1
|
Xu CZ, Gao QY, Gao GH, Chen ZT, Wu MX, Liao GH, Cai YW, Chen N, Wang JF, Zhang HF. FTMT-dependent mitophagy is crucial for ferroptosis resistance in cardiac fibroblast. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119825. [PMID: 39168410 DOI: 10.1016/j.bbamcr.2024.119825] [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: 10/10/2023] [Revised: 07/05/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
Metabolic responses to cellular stress are pivotal in cell ferroptosis, with mitophagy serving as a crucial mechanism in both metabolic processes and ferroptosis. This study aims to elucidate the effects of high glucose on cardiomyocytes (CMs) and cardiac fibroblasts (CFs) regarding ferroptosis and to uncover the underlying mechanisms involved. We examined alterations in glycolysis, mitochondrial oxidative phosphorylation (OXPHOS), and mitophagy, which are essential for metabolic adaptations and ferroptosis. High glucose exposure induced ferroptosis specifically in CMs, while CFs exhibited resistance to ferroptosis, increased glycolytic activity, and no change in OXPHOS. Moreover, high glucose treatment enhanced mitophagy and upregulated mitochondrial ferritin (FTMT). Notably, the combination of FTMT and the autophagy-related protein nuclear receptor coactivator 4 (NCOA4) increased under high glucose conditions. Silencing FTMT significantly impeded mitophagy and eliminated ferroptosis resistance in CFs cultured under high glucose conditions. The transcription factor forkhead box A1 (FOXA1) was upregulated in CFs upon high glucose exposure, playing a crucial role in the increased expression of FTMT. Within the 5'-flanking sequence of the FTMT mRNA, approximately -500 nt from the transcription initiation site, three putative FOXA1 binding sites were identified. High glucose augmented the binding affinity between FOXA1 and these sequences, thereby promoting FTMT transcription. In summary, high glucose upregulated FOXA1 expression and stimulated FTMT promoter activity in CFs, thereby promoting FTMT-dependent mitophagy and conferring ferroptosis resistance in CFs.
Collapse
Affiliation(s)
- Cheng-Zhang Xu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qing-Yuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Guang-Hao Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Mao-Xiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Guang-Hong Liao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yang-Wei Cai
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Nuo Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
2
|
Olkowicz M, Karas A, Berkowicz P, Kaczara P, Jasztal A, Kurylowicz Z, Fedak F, Rosales-Solano H, Roy KS, Kij A, Buczek E, Pawliszyn J, Chlopicki S. Upregulation of ALOX12-12-HETE pathway impairs AMPK-dependent modulation of vascular metabolism in ApoE/LDLR -/- mice. Pharmacol Res 2024; 210:107478. [PMID: 39448044 DOI: 10.1016/j.phrs.2024.107478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Mitochondrial dysfunction and 12-lipoxygenase (ALOX12)-derived 12(S)-HETE production have been associated with vascular inflammation and the pathogenesis of atherosclerosis. However, the role of ALOX12 in regulating vascular energy metabolism in vascular inflammation has not been studied to date. Using mitochondrial and glycolysis functional profiling with the Seahorse extracellular flux analyzer, metabolipidomics, and proteomic analysis (LC-MS/MS), we characterized alterations in vascular energy metabolism in 2- and 6-month-old ApoE/LDLR-/- vs. control C57BL/6 mice. We identified that aorta of 6-month-old ApoE/LDLR-/- mice displayed compromised mitochondrial metabolism manifested by the reduced expression of mitochondrial enzymes, impaired mitochondrial respiration, and consequently diminished respiratory reserve capacity. An increased flux through the glycolysis/lactate shuttle, the hexosamine biosynthetic pathway (HBP), and the pentose phosphate pathway (PPP) was also recognized. Interestingly, ALOX12-12-HETE was the most upregulated axis in eicosanoid metabolism and histological examinations indicated that ApoE/LDLR-/- mice showed increased aortic expression of ALOX12, particularly in early atherosclerotic plaque areas. Remarkably, the joint blocking of ALOX12 and activation of AMPK, but not AMPK activation alone, resulted in the reprogramming of vascular metabolism, with improved mitochondrial respiration and suppressed auxiliary pathways (HBP, PPP, itaconate shunt). In conclusion, excessive activation of the ALOX12-12-HETE pathway in vascular inflammation in early atherosclerosis inhibits AMPK-dependent regulation of vascular metabolism. Consequently, ALOX12 may represent a novel target to boost impaired vascular mitochondrial function in pro-atherosclerotic vascular inflammation.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland.
| | - Agnieszka Karas
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Piotr Berkowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Patrycja Kaczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Zuzanna Kurylowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Filip Fedak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Hernando Rosales-Solano
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Kanchan Sinha Roy
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Department of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, Krakow 31-531, Poland
| |
Collapse
|
3
|
Xiong YB, Huang WY, Ling X, Zhou S, Wang XX, Li XL, Zhou LL. Mitochondrial calcium uniporter promotes kidney aging in mice through inducing mitochondrial calcium-mediated renal tubular cell senescence. Acta Pharmacol Sin 2024; 45:2149-2162. [PMID: 38789496 PMCID: PMC11420221 DOI: 10.1038/s41401-024-01298-5] [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: 12/29/2023] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Renal tubular epithelial cell senescence plays a critical role in promoting and accelerating kidney aging and age-related renal fibrosis. Senescent cells not only lose their self-repair ability, but also can transform into senescence-associated secretory phenotype (SASP) to trigger inflammation and fibrogenesis. Recent studies show that mitochondrial dysfunction is critical for renal tubular cell senescence and kidney aging, and calcium overload and abnormal calcium-dependent kinase activities are involved in mitochondrial dysfunction-associated senescence. In this study we investigated the role of mitochondrial calcium overload and mitochondrial calcium uniporter (MCU) in kidney aging. By comparing the kidney of 2- and 24-month-old mice, we found calcium overload in renal tubular cells of aged kidney, accompanied by significantly elevated expression of MCU. In human proximal renal tubular cell line HK-2, pretreatment with MCU agonist spermine (10 μM) significantly increased mitochondrial calcium accumulation, and induced the production of reactive oxygen species (ROS), leading to renal tubular cell senescence and age-related kidney fibrosis. On the contrary, pretreatment with MCU antagonist RU360 (10 μM) or calcium chelator BAPTA-AM (10 μM) diminished D-gal-induced ROS generation, restored mitochondrial homeostasis, retarded cell senescence, and protected against kidney aging in HK-2 cells. In a D-gal-induced accelerated aging mice model, administration of BAPTA (100 μg/kg. i.p.) every other day for 8 weeks significantly alleviated renal tubuarl cell senescence and fibrosis. We conclude that MCU plays a key role in promoting renal tubular cell senescence and kidney aging. Targeting inhibition on MCU provides a new insight into the therapeutic strategy against kidney aging.
Collapse
Affiliation(s)
- Ya-Bing Xiong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Yan Huang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Xu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Long Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li-Li Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology / Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
Zhu G, Li Y, Gao H, Li X, Fan H, Fan L. Mzb1 Attenuates Atherosclerotic Plaque Vulnerability in ApoE-/- Mice by Alleviating Apoptosis and Modulating Mitochondrial Function. J Cardiovasc Transl Res 2024; 17:782-794. [PMID: 38294627 DOI: 10.1007/s12265-024-10483-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
In this study, we investigated the protective role of Mzb1 in atherosclerotic plaque vulnerability. To explore the impact of Mzb1, we analyzed Mzb1 expression, assessed apoptosis, and evaluated mitochondrial function in atherosclerosis (AS) mouse models and human vascular smooth muscle cells (HVSMCs). We observed a significant decrease in Mzb1 expression in AS mouse models and ox-LDL-treated HVSMCs. Downregulation of Mzb1 increased ox-LDL-induced apoptosis and cholesterol levels of HVSMCs, while Mzb1 overexpression alleviated these effect. Mzb1 was found to enhance mitochondrial function, as evidenced by restored ATP synthesis, mitochondrial membrane potential, and reduced mtROS production. Moreover, Mzb1 overexpression attenuated atherosclerotic plaque vulnerability in ApoE-/- mice. Our findings suggest that Mzb1 overexpression regulates the AMPK/SIRT1 signaling pathway, leading to the attenuation of atherosclerotic plaque vulnerability. This study provides compelling evidence for the protective effect of Mzb1 on atherosclerotic plaques by alleviating apoptosis and modulating mitochondrial function in ApoE-/- mice.
