this study, we investigated whether the histone deacetylase Sirtuin 6 (SIRT6), which mediates deacetylation of histone 3 acetylated at lysine 9 (H3K9ac), is involved pathological myocardial remodelling by regulating myocardial energy metabolism and explored the underlying mechanisms. We generated a TAC mouse model by partial thoracic aortic banding. TAC mice were injected with the SIRT6 agonist MDL-800 at a dose of 65 mg/kg for 8 weeks. At 4, 8 and 12 weeks after TAC, the level of H3K9ac increased gradually, while the expression of SIRT6 and vascular endothelial growth factor A (VEGFA) decreased gradually. MDL-800 reversed the effects of SIRT6 on H3K9ac in TAC mice and promoted the expression of VEGFA in the hearts of TAC mice. MDL-800 also attenuated mitochondria damage and improved mitochondrial respiratory function through upregulating SIRT6 in the hearts of TAC mice. These results revealed a novel mechanism in which SIRT6-mediated H3K9ac level is involved pathological myocardial remodelling in TAC mice through regulating myocardial energy metabolism. These findings may assist in the development of novel methods for preventing and treating pathological myocardial remodelling.
Collapse
Affiliation(s)
- Shuqi Wu
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Jiaojiao Zhang
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Chang Peng
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yixiang Ma
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xiaochun Tian
- Department of Pediatrics, Guizhou Children's HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
11
|
Zhang G, Liu B, Yang Y, Xie S, Chen L, Luo H, Zhong J, Wei Y, Guo F, Gan J, Zhu F, Xu L, Li Q, Shen Y, Zhang H, Liu Y, Li R, Deng H, Yang H. Mitochondrial UQCC3 controls embryonic and tumor angiogenesis by regulating VEGF expression. iScience 2023; 26:107370. [PMID: 37539028 PMCID: PMC10393800 DOI: 10.1016/j.isci.2023.107370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/05/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023] Open
Abstract
Mitochondria play important roles in angiogenesis. However, the mechanisms remain elusive. In this study, we found that mitochondrial ubiquinol-cytochrome c reductase complex assembly factor 3 (UQCC3) is a key regulator of angiogenesis. TALEN-mediated knockout of Uqcc3 in mice caused embryonic lethality at 9.5-10.5 days postcoitum, and vessel density was dramatically reduced. Similarly, knockout of uqcc3 in zebrafish induced lethality post-fertilization and impaired vascular development. Knockout of UQCC3 resulted in slower tumor growth and angiogenesis. Mechanistically, UQCC3 was upregulated under hypoxia, promoted reactive oxygen species (ROS) generation, enhanced HIF-1α stability and increased VEGF expression. Finally, higher expression of UQCC3 was associated with poor prognosis in multiple types tumors, implying a role for UQCC3 in tumor progression. In conclusion, our findings highlight the important contribution of UQCC3 to angiogenesis under both physiological and pathological conditions, indicating the potential of UQCC3 as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Guimin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Binrui Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yun Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuo Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lingcheng Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Zhong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinhao Wei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fengzhu Guo
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jia Gan
- Department of Pathology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R.China
| | - Fan Zhu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiqi Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuge Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huajin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongxin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanshuo Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Le NT. Metabolic regulation of endothelial senescence. Front Cardiovasc Med 2023; 10:1232681. [PMID: 37649668 PMCID: PMC10464912 DOI: 10.3389/fcvm.2023.1232681] [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: 05/31/2023] [Accepted: 07/18/2023] [Indexed: 09/01/2023] Open
Abstract
Endothelial cell (EC) senescence is increasingly recognized as a significant contributor to the development of vascular dysfunction and age-related disorders and diseases, including cancer and cardiovascular diseases (CVD). The regulation of cellular senescence is known to be influenced by cellular metabolism. While extensive research has been conducted on the metabolic regulation of senescence in other cells such as cancer cells and fibroblasts, our understanding of the metabolic regulation of EC senescence remains limited. The specific metabolic changes that drive EC senescence are yet to be fully elucidated. The objective of this review is to provide an overview of the intricate interplay between cellular metabolism and senescence, with a particular emphasis on recent advancements in understanding the metabolic changes preceding cellular senescence. I will summarize the current knowledge on the metabolic regulation of EC senescence, aiming to offer insights into the underlying mechanisms and future research directions.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
13
|
Novorolsky RJ, Kasheke GDS, Hakim A, Foldvari M, Dorighello GG, Sekler I, Vuligonda V, Sanders ME, Renden RB, Wilson JJ, Robertson GS. Preserving and enhancing mitochondrial function after stroke to protect and repair the neurovascular unit: novel opportunities for nanoparticle-based drug delivery. Front Cell Neurosci 2023; 17:1226630. [PMID: 37484823 PMCID: PMC10360135 DOI: 10.3389/fncel.2023.1226630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glia, and neurons that form the basic component of the blood brain barrier. This intricate structure rapidly adjusts cerebral blood flow to match the metabolic needs of brain activity. However, the NVU is exquisitely sensitive to damage and displays limited repair after a stroke. To effectively treat stroke, it is therefore considered crucial to both protect and repair the NVU. Mitochondrial calcium (Ca2+) uptake supports NVU function by buffering Ca2+ and stimulating energy production. However, excessive mitochondrial Ca2+ uptake causes toxic mitochondrial Ca2+ overloading that triggers numerous cell death pathways which destroy the NVU. Mitochondrial damage is one of the earliest pathological events in stroke. Drugs that preserve mitochondrial integrity and function should therefore confer profound NVU protection by blocking the initiation of numerous injury events. We have shown that mitochondrial Ca2+ uptake and efflux in the brain are mediated by the mitochondrial Ca2+ uniporter complex (MCUcx) and sodium/Ca2+/lithium exchanger (NCLX), respectively. Moreover, our recent pharmacological studies have demonstrated that MCUcx inhibition and NCLX activation suppress ischemic and excitotoxic neuronal cell death by blocking mitochondrial Ca2+ overloading. These findings suggest that combining MCUcx inhibition with NCLX activation should markedly protect the NVU. In terms of promoting NVU repair, nuclear hormone receptor activation is a promising approach. Retinoid X receptor (RXR) and thyroid hormone receptor (TR) agonists activate complementary transcriptional programs that stimulate mitochondrial biogenesis, suppress inflammation, and enhance the production of new vascular cells, glia, and neurons. RXR and TR agonism should thus further improve the clinical benefits of MCUcx inhibition and NCLX activation by increasing NVU repair. However, drugs that either inhibit the MCUcx, or stimulate the NCLX, or activate the RXR or TR, suffer from adverse effects caused by undesired actions on healthy tissues. To overcome this problem, we describe the use of nanoparticle drug formulations that preferentially target metabolically compromised and damaged NVUs after an ischemic or hemorrhagic stroke. These nanoparticle-based approaches have the potential to improve clinical safety and efficacy by maximizing drug delivery to diseased NVUs and minimizing drug exposure in healthy brain and peripheral tissues.