Collapse
MESH Headings
- Animals
- Apoptosis
- Plaque, Atherosclerotic
- Disease Models, Animal
- Mice, Knockout, ApoE
- Signal Transduction
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Humans
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Atherosclerosis/prevention & control
- Sirtuin 1/metabolism
- Sirtuin 1/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- AMP-Activated Protein Kinases/metabolism
- Mice, Inbred C57BL
- Cells, Cultured
- Male
- Lipoproteins, LDL/metabolism
- Mitochondria/metabolism
- Mitochondria/pathology
- Rupture, Spontaneous
- Membrane Potential, Mitochondrial
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
- Apolipoproteins E/genetics
- Apolipoproteins E/deficiency
- Adenosine Triphosphate/metabolism
- Aorta/metabolism
- Aorta/pathology
Collapse
Affiliation(s)
- Guanglang Zhu
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, People's Republic of China
| | - Yang Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hongxia Gao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, People's Republic of China
| | - Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, People's Republic of China
| | - Heyu Fan
- School of Arts and Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Longhua Fan
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, People's Republic of China.
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Patterson MT, Xu Y, Hillman H, Osinski V, Schrank PR, Kennedy AE, Barrow F, Zhu A, Tollison S, Shekhar S, Stromnes IM, Tassi I, Wu D, Revelo XS, Binstadt BA, Williams JW. Trem2 Agonist Reprograms Foamy Macrophages to Promote Atherosclerotic Plaque Stability-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:1646-1657. [PMID: 38695172 PMCID: PMC11208052 DOI: 10.1161/atvbaha.124.320797] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/18/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Trem2 (triggering receptor on myeloid cells 2), a surface lipid receptor, is expressed on foamy macrophages within atherosclerotic lesions and regulates cell survival, proliferation, and anti-inflammatory responses. Studies examining the role of Trem2 in atherosclerosis have shown that deletion of Trem2 leads to impaired foamy macrophage lipid uptake, proliferation, survival, and cholesterol efflux. Thus, we tested the hypothesis that administration of a Trem2 agonist antibody (AL002a) to atherogenic mice would enhance macrophage survival and decrease necrotic core formation to improve plaque stability. METHODS To model a therapeutic intervention approach, atherosclerosis-prone mice (Ldlr [low-density lipoprotein receptor]-/-) were fed a high-fat diet for 8 weeks, then transitioned to treatment with AL002a or isotype control for an additional 8 weeks while continuing on a high-fat diet. RESULTS AL002a-treated mice had increased lesion size in both the aortic root and whole mount aorta, which correlated with an expansion of plaque macrophage area. This expansion was due to increased macrophage survival and proliferation in plaques. Importantly, plaques from AL002a-treated mice showed improved features of plaque stability, including smaller necrotic cores, increased fibrous caps, and greater collagen deposition. Single-cell RNA sequencing of whole aorta suspensions from isotype- and AL002a-treated atherosclerotic mice revealed that Trem2 agonism dramatically altered foamy macrophage transcriptome. This included upregulation of oxidative phosphorylation and increased expression of collagen genes. In vitro studies validated that Trem2 agonism with AL002a promoted foamy macrophage oxidized low-density lipoprotein uptake, survival, and cholesterol efflux. CONCLUSIONS Trem2 agonism expands atherosclerotic plaque macrophages by promoting cell survival and proliferation but improves features of plaque stability by rewiring foamy macrophage function to enhance cholesterol efflux and collagen deposition.
Collapse
MESH Headings
- Animals
- Plaque, Atherosclerotic
- Receptors, Immunologic/agonists
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Membrane Glycoproteins/agonists
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Disease Models, Animal
- Mice
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/drug therapy
- Atherosclerosis/prevention & control
- Foam Cells/metabolism
- Foam Cells/pathology
- Foam Cells/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Male
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Receptors, LDL/deficiency
- Cell Proliferation/drug effects
- Diet, High-Fat
- Cell Survival/drug effects
- Necrosis
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
Collapse
Affiliation(s)
- Michael T. Patterson
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Yingzheng Xu
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Hannah Hillman
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Victoria Osinski
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Pediatrics (V.O., B.A.B.), University of Minnesota, Minneapolis
| | - Patricia R. Schrank
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Ainsley E. Kennedy
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Fanta Barrow
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Alisha Zhu
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Samuel Tollison
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Sia Shekhar
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Ingunn M. Stromnes
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Microbiology and Immunology (I.M.S.), University of Minnesota, Minneapolis
| | - Ilaria Tassi
- Alector, Inc, South San Francisco, CA (I.T., D.W.)
- Now with Deep Apple Therapeutics, South San Francisco, CA (I.T.)
| | - Dick Wu
- Alector, Inc, South San Francisco, CA (I.T., D.W.)
| | - Xavier S. Revelo
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| | - Bryce A. Binstadt
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Pediatrics (V.O., B.A.B.), University of Minnesota, Minneapolis
| | - Jesse W. Williams
- Center for Immunology (M.T.P., Y.X., H.H., V.O., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., I.M.S., X.S.R., B.A.B., J.W.W.), University of Minnesota, Minneapolis
- Department of Integrative Biology and Physiology (M.T.P., Y.X., H.H., P.R.S., A.E.K., F.B., A.Z., S.T., S.S., X.S.R., J.W.W.), University of Minnesota, Minneapolis
| |
Collapse
|
6
|
Zhuang F, Huang S, Liu L. PYCR3 modulates mtDNA copy number to drive proliferation and doxorubicin resistance in triple-negative breast cancer. Int J Biochem Cell Biol 2024; 171:106581. [PMID: 38642827 DOI: 10.1016/j.biocel.2024.106581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Triple-negative breast cancer (TNBC) poses significant challenges in treatment due to its aggressive nature and limited therapeutic targets. Understanding the underlying molecular mechanisms driving TNBC progression and chemotherapy resistance is imperative for developing effective therapeutic strategies. Thus, in this study, we aimed to elucidate the role of pyrroline-5-carboxylate reductase 3 (PYCR3) in TNBC pathogenesis and therapeutic response. We observed that PYCR3 is significantly upregulated in TNBC specimens compared to normal breast tissues, correlating with a poorer prognosis in TNBC patients. Knockdown of PYCR3 not only suppresses TNBC cell proliferation but also reverses acquired resistance of TNBC cells to doxorubicin, a commonly used chemotherapeutic agent. Mechanistically, we identified the mitochondrial localization of PYCR3 in TNBC cells and demonstrated its impact on TNBC cell proliferation and sensitivity to doxorubicin through the regulation of mtDNA copy number and mitochondrial respiration. Importantly, Selective reduction of mtDNA copy number using the mtDNA replication inhibitor 2', 3'-dideoxycytidine effectively recapitulates the phenotypic effects observed in PYCR3 knockout, resulting in decreased TNBC cell proliferation and the reversal of doxorubicin resistance through apoptosis induction. Thus, our study underscores the clinical relevance of PYCR3 and highlight its potential as a therapeutic target in TNBC management. By elucidating the functional significance of PYCR3 in TNBC, our findings contribute to a deeper understanding of TNBC biology and provide a foundation for developing novel therapeutic strategies aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Feifei Zhuang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong, China
| | - Shaoyan Huang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong, China
| | - Lei Liu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong, China.
| |
Collapse
|
7
|
Xu X, Qiu F, Yang M, Liu X, Tao S, Zheng B. Unveiling Atherosclerotic Plaque Heterogeneity and SPP1 +/VCAN + Macrophage Subtype Prognostic Significance Through Integrative Single-Cell and Bulk-Seq Analysis. J Inflamm Res 2024; 17:2399-2426. [PMID: 38681071 PMCID: PMC11055562 DOI: 10.2147/jir.s454505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Background Dysregulated macrophages are important causes of Atherosclerosis (AS) formation and increased plaque instability, but the heterogeneity of these plaques and the role of macrophage subtypes in plaque instability have yet to be clarified. Methods This study integrates single-cell and bulk-seq data to analyze atherosclerotic plaques. Unsupervised clustering was used to reveal distinct plaque subtypes, while survival analysis and gene set variation analysis (GSVA) methods helped in understanding their clinical outcomes. Enrichment of differential expression of macrophage genes (DEMGs) score and pseudo-trajectory analysis were utilized to explore the biological functions and differentiation stages of macrophage subtypes in AS progression. Additionally, CellChat and the BayesPrism deconvolution method were used to elucidate macrophage subtype interaction and their prognostic significance at single-cell resolution. Finally, the expression of biomarkers was validated in mouse experiments. Results Three distinct AS plaque subtypes were identified, with cluster 3 plaque subtype being particularly associated with higher immune infiltration and poorer prognosis. The DEMGs score exhibited a significant elevation in three macrophage subtypes (SPP1+/VCAN+ macrophages, IL1B+ macrophages, and FLT3LG+ macrophages), associated with cluster 3 plaque subtype and highlighted the prognostic significance of these subtypes. Activation trajectory of the macrophage subtypes is divided into three states (Pre-branch, Cell fate 1, and Cell fate 2), and Cell fate 2 (SPP1+/VCAN+ macrophages, IL1B+ macrophages, and FLT3LG+ macrophages dominant) exhibiting the highest DEMGs score, distinct interactions with other cell components, and relating to poorer prognosis of ischemic events. This study also uncovered a unique SPP1+/VCAN+ macrophage subtype, rare in quantity but significant in influencing AS progression. Machine learning algorithms identified 10 biomarkers crucial for AS diagnosis. The validation of these biomarkers was performed using Mendelian Randomization analysis and in vitro methods, supporting their relevance in AS pathology. Conclusion Our study provides a comprehensive view of AS plaque heterogeneity and the prognostic significance of macrophage subtypes in plaque instability.
Collapse
Affiliation(s)
- Xiang Xu
- School of Medicine, Yunnan University, Kunming City, Yunnan Province, People’s Republic of China
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Fuling Qiu
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Man Yang
- School of Medicine, Dali University, Dali City, Yunnan Province, People’s Republic of China
| | - Xiaoyong Liu
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Siming Tao
- Department of Cardiology, The Affiliated Hospital of Yunnan University, Kunming City, Yunnan Province, People’s Republic of China
| | - Bingrong Zheng
- School of Medicine, Yunnan University, Kunming City, Yunnan Province, People’s Republic of China
| |
Collapse
|
8
|
Xu R, Li T, Luo J, Zhang X, Wang T, Wang Y, Ma Y, Yang B, Jia J, Dmytriw AA, Li W, Jiao L. PCSK9 increases vulnerability of carotid plaque by promoting mitochondrial dysfunction and apoptosis of vascular smooth muscle cells. CNS Neurosci Ther 2024; 30:e14640. [PMID: 38402551 PMCID: PMC10894644 DOI: 10.1111/cns.14640] [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: 11/13/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been recognized as a novel lipid-lowing target. Recent clinical studies suggested the value of inhibiting PCSK9 in decreasing the vulnerability of coronary plaques. However, the evidence of PCSK9-regulated evolution of unstable carotid plaques is unclear, which has limited the use of PCSK9 inhibitor in carotid plaques. This study aimed to determine the effect and molecular mechanisms of PCSK9 on vulnerability of carotid plaques, to provide potential therapeutic targets for stabilizing carotid plaques. METHODS The expression of PCSK9 in stable and unstable carotid plaques were examined in tissue and plasma. Human aortic vascular smooth muscle cells (VSMCs) and carotid VSMCs were employed to transfect lentivirus for overexpression and knockdown of PCSK9, respectively. Morphological and functional changes of mitochondria were observed by live-cell imaging. Cell apoptosis was evaluated by propidium iodide staining. RNA-sequencing and biological examinations were performed to explore and validate the underlying mechanisms. Truncated plasmids were employed to identify the functional domain of PCSK9 in regulation of VSMCs' mitochondrial morphology, function and apoptosis. RESULTS Clinically, PCSK9 was closely related with vulnerability of human carotid plaques. Increased expression of PCSK9 in human VSMCs was accompanied by higher level of apoptosis. At subcellular level of VSMCs, the morphology of mitochondria was shifted toward the fission state, followed by mitochondrial dysfunction. Inhibition of p38 MAPK activation partially rescued the above morphological and behavioral changes caused by PCSK9. Furthermore, inhibiting of dynamin-related protein 1 (DRP1) attenuated PCSK9-related mitochondrial dysfunction and cell apoptosis. The 1-149aa domain of PCSK9 protein was essential to achieve functional regulation to VSMCs. CONCLUSION Our findings demonstrated that PCSK9 induced morphology-related mitochondrial dysfunction and apoptosis of VSMCs, which may be related to increased vulnerability of carotid plaque.
Collapse
Affiliation(s)
- Ran Xu
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Tianhua Li
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Tao Wang
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Yabing Wang
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Yan Ma
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Bin Yang
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
| | - Jinzhu Jia
- School of Public Health and Center for Statistical SciencePeking UniversityBeijingChina
| | - Adam A. Dmytriw
- Neuroendovascular Program, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern RecognitionInstitute of Automation, Chinese Academy of SciencesBeijingChina
- School of Artificial IntelligenceUniversity of Chinese Academy of SciencesBeijingChina
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI)BeijingChina
- Department of Interventional Neuroradiology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Daepartment of Neurosurgery and Neurology, Jinan Hospital of Xuanwu HospitalShandong First Medical UniversityJinanChina
| |
Collapse
|
9
|
Ma R, Gao QY, Chen ZT, Liao GH, Li ST, Cai JW, Luo NS, Chen H, Zhang HF. SIRT3 suppression resulting from the enhanced β-catenin signaling drives glycolysis and promotes hypoxia-induced cell growth in hepatocellular carcinoma cells. Cell Cycle 2024; 23:435-447. [PMID: 38626328 PMCID: PMC11174062 DOI: 10.1080/15384101.2024.2340864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 04/04/2024] [Indexed: 04/18/2024] Open
Abstract
The precise mechanisms underlying the inhibitory effects of SIRT3, a mitochondrial sirtuin protein, on hepatocellular carcinoma (HCC) development, as well as its impact on mitochondrial respiration, remain poorly understood. We assessed sirtuins 3 (SIRT3) levels in HCC tissues and Huh7 cells cultured under hypoxic condition. We investigated the effects of SIRT3 on cell proliferation, glycolytic metabolism, mitochondrial respiration, mitophagy, and mitochondrial biogenesis in Huh7 cells. Besides, we explored the potential mechanisms regulating SIRT3 expression in hypoxically cultured Huh7 cells. Gradual reduction in SIRT3 expressions were observed in both adjacent tumor tissues and tumor tissues. Similarly, SIRT3 expressions were diminished in Huh7 cells cultured under hypoxic condition. Forced expression of SIRT3 attenuated the growth of hypoxically cultured Huh7 cells. SIRT3 overexpression led to a decrease in extracellular acidification rate while increasing oxygen consumption rate. SIRT3 downregulated the levels of hexokinase 2 and pyruvate kinase M2. Moreover, SIRT3 enhanced mitophagy signaling, as indicated by mtKeima, and upregulated key proteins involved in various mitophagic pathways while reducing intracellular reactive oxygen species levels. Furthermore, SIRT3 increased proxisome proliferator-activated receptor-gamma coactivator 1α levels and the amount of mitochondrial DNA in Huh7 cells. Notably, β-catenin expressions were elevated in Huh7 cells cultured under hypoxic condition. Antagonists and agonists of β-catenin respectively upregulated and downregulated SIRT3 expressions in hypoxically cultured Huh7 cells. The modulationsof glycolysis and mitochondrial respiration represent the primary mechanism through which SIRT3, suppressed by β-catenin, inhibits HCC cell proliferation.