Collapse
Affiliation(s)
- Robyn J. Novorolsky
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Gracious D. S. Kasheke
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Antoine Hakim
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Marianna Foldvari
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Gabriel G. Dorighello
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben Gurion University, Beersheva, Israel
| | | | | | - Robert B. Renden
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, College of Arts and Sciences, Cornell University, Ithaca, NY, United States
| | - George S. Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
14
|
Ma ZR, Li HP, Cai SZ, Du SY, Chen X, Yao J, Cao X, Zhen YF, Wang Q. The mitochondrial protein TIMM44 is required for angiogenesis in vitro and in vivo. Cell Death Dis 2023; 14:307. [PMID: 37147302 PMCID: PMC10163060 DOI: 10.1038/s41419-023-05826-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
The mitochondrial integrity and function in endothelial cells are essential for angiogenesis. TIMM44 (translocase of inner mitochondrial membrane 44) is essential for integrity and function of mitochondria. Here we explored the potential function and the possible mechanisms of TIMM44 in angiogenesis. In HUVECs, human retinal microvascular endothelial cells and hCMEC/D3 brain endothelial cells, silence of TIMM44 by targeted shRNA largely inhibited cell proliferation, migration and in vitro capillary tube formation. TIMM44 silencing disrupted mitochondrial functions in endothelial cells, causing mitochondrial protein input arrest, ATP reduction, ROS production, and mitochondrial depolarization, and leading to apoptosis activation. TIMM44 knockout, by Cas9-sgRNA strategy, also disrupted mitochondrial functions and inhibited endothelial cell proliferation, migration and in vitro capillary tube formation. Moreover, treatment with MB-10 ("MitoBloCK-10"), a TIMM44 blocker, similarly induced mitochondrial dysfunction and suppressed angiogenic activity in endothelial cells. Contrarily, ectopic overexpression of TIMM44 increased ATP contents and augmented endothelial cell proliferation, migration and in vitro capillary tube formation. In adult mouse retinas, endothelial knockdown of TIMM44, by intravitreous injection of endothelial specific TIMM44 shRNA adenovirus, inhibited retinal angiogenesis, causing vascular leakage, acellular capillary growth, and retinal ganglion cells degeneration. Significant oxidative stress was detected in TIMM44-silenced retinal tissues. Moreover, intravitreous injection of MB-10 similarly induced oxidative injury and inhibited retinal angiogenesis in vivo. Together, the mitochondrial protein TIMM44 is important for angiogenesis in vitro and in vivo, representing as a novel and promising therapeutic target of diseases with abnormal angiogenesis.
Collapse
Affiliation(s)
- Zhou-Rui Ma
- Department of Burns and Plastic Surgery, Children's hospital of Soochow University, Suzhou, China
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China
| | - Hong-Peng Li
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Shi-Zhong Cai
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, China
| | - Sheng-Yang Du
- Department of Orthopedics, Xuzhou First People's Hospital, Xuzhou, China
| | - Xia Chen
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, China
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China.
| | - Xu Cao
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China.
- Department of Urology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yun-Fang Zhen
- Department of Orthopedics, Children's hospital of Soochow University, Suzhou, China.
| | - Qian Wang
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, China.
| |
Collapse
|
15
|
Zhang Z, Zhou M, Liu H, Liu W, Chen J. Protective effects of Shen Yuan Dan on myocardial ischemia-reperfusion injury via the regulation of mitochondrial quality control. Cardiovasc Diagn Ther 2023; 13:395-407. [PMID: 37583687 PMCID: PMC10423729 DOI: 10.21037/cdt-23-86] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/19/2023] [Indexed: 08/17/2023]
Abstract
Background Myocardial cell death resulting from ischemia-reperfusion (I/R) injury has been a predominant contributor to morbidity and mortality globally. The mitochondria-centered mechanism plays an important role in the formation of I/R injury. This study intended to discuss the protective mechanism of Shen Yuan Dan (SYD) on cardiomyocytes hypoxia-reoxygenation (H/R) injury via the regulation of mitochondrial quality control (MQC). Additionally, this study clarified the mechanism by which SYD suppressed mitophagy activity through the suppression of the PTEN-induced kinase 1 (PINK1)/Parkin pathway. Methods To induce cellular injury, H9c2 cardiomyocytes were exposed to H/R stimulation. Following the pretreatment with SYD, cardiomyocytes were subjected to H/R stimulation. Mitochondrial membrane potential (MMP), adenosine triphosphate (ATP), superoxide dismutase (SOD), and methane dicarboxylic aldehyde (MDA) were detected to evaluate the degree of cardiomyocyte mitochondrial damage. Laser confocal microscopy was applied to observe the mitochondrial quality, and the messenger (mRNA) levels of mitofusin 1 (Mfn1), mitofusin 2 (Mfn2), optic atrophy protein 1 (Opa1), dynamin-related protein 1 (Drp1), fission 1 (Fis1), and peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) in cardiomyocytes were assessed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Western blotting was employed for the estimation of light chain 3 (LC3)-I, LC3-II, PINK1, and Parkin in cardiomyocytes. Results It was discovered that SYD pretreatment elevated MMP in H/R injury cardiomyocytes, enhanced ATP content, activated SOD activity, and reduced MDA level. SYD treatment increased the mRNA levels of Mfn1, Mfn2, Opa1 and PGC-1α decreased the mRNA levels of Drp1 and Fis1, and reduced the protein levels of LC3, PINK1, and Parkin. Conclusions SYD plays a protective role in H/R injury to cardiomyocytes by regulating mitochondrial quality. Meanwhile, SYD may inhibit mitophagy activity through inhibiting the PINK1/Parkin pathway. This study provides insights into the underlying mechanism of SYD in alleviating myocardial I/R injury.
Collapse
Affiliation(s)
- Zhuhua Zhang
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Affiliated to the Capital Medical University, Beijing, China
| | - Mingxue Zhou
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Affiliated to the Capital Medical University, Beijing, China
| | - Hongxu Liu
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Affiliated to the Capital Medical University, Beijing, China
| | - Wei Liu
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Affiliated to the Capital Medical University, Beijing, China
| | - Jiaping Chen
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Affiliated to the Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Wang Y, Wu J, Wang J, He L, Lai H, Zhang T, Wang X, Li W. Mitochondrial oxidative stress in brain microvascular endothelial cells: Triggering blood-brain barrier disruption. Mitochondrion 2023; 69:71-82. [PMID: 36709855 DOI: 10.1016/j.mito.2023.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Blood-brain barrier disruption plays an important role in central nervous system diseases. This review provides information on the role of mitochondrial oxidative stress in brain microvascular endothelial cells in cellular dysfunction, the disruption of intercellular junctions, transporter dysfunction, abnormal angiogenesis, neurovascular decoupling, and the involvement and aggravation of vascular inflammation and illustrates related molecular mechanisms. In addition, recent drug and nondrug therapies targeting cerebral vascular endothelial cell mitochondria to repair the blood-brain barrier are discussed. This review shows that mitochondrial oxidative stress disorder in brain microvascular endothelial cells plays a key role in the occurrence and development of blood-brain barrier damage and may be critical in various pathological mechanisms of blood-brain barrier damage. These new findings suggest a potential new strategy for the treatment of central nervous system diseases through mitochondrial modulation of cerebral vascular endothelial cells.