Collapse
Affiliation(s)
- Rong Ma
- Department of Infectious Diseases, The First Affiliated Hospital of Chengdu Medical College, Chengdu, PRC
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Qing-Yuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Guang-Hong Liao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Shu-Tai Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Jie-Wen Cai
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Nian-Sang Luo
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| | - Hao Chen
- Department of Gastroenterology, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, PRC
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PRC
| |
Collapse
|
10
|
Ali MA, Gioscia-Ryan R, Yang D, Sutton NR, Tyrrell DJ. Cardiovascular aging: spotlight on mitochondria. Am J Physiol Heart Circ Physiol 2024; 326:H317-H333. [PMID: 38038719 PMCID: PMC11219063 DOI: 10.1152/ajpheart.00632.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/02/2023]
Abstract
Mitochondria are cellular organelles critical for ATP production and are particularly relevant to cardiovascular diseases including heart failure, atherosclerosis, ischemia-reperfusion injury, and cardiomyopathies. With advancing age, even in the absence of clinical disease, mitochondrial homeostasis becomes disrupted (e.g., redox balance, mitochondrial DNA damage, oxidative metabolism, and mitochondrial quality control). Mitochondrial dysregulation leads to the accumulation of damaged and dysfunctional mitochondria, producing excessive reactive oxygen species and perpetuating mitochondrial dysfunction. In addition, mitochondrial DNA, cardiolipin, and N-formyl peptides are potent activators of cell-intrinsic and -extrinsic inflammatory pathways. These age-related mitochondrial changes contribute to the development of cardiovascular diseases. This review covers the impact of aging on mitochondria and links these mechanisms to therapeutic implications for age-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Md Akkas Ali
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Rachel Gioscia-Ryan
- Department of Anesthesiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Dongli Yang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Nadia R Sutton
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - Daniel J Tyrrell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
11
|
Jouabadi SM, Ataabadi EA, Golshiri K, Bos D, Stricker BHC, Danser AHJ, Mattace-Raso F, Roks AJM. Clinical Impact and Mechanisms of Nonatherosclerotic Vascular Aging: The New Kid to Be Blocked. Can J Cardiol 2023; 39:1839-1858. [PMID: 37495207 DOI: 10.1016/j.cjca.2023.07.022] [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: 05/05/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
Ischemic cardiovascular disease and stroke remain the leading cause of global morbidity and mortality. During aging, protective mechanisms in the body gradually deteriorate, resulting in functional, structural, and morphologic changes that affect the vascular system. Because atherosclerotic plaques are not always present along with these alterations, we refer to this kind of vascular aging as nonatherosclerotic vascular aging (NAVA). To maintain proper vascular function during NAVA, it is important to preserve intracellular signalling, prevent inflammation, and block the development of senescent cells. Pharmacologic interventions targeting these components are potential therapeutic approaches for NAVA, with a particular emphasis on inflammation and senescence. This review provides an overview of the pathophysiology of vascular aging and explores potential pharmacotherapies that can improve the function of aged vasculature, focusing on NAVA.
Collapse
Affiliation(s)
- Soroush Mohammadi Jouabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ehsan Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Keivan Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Bruno H C Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Francesco Mattace-Raso
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
12
|
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol 2023; 20:754-777. [PMID: 37193857 DOI: 10.1038/s41569-023-00881-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Normal circulatory function is a key determinant of disease-free life expectancy (healthspan). Indeed, pathologies affecting the cardiovascular system, which are growing in prevalence, are the leading cause of global morbidity, disability and mortality, whereas the maintenance of cardiovascular health is necessary to promote both organismal healthspan and lifespan. Therefore, cardiovascular ageing might precede or even underlie body-wide, age-related health deterioration. In this Review, we posit that eight molecular hallmarks are common denominators in cardiovascular ageing, namely disabled macroautophagy, loss of proteostasis, genomic instability (in particular, clonal haematopoiesis of indeterminate potential), epigenetic alterations, mitochondrial dysfunction, cell senescence, dysregulated neurohormonal signalling and inflammation. We also propose a hierarchical order that distinguishes primary (upstream) from antagonistic and integrative (downstream) hallmarks of cardiovascular ageing. Finally, we discuss how targeting each of the eight hallmarks might be therapeutically exploited to attenuate residual cardiovascular risk in older individuals.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- BioTechMed Graz, Graz, Austria.
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
13
|
Gindri dos Santos B, Goedeke L. Macrophage immunometabolism in diabetes-associated atherosclerosis. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00032. [PMID: 37849988 PMCID: PMC10578522 DOI: 10.1097/in9.0000000000000032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023]
Abstract
Macrophages play fundamental roles in atherosclerotic plaque formation, growth, and regression. These cells are extremely plastic and perform different immune functions depending on the stimuli they receive. Initial in vitro studies have identified specific metabolic pathways that are crucial for the proper function of pro-inflammatory and pro-resolving macrophages. However, the plaque microenvironment, especially in the context of insulin resistance and type 2 diabetes, constantly challenges macrophages with several simultaneous inflammatory and metabolic stimuli, which may explain why atherosclerosis is accelerated in diabetic patients. In this mini review, we discuss how macrophage mitochondrial function and metabolism of carbohydrates, lipids, and amino acids may be affected by this complex plaque microenvironment and how risk factors associated with type 2 diabetes alter the metabolic rewiring of macrophages and disease progression. We also briefly discuss current challenges in assessing macrophage metabolism and identify future tools and possible strategies to alter macrophage metabolism to improve treatment options for diabetes-associated atherosclerosis.
Collapse
Affiliation(s)
- Bernardo Gindri dos Santos
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leigh Goedeke
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine (Endocrinology), The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
Xue S, Su Z, Liu D. Immunometabolism and immune response regulate macrophage function in atherosclerosis. Ageing Res Rev 2023; 90:101993. [PMID: 37379970 DOI: 10.1016/j.arr.2023.101993] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023]
Abstract
Macrophages are crucial in the progression of atherosclerotic cardiovascular disease (ASCVD). In the atherosclerotic lesions, macrophages play a central role in maintaining inflammatory response, promoting plaque development, and facilitating thrombosis. Increasing studies indicate that metabolic reprogramming and immune response mediate macrophage functional changes in all stages of atherosclerosis. In this review article, we explain how metabolic changes in glycolysis, oxidative phosphorylation, the tricarboxylic acid cycle, fatty acid synthesis, fatty acid oxidation, and cholesterol metabolism regulate macrophage function in atherosclerosis. We discuss how immune response to oxidized lipids regulate macrophage function in atherosclerosis. Additionally, we explore how abnormal metabolism leads to macrophage mitochondrial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Sheng Xue
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China.
| | - Zhe Su
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China
| | - Dacheng Liu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China
| |
Collapse
|
15
|
Wojtasińska A, Frąk W, Lisińska W, Sapeda N, Młynarska E, Rysz J, Franczyk B. Novel Insights into the Molecular Mechanisms of Atherosclerosis. Int J Mol Sci 2023; 24:13434. [PMID: 37686238 PMCID: PMC10487483 DOI: 10.3390/ijms241713434] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Atherosclerosis is one of the most fatal diseases in the world. The associated thickening of the arterial wall and its background and consequences make it a very composite disease entity with many mechanisms that lead to its creation. It is an active process, and scientists from various branches are engaged in research, including molecular biologists, cardiologists, and immunologists. This review summarizes the available information on the pathophysiological implications of atherosclerosis, focusing on endothelium dysfunction, inflammatory factors, aging, and uric acid, vitamin D, and miRNA expression as recent evidence of interactions of the molecular and cellular elements. Analyzing new discoveries for the underlying causes of this condition assists the general research to improve understanding of the mechanism of pathophysiology and thus prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Armanda Wojtasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Natalia Sapeda
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| |
Collapse
|
16
|
van Eeden C, Redmond D, Mohazab N, Larché MJ, Mason AL, Cohen Tervaert JW, Osman MS. Evidence of a Novel Mitochondrial Signature in Systemic Sclerosis Patients with Chronic Fatigue Syndrome. Int J Mol Sci 2023; 24:12057. [PMID: 37569433 PMCID: PMC10418481 DOI: 10.3390/ijms241512057] [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: 06/28/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Symptoms of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are common in rheumatic diseases, but no studies report the frequency of these in early systemic sclerosis. There are no known biomarkers that can distinguish between patients with ME/CFS, although mitochondrial abnormalities are often demonstrated. We sought to assess the prevalence of ME/CFS in limited cutaneous SSc (lcSSc) patients early in their disease (<5 years from the onset of non-Raynaud's symptoms) and to determine if alterations in mitochondrial electron transport chain (ETC) transcripts and mitochondrial DNA (mtDNA) integrity could be used to distinguish between fatigued and non-fatigued patients. All SSc patients met ACR/EULAR classification criteria. ME/CFS-related symptoms were assessed through validated questionnaires, and the expression of ETC transcripts and mtDNA integrity were quantified via qPCR. SSc patients with ME/CFS could be distinguished from non-fatigued patients through ETC gene analysis; specifically, reduced expression of ND4 and CyB and increased expression of Cox7C. ND4 and CyB expression correlated with indicators of disease severity. Further prospective and functional studies are needed to determine if this altered signature can be further utilized to better identify ME/CFS in SSc patients, and whether ME/CFS in early SSc disease could predict more severe disease outcomes.
Collapse
Affiliation(s)
- Charmaine van Eeden
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Desiree Redmond
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Naima Mohazab
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Maggie J. Larché
- Division of Rheumatology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Andrew L. Mason
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Jan Willem Cohen Tervaert
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Mohammed S. Osman
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
17
|
Liu S, Zheng Z, Zhao Y, Yao H, Zhang L, Chen C, Jin S. DRP1 knockdown and atorvastatin alleviate ox-LDL-induced vascular endothelial cells injury: DRP1 is a potential target for preventing atherosclerosis. Exp Cell Res 2023:113688. [PMID: 37315759 DOI: 10.1016/j.yexcr.2023.113688] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/03/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023]
Abstract
Vascular endothelial cells (VECs) injury is the first step in the pathogenesis of atherosclerosis (AS). Mitochondrial dysfunction plays a significant role in VECs injury, but the underlying mechanisms are still unclear. Here, the human umbilical vein endothelial cells were exposed to 100 μg/mL oxidized low-density lipoprotein for 24 h to establish AS model in vitro. We reported that mitochondrial dynamics disorder is a prominent feature of VECs in AS models and associated with mitochondrial dysfunction. Moreover, the knockdown of dynamin-related protein 1 (DRP1) in AS model significantly alleviated the mitochondrial dynamics disorder and VECs injury. On the contrary, DRP1 overexpression significantly aggravated this injury. Interestingly, atorvastatin (ATV), a classical anti-atherosclerotic drug, prominently inhibited the expression of DRP1 in AS models and similarly alleviated the mitochondrial dynamics disorder and VECs injury in vitro and in vivo. At the same time, we found that ATV alleviated VECs damage but did not significantly reduce lipid concentration in vivo. Our findings provide a potential therapeutic target of AS and a new mechanism of the anti-atherosclerotic effect of ATV.
Collapse
Affiliation(s)
- Shengnan Liu
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Zhinan Zheng
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Yingyin Zhao
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Hanming Yao
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Lizhen Zhang
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Cui Chen
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Sanqing Jin
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China.
| |
Collapse
|
18
|
Li J, Li X, Song S, Sun Z, Li Y, Yang L, Xie Z, Cai Y, Zhao Y. Mitochondria spatially and temporally modulate VSMC phenotypes via interacting with cytoskeleton in cardiovascular diseases. Redox Biol 2023; 64:102778. [PMID: 37321061 DOI: 10.1016/j.redox.2023.102778] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
Cardiovascular diseases caused by atherosclerosis (AS) seriously endanger human health, which is closely related to vascular smooth muscle cell (VSMC) phenotypes. VSMC phenotypic transformation is marked by the alteration of phenotypic marker expression and cellular behaviour. Intriguingly, the mitochondrial metabolism and dynamics altered during VSMC phenotypic transformation. Firstly, this review combs VSMC mitochondrial metabolism in three aspects: mitochondrial ROS generation, mutated mitochondrial DNA (mtDNA) and calcium metabolism respectively. Secondly, we summarized the role of mitochondrial dynamics in regulating VSMC phenotypes. We further emphasized the association between mitochondria and cytoskelton via presenting cytoskeletal support during mitochondrial dynamics process, and discussed its impact on their respective dynamics. Finally, considering that both mitochondria and cytoskeleton are mechano-sensitive organelles, we demonstrated their direct and indirect interaction under extracellular mechanical stimuli through several mechano-sensitive signaling pathways. We additionally discussed related researches in other cell types in order to inspire deeper thinking and reasonable speculation of potential regulatory mechanism in VSMC phenotypic transformation.
Collapse
Affiliation(s)
- Jingwen Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Xinyue Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Sijie Song
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhengwen Sun
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yuanzhu Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Long Yang
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhenhong Xie
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yikui Cai
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yinping Zhao
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
19
|
Becker PH, Thérond P, Gaignard P. Targeting mitochondrial function in macrophages: A novel treatment strategy for atherosclerotic cardiovascular disease? Pharmacol Ther 2023; 247:108441. [PMID: 37201736 DOI: 10.1016/j.pharmthera.2023.108441] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023]
Abstract
Atherosclerotic cardiovascular disease is a major cause of morbidity and mortality due to chronic arterial injury caused by hyperlipidemia, hypertension, inflammation and oxidative stress. Recent studies have shown that the progression of this disease is associated with mitochondrial dysfunction and with the accumulation of mitochondrial alterations within macrophages of atherosclerotic plaques. These alterations contribute to processes of inflammation and oxidative stress. Among the many players involved, macrophages play a pivotal role in atherogenesis as they can exert both beneficial and deleterious effects due to their anti- and pro-inflammatory properties. Their atheroprotective functions, such as cholesterol efflux and efferocytosis, as well as the maintenance of their polarization towards an anti-inflammatory state, are particularly dependent on mitochondrial metabolism. Moreover, in vitro studies have demonstrated deleterious effects of oxidized LDL on macrophage mitochondrial function, resulting in a switch to a pro-inflammatory state and to a potential loss of atheroprotective capacity. Therefore, preservation of mitochondrial function is now considered a legitimate therapeutic strategy. This review focuses on the potential therapeutic strategies that could improve the mitochondrial function of macrophages, enabling them to maintain their atheroprotective capacity. These emerging therapies could play a valuable role in counteracting the progression of atherosclerotic lesions and possibly inducing their regression.