Collapse
Affiliation(s)
- Yi Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jing Wu
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jiexin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Linxi He
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Han Lai
- School of Foreign Languages, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Tian Zhang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Xin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| |
Collapse
|
17
|
Zhang X, Sun J, Zhou M, Li C, Zhu Z, Gan X. The role of mitochondria in the peri-implant microenvironment. Exp Physiol 2023; 108:398-411. [PMID: 36648334 PMCID: PMC10103875 DOI: 10.1113/ep090988] [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: 11/13/2022] [Accepted: 12/12/2022] [Indexed: 01/18/2023]
Abstract
NEW FINDINGS What is the topic of this review? In this review, we consider the key role of mitochondria in the peri-implant milieu, including the regulation of mitochondrial reactive oxygen species and mitochondrial metabolism in angiogenesis, the polarization of macrophage immune responses, and bone formation and bone resorption during osseointegration. What advances does it highlight? Mitochondria contribute to the behaviours of peri-implant cell lines based on metabolic and reactive oxygen species signalling modulations, which will contribute to the research field and the development of new treatment strategies for improving implant success. ABSTRACT Osseointegration is a dynamic biological process in the local microenvironment adjacent to a bone implant, which is crucial for implant performance and success of the implant surgery. Recently, the role of mitochondria in the peri-implant microenvironment during osseointegration has gained much attention. Mitochondrial regulation has been verified to be essential for cellular events in osseointegration and as a therapeutic target for peri-implant diseases in the peri-implant microenvironment. In this review, we summarize our current knowledge of the key role of mitochondria in the peri-implant milieu, including the regulation of mitochondrial reactive oxygen species and mitochondrial metabolism in angiogenesis, the polarization of macrophage immune responses, and bone formation and resorption during osseointegration, which will contribute to the research field and the development of new treatment strategies to improve implant success. In addition, we indicate limitations in our current understanding of the regulation of mitochondria in osseointegration and suggest topics for further study.
Collapse
Affiliation(s)
- Xidan Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiyu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Min Zhou
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Chen Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Zhuoli Zhu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xueqi Gan
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
18
|
Jing W, Liu C, Su C, Liu L, Chen P, Li X, Zhang X, Yuan B, Wang H, Du X. Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Front Immunol 2023; 14:1107670. [PMID: 36845127 PMCID: PMC9948260 DOI: 10.3389/fimmu.2023.1107670] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, pannus formation, and bone and cartilage damage. It has a high disability rate. The hypoxic microenvironment of RA joints can cause reactive oxygen species (ROS) accumulation and mitochondrial damage, which not only affect the metabolic processes of immune cells and pathological changes in fibroblastic synovial cells but also upregulate the expression of several inflammatory pathways, ultimately promoting inflammation. Additionally, ROS and mitochondrial damage are involved in angiogenesis and bone destruction, thereby accelerating RA progression. In this review, we highlighted the effects of ROS accumulation and mitochondrial damage on inflammatory response, angiogenesis, bone and cartilage damage in RA. Additionally, we summarized therapies that target ROS or mitochondria to relieve RA symptoms and discuss the gaps in research and existing controversies, hoping to provide new ideas for research in this area and insights for targeted drug development in RA.
Collapse
Affiliation(s)
- Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chenghong Su
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangjun Li
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
19
|
Bai G, Jiang X, Qin J, Zou Y, Zhang W, Teng T, Shi B, Sun H. Perinatal exposure to glyphosate-based herbicides impairs progeny health and placental angiogenesis by disturbing mitochondrial function. ENVIRONMENT INTERNATIONAL 2022; 170:107579. [PMID: 36265358 DOI: 10.1016/j.envint.2022.107579] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Glyphosate-based herbicides (GBHs) are the most widely used pesticide worldwide and can provoke placental injury. However, whether and how GBHs damage angiogenesis in the placenta is not yet known. This work evaluated the safety of glyphosate on pregnant sows based on the limit level by governments and investigated the effects and mechanism of Low-GBHs (20 mg/kg) and High-GBHs (100 mg/kg) exposure on placental angiogenesis. Results showed that gestational exposure to GBHs decreased placental vessel density and cell multiplication by interfering with the expression of VEGFA, PLGF, VEGFr2 and Hand2 (indicators of angiogenesis), which may be in relation to oxidative stress-induced disorders of mitochondrial fission and fusion as well as the impaired function of the mitochondrial respiratory chain. Additionally, GBHs destroyed barrier function and nutrient transport in the placenta, and was accompanied by jejunum oxidative stress in newborn piglets. However, GBHs exposure had no significant differences on sow reproductive performance. As a natural antioxidant, betaine treatment protected placenta and newborn piglets against GBHs-induced damage. In conclusion, GBHs impaired placental angiogenesis and function and further damaged the health of postnatal progeny, these effects may be linked to mitochondrial dysfunction. Betaine treatment following glyphosate exposure provided modest relief.
Collapse
Affiliation(s)
- Guangdong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Xu Jiang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Jianwei Qin
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Yingbin Zou
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Wentao Zhang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Teng Teng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China.
| | - Haoyang Sun
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
20
|
Mitochondria-Targeted Delivery of Camptothecin Based on HPMA Copolymer for Metastasis Suppression. Pharmaceutics 2022; 14:pharmaceutics14081534. [PMID: 35893790 PMCID: PMC9331251 DOI: 10.3390/pharmaceutics14081534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/10/2022] [Accepted: 07/20/2022] [Indexed: 01/02/2023] Open
Abstract
Poor anti-metastasis effects and side-effects remain a challenge for the clinical application of camptothecin (CPT). Mitochondria can be a promising target for the treatment of metastatic tumors due to their vital roles in providing energy supply, upregulating pro-metastatic factors, and controlling cell-death signaling. Thus, selectively delivering CPT to mitochondria appears to be a feasible way of improving the anti-metastasis effect and reducing adverse effects. Here, we established a 2-(dimethylamino) ethyl methacrylate (DEA)-modified N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer–CPT conjugate (P-DEA-CPT) to mediate the mitochondrial accumulation of CPT. The mitochondria-targeted P-DEA-CPT could overcome multiple barriers by quickly internalizing into 4T1 cells, then escaping from lysosome, and sufficiently accumulating in mitochondria. Subsequently, P-DEA-CPT greatly damaged mitochondrial function, leading to the reactive oxide species (ROS) elevation, energy depletion, apoptosis amplification, and tumor metastasis suppression. Consequently, P-DEA-CPT successfully inhibited both primary tumor growth and distant metastasis in vivo. Furthermore, our studies revealed that the mechanism underlying the anti-metastasis capacity of P-DEA-CPT was partially via downregulation of various pro-metastatic proteins, such as hypoxia induction factor-1α (HIF-1α), matrix metalloproteinases-2 (MMP-2), and vascular endothelial growth factor (VEGF). This study provided the proof of concept that escorting CPT to mitochondria via a mitochondrial targeting strategy could be a promising approach for anti-metastasis treatment.