Collapse
Affiliation(s)
- Pierre-Hadrien Becker
- Université Paris-Saclay, EA 7357, Lipides: Systèmes Analytiques et Biologiques, Châtenay-Malabry 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre 94270, France.
| | - Patrice Thérond
- Université Paris-Saclay, EA 7357, Lipides: Systèmes Analytiques et Biologiques, Châtenay-Malabry 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre 94270, France
| | - Pauline Gaignard
- Université Paris-Saclay, EA 7357, Lipides: Systèmes Analytiques et Biologiques, Châtenay-Malabry 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre 94270, France
| |
Collapse
|
20
|
Zhang Y, Weng J, Huan L, Sheng S, Xu F. Mitophagy in atherosclerosis: from mechanism to therapy. Front Immunol 2023; 14:1165507. [PMID: 37261351 PMCID: PMC10228545 DOI: 10.3389/fimmu.2023.1165507] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
Mitophagy is a type of autophagy that can selectively eliminate damaged and depolarized mitochondria to maintain mitochondrial activity and cellular homeostasis. Several pathways have been found to participate in different steps of mitophagy. Mitophagy plays a significant role in the homeostasis and physiological function of vascular endothelial cells, vascular smooth muscle cells, and macrophages, and is involved in the development of atherosclerosis (AS). At present, many medications and natural chemicals have been shown to alter mitophagy and slow the progression of AS. This review serves as an introduction to the field of mitophagy for researchers interested in targeting this pathway as part of a potential AS management strategy.
Collapse
Affiliation(s)
- Yanhong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiajun Weng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| | - Luyao Huan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Song Sheng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
21
|
Vyas HS, Jadeja RN, Vohra A, Upadhyay KK, Thounaojam MC, Bartoli M, Devkar RV. CORM-A1 Alleviates Pro-Atherogenic Manifestations via miR-34a-5p Downregulation and an Improved Mitochondrial Function. Antioxidants (Basel) 2023; 12:antiox12050997. [PMID: 37237862 DOI: 10.3390/antiox12050997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Atherogenesis involves multiple cell types undergoing robust metabolic processes resulting in mitochondrial dysfunction, elevated reactive oxygen species (ROS), and consequent oxidative stress. Carbon monoxide (CO) has been recently explored for its anti-atherogenic potency; however, the effects of CO on ROS generation and mitochondrial dysfunction in atherosclerosis remain unexplored. Herein, we describe the anti-atherogenic efficacy of CORM-A1, a CO donor, in in vitro (ox-LDL-treated HUVEC and MDMs) and in vivo (atherogenic diet-fed SD rats) experimental models. In agreement with previous data, we observed elevated miR-34a-5p levels in all our atherogenic model systems. Administration of CO via CORM-A1 accounted for positive alterations in the expression of miR-34a-5p and transcription factors/inhibitors (P53, NF-κB, ZEB1, SNAI1, and STAT3) and DNA methylation pattern, thereby lowering its countenance in atherogenic milieu. Inhibition of miR-34a-5p expression resulted in restoration of SIRT-1 levels and of mitochondrial biogenesis. CORM-A1 supplementation further accounted for improvement in cellular and mitochondrial antioxidant capacity and subsequent reduction in ROS. Further and most importantly, CORM-A1 restored cellular energetics by improving overall cellular respiration in HUVECs, as evidenced by restored OCR and ECAR rates, whereas a shift from non-mitochondrial to mitochondrial respiration was observed in atherogenic MDMs, evidenced by unaltered glycolytic respiration and maximizing OCR. In agreement with these results, CORM-A1 treatment also accounted for elevated ATP production in both in vivo and in vitro experimental models. Cumulatively, our studies demonstrate for the first time the mechanism of CORM-A1-mediated amelioration of pro-atherogenic manifestations through inhibition of miR-34a-5p expression in the atherogenic milieu and consequential rescue of SIRT1-mediated mitochondrial biogenesis and respiration.
Collapse
Affiliation(s)
- Hitarthi S Vyas
- Chronobiology and Metabolic Endocrinology Lab, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, India
| | - Ravirajsinh N Jadeja
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| | - Aliasgar Vohra
- Chronobiology and Metabolic Endocrinology Lab, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, India
| | - Kapil K Upadhyay
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Menaka C Thounaojam
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Ranjitsinh V Devkar
- Chronobiology and Metabolic Endocrinology Lab, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, India
| |
Collapse
|
22
|
San-Millán I. The Key Role of Mitochondrial Function in Health and Disease. Antioxidants (Basel) 2023; 12:antiox12040782. [PMID: 37107158 PMCID: PMC10135185 DOI: 10.3390/antiox12040782] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
The role of mitochondrial function in health and disease has become increasingly recognized, particularly in the last two decades. Mitochondrial dysfunction as well as disruptions of cellular bioenergetics have been shown to be ubiquitous in some of the most prevalent diseases in our society, such as type 2 diabetes, cardiovascular disease, metabolic syndrome, cancer, and Alzheimer's disease. However, the etiology and pathogenesis of mitochondrial dysfunction in multiple diseases have yet to be elucidated, making it one of the most significant medical challenges in our history. However, the rapid advances in our knowledge of cellular metabolism coupled with the novel understanding at the molecular and genetic levels show tremendous promise to one day elucidate the mysteries of this ancient organelle in order to treat it therapeutically when needed. Mitochondrial DNA mutations, infections, aging, and a lack of physical activity have been identified to be major players in mitochondrial dysfunction in multiple diseases. This review examines the complexities of mitochondrial function, whose ancient incorporation into eukaryotic cells for energy purposes was key for the survival and creation of new species. Among these complexities, the tightly intertwined bioenergetics derived from the combustion of alimentary substrates and oxygen are necessary for cellular homeostasis, including the production of reactive oxygen species. This review discusses different etiological mechanisms by which mitochondria could become dysregulated, determining the fate of multiple tissues and organs and being a protagonist in the pathogenesis of many non-communicable diseases. Finally, physical activity is a canonical evolutionary characteristic of humans that remains embedded in our genes. The normalization of a lack of physical activity in our modern society has led to the perception that exercise is an "intervention". However, physical activity remains the modus vivendi engrained in our genes and being sedentary has been the real intervention and collateral effect of modern societies. It is well known that a lack of physical activity leads to mitochondrial dysfunction and, hence, it probably becomes a major etiological factor of many non-communicable diseases affecting modern societies. Since physical activity remains the only stimulus we know that can improve and maintain mitochondrial function, a significant emphasis on exercise promotion should be imperative in order to prevent multiple diseases. Finally, in populations with chronic diseases where mitochondrial dysfunction is involved, an individualized exercise prescription should be crucial for the "metabolic rehabilitation" of many patients. From lessons learned from elite athletes (the perfect human machines), it is possible to translate and apply multiple concepts to the betterment of populations with chronic diseases.