Collapse
|
21
|
Abstract
Mice with insulin receptor (IR)-deficient astrocytes (GFAP-IR knockout [KO] mice) show blunted responses to insulin and reduced brain glucose uptake, whereas IR-deficient astrocytes show disturbed mitochondrial responses to glucose. While exploring the functional impact of disturbed mitochondrial function in astrocytes, we observed that GFAP-IR KO mice show uncoupling of brain blood flow with glucose uptake. Since IR-deficient astrocytes show higher levels of reactive oxidant species (ROS), this leads to stimulation of hypoxia-inducible factor-1α and, consequently, of the vascular endothelial growth factor angiogenic pathway. Indeed, GFAP-IR KO mice show disturbed brain vascularity and blood flow that is normalized by treatment with the antioxidant N-acetylcysteine (NAC). NAC ameliorated high ROS levels, normalized angiogenic signaling and mitochondrial function in IR-deficient astrocytes, and normalized neurovascular coupling in GFAP-IR KO mice. Our results indicate that by modulating glucose uptake and angiogenesis, insulin receptors in astrocytes participate in neurovascular coupling.
Collapse
|
22
|
Mitochondrial mutations alter endurance exercise response and determinants in mice. Proc Natl Acad Sci U S A 2022; 119:e2200549119. [PMID: 35482926 PMCID: PMC9170171 DOI: 10.1073/pnas.2200549119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Primary mitochondrial diseases (PMDs) are the most prevalent inborn metabolic disorders, affecting an estimated 1 in 4,200 individuals. Endurance exercise is generally known to improve mitochondrial function, but its indication in the heterogeneous group of PMDs is unclear. We determined the relationship between mitochondrial mutations, endurance exercise response, and the underlying molecular pathways in mice with distinct mitochondrial mutations. This revealed that mitochondria are crucial regulators of exercise capacity and exercise response. Endurance exercise proved to be mostly beneficial across the different mitochondrial mutant mice with the exception of a worsened dilated cardiomyopathy in ANT1-deficient mice. Thus, therapeutic exercises, especially in patients with PMDs, should take into account the physical and mitochondrial genetic status of the patient. Primary mitochondrial diseases (PMDs) are a heterogeneous group of metabolic disorders that can be caused by hundreds of mutations in both mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) genes. Current therapeutic approaches are limited, although one approach has been exercise training. Endurance exercise is known to improve mitochondrial function in heathy subjects and reduce risk for secondary metabolic disorders such as diabetes or neurodegenerative disorders. However, in PMDs the benefit of endurance exercise is unclear, and exercise might be beneficial for some mitochondrial disorders but contraindicated in others. Here we investigate the effect of an endurance exercise regimen in mouse models for PMDs harboring distinct mitochondrial mutations. We show that while an mtDNA ND6 mutation in complex I demonstrated improvement in response to exercise, mice with a CO1 mutation affecting complex IV showed significantly fewer positive effects, and mice with an ND5 complex I mutation did not respond to exercise at all. For mice deficient in the nDNA adenine nucleotide translocase 1 (Ant1), endurance exercise actually worsened the dilated cardiomyopathy. Correlating the gene expression profile of skeletal muscle and heart with the physiologic exercise response identified oxidative phosphorylation, amino acid metabolism, matrisome (extracellular matrix [ECM]) structure, and cell cycle regulation as key pathways in the exercise response. This emphasizes the crucial role of mitochondria in determining the exercise capacity and exercise response. Consequently, the benefit of endurance exercise in PMDs strongly depends on the underlying mutation, although our results suggest a general beneficial effect.
Collapse
|
23
|
Zaccagnini G, Greco S, Voellenkle C, Gaetano C, Martelli F. miR-210 hypoxamiR in Angiogenesis and Diabetes. Antioxid Redox Signal 2022; 36:685-706. [PMID: 34521246 DOI: 10.1089/ars.2021.0200] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: microRNA-210 (miR-210) is the master hypoxia-inducible miRNA (hypoxamiR) since it has been found to be significantly upregulated under hypoxia in a wide range of cell types. Recent advances: Gene ontology analysis of its targets indicates that miR-210 modulates several aspects of cellular response to hypoxia. Due to its high pleiotropy, miR-210 not only plays a protective role by fine-tuning mitochondrial metabolism and inhibiting red-ox imbalance and apoptosis, but it can also promote cell proliferation, differentiation, and migration, substantially contributing to angiogenesis. Critical issues: As most miRNAs, modulating different gene pathways, also miR-210 can potentially lead to different and even opposite effects, depending on the physio-pathological contexts in which it acts. Future direction: The use of miRNAs as therapeutics is a fast growing field. This review aimed at highlighting the role of miR-210 in angiogenesis in the context of ischemic cardiovascular diseases and diabetes in order to clarify the molecular mechanisms underpinning miR-210 action. Particular attention will be dedicated to experimentally validated miR-210 direct targets involved in cellular processes related to angiogenesis and diabetes mellitus, such as mitochondrial metabolism, redox balance, apoptosis, migration, and adhesion. Antioxid. Redox Signal. 36, 685-706.
Collapse
Affiliation(s)
- Germana Zaccagnini
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| |
Collapse
|
24
|
Wikramanayake TC, Chéret J, Sevilla A, Birch-Machin M, Paus R. Targeting mitochondria in dermatological therapy: Beyond oxidative damage and skin aging. Expert Opin Ther Targets 2022; 26:233-259. [PMID: 35249436 DOI: 10.1080/14728222.2022.2049756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The analysis of the role of the mitochondria in oxidative damage and skin aging is a significant aspect of dermatological research. Mitochondria generate most reactive oxygen species (ROS); however, excessive ROS are cytotoxic and DNA-damaging and promote (photo-)aging. ROS also possesses key physiological and regulatory functions and mitochondrial dysfunction is prominent in several skin diseases including skin cancers. Although many standard dermatotherapeutics modulate mitochondrial function, dermatological therapy rarely targets the mitochondria. Accordingly, there is a rationale for "mitochondrial dermatology"-based approaches to be applied to therapeutic research. AREAS COVERED This paper examines the functions of mitochondria in cutaneous physiology beyond energy (ATP) and ROS production. Keratinocyte differentiation and epidermal barrier maintenance, appendage morphogenesis and homeostasis, photoaging and skin cancer are considered. Based on related PubMed search results, the paper evaluates thyroid hormones, glucocorticoids, Vitamin D3 derivatives, retinoids, cannabinoid receptor agonists, PPARγ agonists, thyrotropin, and thyrotropin-releasing hormone as instructive lead compounds. Moreover, the mitochondrial protein MPZL3 as a promising new drug target for future "mitochondrial dermatology" is highlighted. EXPERT OPINION Future dermatological therapeutic research should have a mitochondrial medicine emphasis. Focusing on selected lead agents, protein targets, in silico drug design, and model diseases will fertilize a mito-centric approach.
Collapse
Affiliation(s)
- Tongyu C Wikramanayake
- Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, U.S.A.,Molecular Cell and Developmental Biology Program, University of Miami Miller School of Medicine, Miami, FL, U.S.A
| | - Jérémy Chéret
- Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, U.S.A
| | - Alec Sevilla
- Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, U.S.A
| | - Mark Birch-Machin
- Dermatological Sciences, Translational and Clinical Research Institute, and The UK National Innovation Centre for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Ralf Paus
- Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, U.S.A.,Monasterium Laboratory, Münster, Germany.,Centre for Dermatology Research, University of Manchester, and NIHR Manchester Biomedical Research Centre, Manchester, UK
| |
Collapse
|
25
|
Vera DB, Fredes AN, Garrido MP, Romero C. Role of Mitochondria in Interplay between NGF/TRKA, miR-145 and Possible Therapeutic Strategies for Epithelial Ovarian Cancer. LIFE (BASEL, SWITZERLAND) 2021; 12:life12010008. [PMID: 35054401 PMCID: PMC8779980 DOI: 10.3390/life12010008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
Ovarian cancer is the most lethal gynecological neoplasm, and epithelial ovarian cancer (EOC) accounts for 90% of ovarian malignancies. The 5-year survival is less than 45%, and, unlike other types of cancer, the proportion of women who die from this disease has not improved in recent decades. Nerve growth factor (NGF) and tropomyosin kinase A (TRKA), its high-affinity receptor, play a crucial role in pathogenesis through cell proliferation, angiogenesis, invasion, and migration. NGF/TRKA increase their expression during the progression of EOC by upregulation of oncogenic proteins as vascular endothelial growth factor (VEGF) and c-Myc. Otherwise, the expression of most oncoproteins is regulated by microRNAs (miRs). Our laboratory group reported that the tumoral effect of NGF/TRKA depends on the regulation of miR-145 levels in EOC. Currently, mitochondria have been proposed as new therapeutic targets to activate the apoptotic pathway in the cancer cell. The mitochondria are involved in a myriad of functions as energy production, redox control, homeostasis of Ca+2, and cell death. We demonstrated that NGF stimulation produces an augment in the Bcl-2/BAX ratio, which supports the anti-apoptotic effects of NGF in EOC cells. The review aimed to discuss the role of mitochondria in the interplay between NGF/TRKA and miR-145 and possible therapeutic strategies that may decrease mortality due to EOC.
Collapse
Affiliation(s)
- Daniela B. Vera
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
| | - Allison N. Fredes
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
| | - Maritza P. Garrido
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
- Obstetrics and Gynecology Departament, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.P.G.); (C.R.)
| | - Carmen Romero
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital University of Chile, Santiago 8380456, Chile; (D.B.V.); (A.N.F.)
- Obstetrics and Gynecology Departament, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.P.G.); (C.R.)
| |
Collapse
|
26
|
Su CT, Jao TM, Urban Z, Huang YJ, See DHW, Tsai YC, Lin WC, Huang JW. LTBP4 affects renal fibrosis by influencing angiogenesis and altering mitochondrial structure. Cell Death Dis 2021; 12:943. [PMID: 34645813 PMCID: PMC8514500 DOI: 10.1038/s41419-021-04214-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/25/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022]
Abstract
Transforming growth factor beta (TGFβ) signalling regulates extracellular matrix accumulation known to be essential for the pathogenesis of renal fibrosis; latent transforming growth factor beta binding protein 4 (LTBP4) is an important regulator of TGFβ activity. To date, the regulation of LTBP4 in renal fibrosis remains unknown. Herein, we report that LTBP4 is upregulated in patients with chronic kidney disease and fibrotic mice kidneys created by unilateral ureteral obstruction (UUO). Mice lacking the short LTBP4 isoform (Ltbp4S-/-) exhibited aggravated tubular interstitial fibrosis (TIF) after UUO, indicating that LTBP4 potentially protects against TIF. Transcriptomic analysis of human proximal tubule cells overexpressing LTBP4 revealed that LTBP4 influences angiogenic pathways; moreover, these cells preserved better mitochondrial respiratory functions and expressed higher vascular endothelial growth factor A (VEGFA) compared to wild-type cells under hypoxia. Results of the tube formation assay revealed that additional LTBP4 in human umbilical vein endothelial cell supernatant stimulates angiogenesis with upregulated vascular endothelial growth factor receptors (VEGFRs). In vivo, aberrant angiogenesis, abnormal mitochondrial morphology and enhanced oxidative stress were observed in Ltbp4S-/- mice after UUO. These results reveal novel molecular functions of LTBP4 stimulating angiogenesis and potentially impacting mitochondrial structure and function. Collectively, our findings indicate that LTBP4 protects against disease progression and may be of therapeutic use in renal fibrosis.
Collapse
Affiliation(s)
- Chi-Ting Su
- Renal Division, Department of Internal medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, National Taiwan University Cancer Centre Hospital, Taipei, Taiwan
| | - Tzu-Ming Jao
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
- Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Zsolt Urban
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yue-Jhu Huang
- Renal Division, Department of Internal medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Daniel H W See
- Renal Division, Department of Internal medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Yao-Chou Tsai
- Renal Division, Department of Internal medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Wei-Chou Lin
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jenq-Wen Huang
- Renal Division, Department of Internal medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan.
| |
Collapse
|
27
|
Li B, Chen K, Liu F, Zhang J, Chen X, Chen T, Chen Q, Yao Y, Hu W, Wang L, Wu Y. Developmental Angiogenesis Requires the Mitochondrial Phenylalanyl-tRNA Synthetase. Front Cardiovasc Med 2021; 8:724846. [PMID: 34540921 PMCID: PMC8440837 DOI: 10.3389/fcvm.2021.724846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/04/2021] [Indexed: 12/03/2022] Open
Abstract
Background: Mitochondrial aminoacyl-tRNA synthetases (mtARSs) catalyze the binding of specific amino acids to their cognate tRNAs and play an essential role in the synthesis of proteins encoded by mitochondrial DNA. Defects in mtARSs have been linked to human diseases, but their tissue-specific pathophysiology remains elusive. Here we examined the role of mitochondrial phenylalanyl-tRNA synthetase (FARS2) in developmental angiogenesis and its potential contribution to the pathogenesis of cardiovascular disease. Methods: Morpholinos were injected into fertilized zebrafish ova to establish an in vivo fars2 knock-down model. A visualization of the vasculature was achieved by using Tg (fli1: EGFP)y1 transgenic zebrafish. In addition, small interference RNAs (siRNAs) were transferred into human umbilical vein endothelial cells (HUVECs) to establish an in vitro FARS2 knock-down model. Cell motility, proliferation, and tubulogenesis were determined using scratch-wound CCK8, transwell-based migration, and tube formation assays. In addition, mitochondria- and non-mitochondria-related respiration were evaluated using a Seahorse XF24 analyzer and flow cytometry assays. Analyses of the expression levels of transcripts and proteins were performed using qRT-PCR and western blotting, respectively. Results: The knock-down of fars2 hampered the embryonic development in zebrafish and delayed the formation of the vasculature in Tg (fli1: EGFP)y1 transgenic zebrafish. In addition, the siRNA-mediated knock-down of FARS2 impaired angiogenesis in HUVECs as indicated by decreased cell motility and tube formation capacity. The knock-down of FARS2 also produced variable decreases in mitochondrial- and non-mitochondrial respiration in HUVECs and disrupted the regulatory pathways of angiogenesis in both HUVECs and zebrafish. Conclusion: Our current work offers novel insights into angiogenesis defects and cardiovascular diseases induced by FARS2 deficiency.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Kun Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Medical University, Xi'an, China
| | - Fangfang Liu
- Department of Neurosciences, Air Force Medical University, Xi'an, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Northwest University, Xi'an, China
| | - Xihui Chen
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Tangdong Chen
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Qi Chen
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Yan Yao
- Department of Clinical Medicine, Yan'an University, Yan'an, China
| | - Weihong Hu
- Department of Clinical Medicine, Yan'an University, Yan'an, China
| | - Li Wang
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China.,School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Yuanming Wu
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| |
Collapse
|
28
|
Solly EL, Psaltis PJ, Bursill CA, Tan JTM. The Role of miR-181c in Mechanisms of Diabetes-Impaired Angiogenesis: An Emerging Therapeutic Target for Diabetic Vascular Complications. Front Pharmacol 2021; 12:718679. [PMID: 34483928 PMCID: PMC8414254 DOI: 10.3389/fphar.2021.718679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is estimated to affect up to 700 million people by the year 2045, contributing to an immense health and economic burden. People living with diabetes have a higher risk of developing numerous debilitating vascular complications, leading to an increased need for medical care, a reduced quality of life and increased risk of early death. Current treatments are not satisfactory for many patients who suffer from impaired angiogenesis in response to ischaemia, increasing their risk of ischaemic cardiovascular conditions. These vascular pathologies are characterised by endothelial dysfunction and abnormal angiogenesis, amongst a host of impaired signaling pathways. Therapeutic stimulation of angiogenesis holds promise for the treatment of diabetic vascular complications that stem from impaired ischaemic responses. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis to improve ischaemic complications such as ischaemic heart disease and peripheral artery disease, highlighting the immense unmet need. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis in a clinical setting, highlighting the immense unmet need. MicroRNAs (miRNAs) are emerging as powerful targets for multifaceted diseases including diabetes and cardiovascular disease. This review highlights the potential role of microRNAs as therapeutic targets for rescuing diabetes-impaired angiogenesis, with a specific focus on miR-181c, which we have previously identified as an important angiogenic regulator. Here we summarise the pathways currently known to be regulated by miR-181c, which include the classical angiogenesis pathways that are dysregulated in diabetes, mitochondrial function and axonal guidance, and describe how these relate both directly and indirectly to angiogenesis. The pleiotropic actions of miR-181c across multiple key angiogenic signaling pathways and critical cellular processes highlight its therapeutic potential as a novel target for treating diabetic vascular complications.