Collapse
Affiliation(s)
- Iñigo San-Millán
- Department of Human Physiology and Nutrition, University of Colorado, Colorado Springs, CO 80198, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
23
|
The Drp1-Mediated Mitochondrial Fission Protein Interactome as an Emerging Core Player in Mitochondrial Dynamics and Cardiovascular Disease Therapy. Int J Mol Sci 2023; 24:ijms24065785. [PMID: 36982862 PMCID: PMC10057413 DOI: 10.3390/ijms24065785] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Mitochondria, the membrane-bound cell organelles that supply most of the energy needed for cell function, are highly regulated, dynamic organelles bearing the ability to alter both form and functionality rapidly to maintain normal physiological events and challenge stress to the cell. This amazingly vibrant movement and distribution of mitochondria within cells is controlled by the highly coordinated interplay between mitochondrial dynamic processes and fission and fusion events, as well as mitochondrial quality-control processes, mainly mitochondrial autophagy (also known as mitophagy). Fusion connects and unites neighboring depolarized mitochondria to derive a healthy and distinct mitochondrion. In contrast, fission segregates damaged mitochondria from intact and healthy counterparts and is followed by selective clearance of the damaged mitochondria via mitochondrial specific autophagy, i.e., mitophagy. Hence, the mitochondrial processes encompass all coordinated events of fusion, fission, mitophagy, and biogenesis for sustaining mitochondrial homeostasis. Accumulated evidence strongly suggests that mitochondrial impairment has already emerged as a core player in the pathogenesis, progression, and development of various human diseases, including cardiovascular ailments, the leading causes of death globally, which take an estimated 17.9 million lives each year. The crucial factor governing the fission process is the recruitment of dynamin-related protein 1 (Drp1), a GTPase that regulates mitochondrial fission, from the cytosol to the outer mitochondrial membrane in a guanosine triphosphate (GTP)-dependent manner, where it is oligomerized and self-assembles into spiral structures. In this review, we first aim to describe the structural elements, functionality, and regulatory mechanisms of the key mitochondrial fission protein, Drp1, and other mitochondrial fission adaptor proteins, including mitochondrial fission 1 (Fis1), mitochondrial fission factor (Mff), mitochondrial dynamics 49 (Mid49), and mitochondrial dynamics 51 (Mid51). The core area of the review focuses on the recent advances in understanding the role of the Drp1-mediated mitochondrial fission adaptor protein interactome to unravel the missing links of mitochondrial fission events. Lastly, we discuss the promising mitochondria-targeted therapeutic approaches that involve fission, as well as current evidence on Drp1-mediated fission protein interactions and their critical roles in the pathogeneses of cardiovascular diseases (CVDs).
Collapse
|
24
|
Onnis C, Muscogiuri G, Cademartiri F, Fanni D, Faa G, Gerosa C, Mannelli L, Suri JS, Sironi S, Montisci R, Saba L. Non-invasive coronary imaging in elderly population. Eur J Radiol 2023; 162:110794. [PMID: 37001255 DOI: 10.1016/j.ejrad.2023.110794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Age is a non-modifiable cardiovascular risk factor, which leads to development and progression of chronic conditions, such as coronary artery disease, by promoting atherosclerosis. Aging is responsible for morphological structure changes of the coronary arteries and specific atherosclerotic plaque features, which can be studied with non-invasive coronary imaging techniques, particularly coronary CT angiography. The aim of this review is to evaluate current knowledge on this technique applied to the elderly population, and to describe CAD manifestation and plaque features of coronary atherosclerosis in this particular set of patients. We also discuss the clinical implication of frailty assessment and customization of diagnostic strategies in order to shift the approach from disease-centered to patient-centered care.
Collapse
|
25
|
Xia Y, Zhang X, An P, Luo J, Luo Y. Mitochondrial Homeostasis in VSMCs as a Central Hub in Vascular Remodeling. Int J Mol Sci 2023; 24:ijms24043483. [PMID: 36834896 PMCID: PMC9961025 DOI: 10.3390/ijms24043483] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Vascular remodeling is a common pathological hallmark of many cardiovascular diseases. Vascular smooth muscle cells (VSMCs) are the predominant cell type lining the tunica media and play a crucial role in maintaining aortic morphology, integrity, contraction and elasticity. Their abnormal proliferation, migration, apoptosis and other activities are tightly associated with a spectrum of structural and functional alterations in blood vessels. Emerging evidence suggests that mitochondria, the energy center of VSMCs, participate in vascular remodeling through multiple mechanisms. For example, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α)-mediated mitochondrial biogenesis prevents VSMCs from proliferation and senescence. The imbalance between mitochondrial fusion and fission controls the abnormal proliferation, migration and phenotypic transformation of VSMCs. Guanosine triphosphate-hydrolyzing enzymes, including mitofusin 1 (MFN1), mitofusin 2 (MFN2), optic atrophy protein 1 (OPA1) and dynamin-related protein 1 (DRP1), are crucial for mitochondrial fusion and fission. In addition, abnormal mitophagy accelerates the senescence and apoptosis of VSMCs. PINK/Parkin and NIX/BINP3 pathways alleviate vascular remodeling by awakening mitophagy in VSMCs. Mitochondrial DNA (mtDNA) damage destroys the respiratory chain of VSMCs, resulting in excessive ROS production and decreased ATP levels, which are related to the proliferation, migration and apoptosis of VSMCs. Thus, maintaining mitochondrial homeostasis in VSMCs is a possible way to relieve pathologic vascular remodeling. This review aims to provide an overview of the role of mitochondria homeostasis in VSMCs during vascular remodeling and potential mitochondria-targeted therapies.
Collapse
|
26
|
Song J, Farris D, Ariza P, Moorjani S, Varghese M, Blin M, Chen J, Tyrrell D, Zhang M, Singer K, Salmon M, Goldstein DR. Age-associated adipose tissue inflammation promotes monocyte chemotaxis and enhances atherosclerosis. Aging Cell 2023; 22:e13783. [PMID: 36683460 PMCID: PMC9924943 DOI: 10.1111/acel.13783] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 10/31/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023] Open
Abstract
Although aging enhances atherosclerosis, we do not know if this occurs via alterations in circulating immune cells, lipid metabolism, vasculature, or adipose tissue. Here, we examined whether aging exerts a direct pro-atherogenic effect on adipose tissue in mice. After demonstrating that aging augmented the inflammatory profile of visceral but not subcutaneous adipose tissue, we transplanted visceral fat from young or aged mice onto the right carotid artery of Ldlr-/- recipients. Aged fat transplants not only increased atherosclerotic plaque size with increased macrophage numbers in the adjacent carotid artery, but also in distal vascular territories, indicating that aging of the adipose tissue enhances atherosclerosis via secreted factors. By depleting macrophages from the visceral fat, we identified that adipose tissue macrophages are major contributors of the secreted factors. To identify these inflammatory factors, we found that aged fat transplants secreted increased levels of the inflammatory mediators TNFα, CXCL2, and CCL2, which synergized to promote monocyte chemotaxis. Importantly, the combined blockade of these inflammatory mediators impeded the ability of aged fat transplants to enhance atherosclerosis. In conclusion, our study reveals that aging enhances atherosclerosis via increased inflammation of visceral fat. Our study suggests that future therapies targeting the visceral fat may reduce atherosclerosis disease burden in the expanding older population.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Diana Farris
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Paola Ariza
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Smriti Moorjani
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Mita Varghese
- Department of Pediatrics, Division of EndocrinologyUniversity of MichiganAnn ArborMichiganUSA
| | - Muriel Blin
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Judy Chen
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel Tyrrell
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Min Zhang
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Kanakadurga Singer
- Department of Pediatrics, Division of EndocrinologyUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Morgan Salmon
- Department of Cardiac SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
27
|
Li Y, Yang S, Jin X, Li D, Lu J, Wang X, Wu M. Mitochondria as novel mediators linking gut microbiota to atherosclerosis that is ameliorated by herbal medicine: A review. Front Pharmacol 2023; 14:1082817. [PMID: 36733506 PMCID: PMC9886688 DOI: 10.3389/fphar.2023.1082817] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Atherosclerosis (AS) is the main cause of cardiovascular disease (CVD) and is characterized by endothelial damage, lipid deposition, and chronic inflammation. Gut microbiota plays an important role in the occurrence and development of AS by regulating host metabolism and immunity. As human mitochondria evolved from primordial bacteria have homologous characteristics, they are attacked by microbial pathogens as target organelles, thus contributing to energy metabolism disorders, oxidative stress, and apoptosis. Therefore, mitochondria may be a key mediator of intestinal microbiota disorders and AS aggravation. Microbial metabolites, such as short-chain fatty acids, trimethylamine, hydrogen sulfide, and bile acids, also affect mitochondrial function, including mtDNA mutation, oxidative stress, and mitophagy, promoting low-grade inflammation. This further damages cellular homeostasis and the balance of innate immunity, aggravating AS. Herbal medicines and their monomers can effectively ameliorate the intestinal flora and their metabolites, improve mitochondrial function, and inhibit atherosclerotic plaques. This review focuses on the interaction between gut microbiota and mitochondria in AS and explores a therapeutic strategy for restoring mitochondrial function and intestinal microbiota disorders using herbal medicines, aiming to provide new insights for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yujuan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Beijing University of Chinese Medicine, Beijing, China
| | - Xinyue Wang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Min Wu,
| |
Collapse
|
28
|
Mitochondrial Dysfunction: The Hidden Player in the Pathogenesis of Atherosclerosis? Int J Mol Sci 2023; 24:ijms24021086. [PMID: 36674602 PMCID: PMC9861427 DOI: 10.3390/ijms24021086] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a multifactorial inflammatory pathology that involves metabolic processes. Improvements in therapy have drastically reduced the prognosis of cardiovascular disease. Nevertheless, a significant residual risk is still relevant, and is related to unmet therapeutic targets. Endothelial dysfunction and lipid infiltration are the primary causes of atherosclerotic plaque progression. In this contest, mitochondrial dysfunction can affect arterial wall cells, in particular macrophages, smooth muscle cells, lymphocytes, and endothelial cells, causing an increase in reactive oxygen species (ROS), leading to oxidative stress, chronic inflammation, and intracellular lipid deposition. The detection and characterization of mitochondrial DNA (mtDNA) is crucial for assessing mitochondrial defects and should be considered the goal for new future therapeutic interventions. In this review, we will focus on a new idea, based on the analysis of data from many research groups, namely the link between mitochondrial impairment and endothelial dysfunction and, in particular, its effect on atherosclerosis and aging. Therefore, we discuss known and novel mitochondria-targeting therapies in the contest of atherosclerosis.