Collapse
Affiliation(s)
- Emma L Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
29
|
Chae SA, Son JS, Du M. Prenatal exercise in fetal development: a placental perspective. FEBS J 2021; 289:3058-3071. [PMID: 34449982 DOI: 10.1111/febs.16173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Maternal obesity (MO) and gestational diabetes mellitus (GDM) are common in Western societies, which impair fetal development and predispose offspring to metabolic dysfunction. Placenta is the organ linking the mother to her fetus, and MO suppresses the development of vascular system and expression of nutrient transporters in placenta, thereby affecting fetal development. For maintaining its proper physiological function, placenta is energy demanding, which is met through extensive oxidative phosphorylation. However, the oxidative capacity of placenta is suppressed due to MO and GDM. Recently, several studies showed that physical activity during pregnancy enhances oxidative metabolism and improves placental function, which might be partially mediated by exerkines, referring to cytokines elicited by exercise. In addition, as an endocrine organ, placenta secretes cytokines, termed placentokines, including apelin, superoxide dismutase 3, irisin, and adiponectin, which mediate fetal development and maternal metabolism. Possible molecular mechanisms linking maternal exercise and placentokines to placental and fetal development are further discussed. As an emerging field, up to now, available studies are limited, mostly conducted in rodents. Given the epidemics of obesity and metabolic disorders, as well as the prevalence of maternal sedentary lifestyle, the effects of exercise of pregnant women on placental function and placentokine secretion, as well as their impacts on fetal development, need to be further examined.
Collapse
Affiliation(s)
- Song Ah Chae
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Jun Seok Son
- Laboratory of Perinatal Kinesioepigenetics, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
30
|
Correia Y, Scheel J, Gupta S, Wang K. Placental mitochondrial function as a driver of angiogenesis and placental dysfunction. Biol Chem 2021; 402:887-909. [PMID: 34218539 DOI: 10.1515/hsz-2021-0121] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022]
Abstract
The placenta is a highly vascularized and complex foetal organ that performs various tasks, crucial to a healthy pregnancy. Its dysfunction leads to complications such as stillbirth, preeclampsia, and intrauterine growth restriction. The specific cause of placental dysfunction remains unknown. Recently, the role of mitochondrial function and mitochondrial adaptations in the context of angiogenesis and placental dysfunction is getting more attention. The required energy for placental remodelling, nutrient transport, hormone synthesis, and the reactive oxygen species leads to oxidative stress, stemming from mitochondria. Mitochondria adapt to environmental changes and have been shown to adjust their oxygen and nutrient use to best support placental angiogenesis and foetal development. Angiogenesis is the process by which blood vessels form and is essential for the delivery of nutrients to the body. This process is regulated by different factors, pro-angiogenic factors and anti-angiogenic factors, such as sFlt-1. Increased circulating sFlt-1 levels have been linked to different preeclamptic phenotypes. One of many effects of increased sFlt-1 levels, is the dysregulation of mitochondrial function. This review covers mitochondrial adaptations during placentation, the importance of the anti-angiogenic factor sFlt-1in placental dysfunction and its role in the dysregulation of mitochondrial function.
Collapse
Affiliation(s)
- Yolanda Correia
- Aston Medical School, College of Health & Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Julia Scheel
- Department of Systems Biology and Bioinformatics, University of Rostock, D-18051 Rostock, Germany
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, D-18051 Rostock, Germany
| | - Keqing Wang
- Aston Medical School, College of Health & Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| |
Collapse
|
31
|
Local complement factor H protects kidney endothelial cell structure and function. Kidney Int 2021; 100:824-836. [PMID: 34139209 DOI: 10.1016/j.kint.2021.05.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 05/09/2021] [Accepted: 05/20/2021] [Indexed: 12/25/2022]
Abstract
Factor H (FH) is a critical regulator of the alternative complement pathway and its deficiency or mutation underlie kidney diseases such as dense deposit disease. Since vascular dysfunction is an important facet of kidney disease, maintaining optimal function of the lining endothelial cells is important for vascular health. To investigate the molecular mechanisms that are regulated by FH in endothelial cells, FH deficient and sufficient mouse kidney endothelial cell cultures were established. Endothelial FH deficiency resulted in cytoskeletal remodeling, increased angiogenic potential, loss of cellular layer integrity and increased cell proliferation. FH reconstitution prevented these FH-dependent proliferative changes. Respiratory flux analysis showed reduced basal mitochondrial respiration, ATP production and maximal respiratory capacity in FH deficient endothelial cells, while proton leak remained unaltered. Similar changes were observed in FH deficient human glomerular endothelial cells indicating the translational potential of these studies. Gene expression analysis revealed that the FH-dependent gene changes in mouse kidney endothelial cells include significant upregulation of genes involved in inflammation and the complement system. The transcription factor nuclear factor-kB, that regulates many biological processes, was translocated from the cytoplasm to the nucleus in the absence of FH. Thus, our studies show the functional relevance of intrinsic FH in kidney endothelial cells in man and mouse.
Collapse
|
32
|
Zhou H, He Y, Zhu J, Lin X, Chen J, Shao C, Wan H, Yang J. Guhong Injection Protects Against Apoptosis in Cerebral Ischemia by Maintaining Cerebral Microvasculature and Mitochondrial Integrity Through the PI3K/AKT Pathway. Front Pharmacol 2021; 12:650983. [PMID: 34054531 PMCID: PMC8155598 DOI: 10.3389/fphar.2021.650983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/23/2021] [Indexed: 01/22/2023] Open
Abstract
Guhong injection (GHI) can be used for the treatment of ischemic stroke. We investigated the antiapoptotic activity of GHI, its ability to repair the cerebral microvessels and mitochondria, and the PI3K/AKT signaling pathway of GHI against cerebral ischemia. Western blot and immunohistochemical analyses were used to determine the expression of cleaved caspase-3, B-cell lymphoma-2 (Bcl-2), cytochrome c (Cyt-c), basic fibroblast growth factor (BFGF), vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1), and proteins in the PI3K/AKT signaling pathway. Transmission electron microscopy and scanning electron microscopy were used to evaluate the structures of the cerebral microvasculature and cells. Hoechst 33342 staining was used to evaluate the nuclear morphology. FITC-AV/PI double staining was used to measure the antiapoptotic effects. The fluorescent dye JC-1 was used to measure mitochondrial membrane potential. The enzyme-linked immunosorbent assay (ELISA) was used to detect the activities of matrix metalloproteinase-9 (MMP-9). Biochemical assay kits were used to detect the activities of lactate dehydrogenase (LDH), superoxide dismutase (SOD), and malondialdehyde (MDA). Compared with the middle cerebral artery occlusion (MCAO) group, there was decreased infarct volume and significantly improved neurological deficits in the GHI group. In addition, the expression of Bcl-2 was significantly upregulated, while the expression of Cyt-c, Bax, and cleaved caspase-3 was notably downregulated. GHI administration attenuated the pathological change and morphology of the cerebral microvasculature, and immunohistochemical staining indicated that the expressions of BFGF, VEGF, and TGF-β1 were significantly increased. The cell morphology, cell viability, cell nuclei characteristics, and mitochondrial morphology normalized following GHI treatment, which decreased the release of Cyt-c and the mitochondrial membrane potential. The levels of LDH, MMP-9, and MDA decreased, while SOD increased. Moreover, GHI administration inhibited the activation of the PI3K/AKT signaling pathway in rat brain microvascular endothelial cells (rBMECs) following oxygen/glucose deprivation (OGD) injury. Therefore, our results show that GHI administration resulted in antiapoptosis of cerebral cells and repair of cerebral microvessels and mitochondria via the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Huifen Zhou
- Institute of Cardiovascular-Cranial Disease, School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Zhu
- Institute of Cardiovascular-Cranial Disease, School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaojie Lin
- Institute of Cardiovascular-Cranial Disease, School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Juan Chen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chongyu Shao
- Institute of Cardiovascular-Cranial Disease, School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- Institute of Cardiovascular-Cranial Disease, School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiehong Yang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
33
|
Lopes SV, Collins MN, Reis RL, Oliveira JM, Silva-Correia J. Vascularization Approaches in Tissue Engineering: Recent Developments on Evaluation Tests and Modulation. ACS APPLIED BIO MATERIALS 2021; 4:2941-2956. [DOI: 10.1021/acsabm.1c00051] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Soraia V. Lopes
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maurice N. Collins
- Bernal Institute, School of Engineering, University of Limerick, Limerick V94 T9PX, Ireland
| | - Rui L. Reis
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
34
|
Lin Z, Lin X, Chen J, Huang G, Chen T, Zheng L. Mitofusin-2 is a novel anti-angiogenic factor in pancreatic cancer. J Gastrointest Oncol 2021; 12:484-495. [PMID: 34012642 DOI: 10.21037/jgo-21-176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Aberrant expression of mitofusin-2 (MFN2) has been found to be associated with vascular endothelial growth factor A (VEGFA)-mediated angiogenesis in human umbilical vein endothelial cells (HUVECs). This study aimed to investigate the expression of MFN2 in pancreatic cancer (PC) and the role of MFN2 in vascular endothelial cell growth and angiogenesis. Methods Protein and mRNA expression of MFN2 and VEGFA were measured. The CCK-8 assay, tube formation assay, flow cytometry, and transmission electron microscopy were used to examine the effects of MFN2 overexpression on HUVEC growth, angiogenesis, and apoptosis. Western blot and immunocytochemical staining were conducted to measure alterations in cell cycle and apoptosis regulators and vascular endothelial growth factor receptor 2 (VEGFR2), angiopoietin-1 gene (ANGPT1), and tissue inhibitor of metalloproteinase 1 (TIMP1) expression in HUVECs. Results The results showed that MFN2 levels were significantly decreased in tumor tissues. Contrasting results were observed for VEGFA mRNA levels. MFN2 overexpression inhibited cell growth while promoting the formation of apoptotic bodies in HUVECs. Additionally, MFN2 overexpression enhanced the protein expression of p21 and p27 while attenuating the expression of proliferating cell nuclear antigen, VEGFA, VEGFR2, ANGPT1, and TIPM1 in HUVECs. Conclusions In conclusion, MFN2 expression negatively correlates with VEGFA expression in PC and inhibits endothelial cell growth and angiogenesis.
Collapse
Affiliation(s)
- Zhichuan Lin
- Department of Hepatobiliary Surgery, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou, China
| | - Xiaoyi Lin
- Department of Hepatobiliary Surgery, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou, China
| | - Jinhong Chen
- Department of Hepatobiliary Surgery, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou, China
| | - Guoqiang Huang
- Department of Hepatobiliary Surgery, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou, China
| | - Tangen Chen
- Department of Hepatobiliary Surgery, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou, China
| | - Liling Zheng
- Pediatric Intensive Care Unit, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou, China
| |
Collapse
|
35
|
Bao X, Zhang J, Huang G, Yan J, Xu C, Dou Z, Sun C, Zhang H. The crosstalk between HIFs and mitochondrial dysfunctions in cancer development. Cell Death Dis 2021; 12:215. [PMID: 33637686 PMCID: PMC7910460 DOI: 10.1038/s41419-021-03505-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022]
Abstract
Mitochondria are essential cellular organelles that are involved in regulating cellular energy, metabolism, survival, and proliferation. To some extent, cancer is a genetic and metabolic disease that is closely associated with mitochondrial dysfunction. Hypoxia-inducible factors (HIFs), which are major molecules that respond to hypoxia, play important roles in cancer development by participating in multiple processes, such as metabolism, proliferation, and angiogenesis. The Warburg phenomenon reflects a pseudo-hypoxic state that activates HIF-1α. In addition, a product of the Warburg effect, lactate, also induces HIF-1α. However, Warburg proposed that aerobic glycolysis occurs due to a defect in mitochondria. Moreover, both HIFs and mitochondrial dysfunction can lead to complex reprogramming of energy metabolism, including reduced mitochondrial oxidative metabolism, increased glucose uptake, and enhanced anaerobic glycolysis. Thus, there may be a connection between HIFs and mitochondrial dysfunction. In this review, we systematically discuss the crosstalk between HIFs and mitochondrial dysfunctions in cancer development. Above all, the stability and activity of HIFs are closely influenced by mitochondrial dysfunction related to tricarboxylic acid cycle, electron transport chain components, mitochondrial respiration, and mitochondrial-related proteins. Furthermore, activation of HIFs can lead to mitochondrial dysfunction by affecting multiple mitochondrial functions, including mitochondrial oxidative capacity, biogenesis, apoptosis, fission, and autophagy. In general, the regulation of tumorigenesis and development by HIFs and mitochondrial dysfunction are part of an extensive and cooperative network.
Collapse
Affiliation(s)
- Xingting Bao
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Jinhua Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Guomin Huang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Junfang Yan
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Caipeng Xu
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Zhihui Dou
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Chao Sun
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
| | - Hong Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Guangdong, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
| |
Collapse
|
36
|
Schnabel B, Kuhrt H, Wiedemann P, Bringmann A, Hollborn M. Osmotic regulation of aquaporin-8 expression in retinal pigment epithelial cells in vitro: Dependence on K ATP channel activation. Mol Vis 2020; 26:797-817. [PMID: 33456300 PMCID: PMC7803296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 12/28/2020] [Indexed: 12/02/2022] Open
Abstract
PURPOSE The expression of aquaporin-8 (AQP8), which plays a crucial role in the maintenance of the cellular fluid and electrolyte balance, was shown to be increased in RPE cells under hyperosmotic conditions. The aim of the present study was to investigate the mechanisms of hyperosmotic AQP8 gene expression and the localization of AQP8 in cultured human RPE cells. METHODS Hyperosmolarity was produced with the addition of 100 mM NaCl or 200 mM sucrose. Hypoxia was induced by cell culture in a 0.2% O2 atmosphere or the addition of the hypoxia mimetic CoCl2. Oxidative stress was induced by the addition of H2O2. Gene expression was determined with real-time RT-PCR analysis. AQP8 protein localization and secretion of VEGF were evaluated with immunocytochemistry, western blotting, and enzyme-linked immunosorbent assay (ELISA). RESULTS Immunocytochemical and western blot data suggest that the AQP8 protein is mainly located in the mitochondria. Extracellular hyperosmolarity, hypoxia, and oxidative stress induced increases in AQP8 gene expression. Hyperosmotic AQP8 gene expression was reduced by inhibitors of the p38 MAPK and PI3K signal transduction pathways, and by JAK2 and PLA2 inhibitors, and was in part mediated by the transcriptional activity of CREB. Hyperosmotic AQP8 gene expression was also reduced by autocrine/paracrine interleukin-1 signaling, the sulfonylureas glibenclamide and glipizide, which are known inhibitors of KATP channel activation, and a pannexin-blocking peptide. The KATP channel opener pinacidil increased the expression of AQP8 under control conditions. The cells contained Kir6.1 and SUR2B gene transcripts and displayed Kir6.1 immunoreactivity. siRNA-mediated knockdown of AQP8 caused increases in hypoxic VEGF gene expression and secretion and decreased cell viability under control, hyperosmotic, and hypoxic conditions. CONCLUSIONS The data indicate that hyperosmotic expression of AQP8 in RPE cells is dependent on the activation of KATP channels. The data suggest that AQP8 activity decreases the hypoxic VEGF expression and improves the viability of RPE cells which may have impact for ischemic retinal diseases like diabetic retinopathy and age-related macular degeneration.
Collapse
Affiliation(s)
- Benjamin Schnabel
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Heidrun Kuhrt
- Institute of Anatomy, Medical Faculty, University of Leipzig, Germany
| | - Peter Wiedemann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Margrit Hollborn
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
37
|
Cross-Talk between NADPH Oxidase and Mitochondria: Role in ROS Signaling and Angiogenesis. Cells 2020; 9:cells9081849. [PMID: 32781794 PMCID: PMC7466096 DOI: 10.3390/cells9081849] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, a new vessel formation from the pre-existing ones, is essential for embryonic development, wound repair and treatment of ischemic heart and limb diseases. However, dysregulated angiogenesis contributes to various pathologies such as diabetic retinopathy, atherosclerosis and cancer. Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) as well as mitochondria play an important role in promoting the angiogenic switch from quiescent endothelial cells (ECs). However, how highly diffusible ROS produced from different sources and location can communicate with each other to regulate angiogenesis remains unclear. To detect a localized ROS signal in distinct subcellular compartments in real time in situ, compartment-specific genetically encoded redox-sensitive fluorescence biosensors have been developed. Recently, the intercellular communication, “cross-talk”, between ROS derived from NOX and mitochondria, termed “ROS-induced ROS release”, has been proposed as a mechanism for ROS amplification at distinct subcellular compartments, which are essential for activation of redox signaling. This “ROS-induced ROS release” may represent a feed-forward mechanism of localized ROS production to maintain sustained signaling, which can be targeted under pathological conditions with oxidative stress or enhanced to promote therapeutic angiogenesis. In this review, we summarize the recent knowledge regarding the role of the cross-talk between NOX and mitochondria organizing the sustained ROS signaling involved in VEGF signaling, neovascularization and tissue repair.
Collapse
|
38
|
Wang J, Toan S, Zhou H. Mitochondrial quality control in cardiac microvascular ischemia-reperfusion injury: New insights into the mechanisms and therapeutic potentials. Pharmacol Res 2020; 156:104771. [PMID: 32234339 DOI: 10.1016/j.phrs.2020.104771] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022]
Abstract
Thrombolytic therapy and revascularization strategies create a complete recanalization of the occluded epicardial coronary artery in patients with myocardial infarction (MI). However, about 35 % of patients still experience an impaired myocardial reperfusion, which is termed a no-reflow phenomenon mainly caused by cardiac microvascular ischemia-reperfusion (I/R) injury. Mitochondria are essential for microvascular endothelial cells' survival, both because of their roles as metabolic energy producers and as regulators of programmed cell death. Mitochondrial structure and function are regulated by a mitochondrial quality control (MQC) system, a series of processes including mitochondrial biogenesis, mitochondrial dynamics/mitophagy, mitochondrial proteostasis, and mitochondria-mediated cell death. Our review discusses the MQC mechanisms and how they are linked to cardiac microvascular I/R injury. Additionally, we will summarize the molecular basis that results in defective MQC mechanisms and present potential therapeutic interventions for improving MQC in cardiac microvascular I/R injury.
Collapse
Affiliation(s)
- Jin Wang
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN 55812, USA
| | - Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
39
|
Mitochondrial MiRNA in Cardiovascular Function and Disease. Cells 2019; 8:cells8121475. [PMID: 31766319 PMCID: PMC6952824 DOI: 10.3390/cells8121475] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs functioning as crucial post-transcriptional regulators of gene expression involved in cardiovascular development and health. Recently, mitochondrial miRNAs (mitomiRs) have been shown to modulate the translational activity of the mitochondrial genome and regulating mitochondrial protein expression and function. Although mitochondria have been verified to be essential for the development and as a therapeutic target for cardiovascular diseases, we are just beginning to understand the roles of mitomiRs in the regulation of crucial biological processes, including energy metabolism, oxidative stress, inflammation, and apoptosis. In this review, we summarize recent findings regarding how mitomiRs impact on mitochondrial gene expression and mitochondrial function, which may help us better understand the contribution of mitomiRs to both the regulation of cardiovascular function under physiological conditions and the pathogenesis of cardiovascular diseases.
Collapse
|