Collapse
|
29
|
Qu K, Yan F, Qin X, Zhang K, He W, Dong M, Wu G. Mitochondrial dysfunction in vascular endothelial cells and its role in atherosclerosis. Front Physiol 2022; 13:1084604. [PMID: 36605901 PMCID: PMC9807884 DOI: 10.3389/fphys.2022.1084604] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
The mitochondria are essential organelles that generate large amounts of ATP via the electron transport chain (ECT). Mitochondrial dysfunction causes reactive oxygen species accumulation, energy stress, and cell death. Endothelial mitochondrial dysfunction is an important factor causing abnormal function of the endothelium, which plays a central role during atherosclerosis development. Atherosclerosis-related risk factors, including high glucose levels, hypertension, ischemia, hypoxia, and diabetes, promote mitochondrial dysfunction in endothelial cells. This review summarizes the physiological and pathophysiological roles of endothelial mitochondria in endothelial function and atherosclerosis.
Collapse
Affiliation(s)
- Kai Qu
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Fang Yan
- Department of Geriatrics, Geriatric Diseases Institute of Chengdu, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China,Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Xian Qin
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Kun Zhang
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Wen He
- Department of Geriatrics, Clinical trial center, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China,*Correspondence: Mingqing Dong, ; Guicheng Wu,
| | - Guicheng Wu
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,*Correspondence: Mingqing Dong, ; Guicheng Wu,
| |
Collapse
|
30
|
Liu Y, Huang Y, Xu C, An P, Luo Y, Jiao L, Luo J, Li Y. Mitochondrial Dysfunction and Therapeutic Perspectives in Cardiovascular Diseases. Int J Mol Sci 2022; 23:16053. [PMID: 36555691 PMCID: PMC9788331 DOI: 10.3390/ijms232416053] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/21/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
High mortality rates due to cardiovascular diseases (CVDs) have attracted worldwide attention. It has been reported that mitochondrial dysfunction is one of the most important mechanisms affecting the pathogenesis of CVDs. Mitochondrial DNA (mtDNA) mutations may result in impaired oxidative phosphorylation (OXPHOS), abnormal respiratory chains, and ATP production. In dysfunctional mitochondria, the electron transport chain (ETC) is uncoupled and the energy supply is reduced, while reactive oxygen species (ROS) production is increased. Here, we discussed and analyzed the relationship between mtDNA mutations, impaired mitophagy, decreased OXPHOS, elevated ROS, and CVDs from the perspective of mitochondrial dysfunction. Furthermore, we explored current potential therapeutic strategies for CVDs by eliminating mtDNA mutations (e.g., mtDNA editing and mitochondrial replacement), enhancing mitophagy, improving OXPHOS capacity (e.g., supplement with NAD+, nicotinamide riboside (NR), nicotinamide mononucleotide (NMN), and nano-drug delivery), and reducing ROS (e.g., supplement with Coenzyme Q10 and other antioxidants), and dissected their respective advantages and limitations. In fact, some therapeutic strategies are still a long way from achieving safe and effective clinical treatment. Although establishing effective and safe therapeutic strategies for CVDs remains challenging, starting from a mitochondrial perspective holds bright prospects.
Collapse
Affiliation(s)
- Yu Liu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Yuejia Huang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Chong Xu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Peng An
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yongting Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Lei Jiao
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Junjie Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yongzhi Li
- China Astronaut Research and Training Center, Beijing 100094, China
| |
Collapse
|
31
|
Li YJ, Jin X, Li D, Lu J, Zhang XN, Yang SJ, Zhao YX, Wu M. New insights into vascular aging: Emerging role of mitochondria function. Biomed Pharmacother 2022; 156:113954. [DOI: 10.1016/j.biopha.2022.113954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
|
32
|
Choudhury D, Rong N, Ikhapoh I, Rajabian N, Tseropoulos G, Wu Y, Mehrotra P, Thiyagarajan R, Shahini A, Seldeen KL, Troen B, Lei P, Andreadis ST. Inhibition of glutaminolysis restores mitochondrial function in senescent stem cells. Cell Rep 2022; 41:111744. [PMID: 36450260 PMCID: PMC9809151 DOI: 10.1016/j.celrep.2022.111744] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/07/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Mitochondrial dysfunction, a hallmark of aging, has been associated with the onset of aging phenotypes and age-related diseases. Here, we report that impaired mitochondrial function is associated with increased glutamine catabolism in senescent human mesenchymal stem cells (MSCs) and myofibroblasts derived from patients suffering from Hutchinson-Gilford progeria syndrome. Increased glutaminase (GLS1) activity accompanied by loss of urea transporter SLC14A1 induces urea accumulation, mitochondrial dysfunction, and DNA damage. Conversely, blocking GLS1 activity restores mitochondrial function and leads to amelioration of aging hallmarks. Interestingly, GLS1 expression is regulated through the JNK pathway, as demonstrated by chemical and genetic inhibition. In agreement with our in vitro findings, tissues isolated from aged or progeria mice display increased urea accumulation and GLS1 activity, concomitant with declined mitochondrial function. Inhibition of glutaminolysis in progeria mice improves mitochondrial respiratory chain activity, suggesting that targeting glutaminolysis may be a promising strategy for restoring age-associated loss of mitochondrial function.
Collapse
Affiliation(s)
- Debanik Choudhury
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Na Rong
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Izuagie Ikhapoh
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Nika Rajabian
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Yulun Wu
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Pihu Mehrotra
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Ramkumar Thiyagarajan
- Department of Medicine, Division of Geriatrics and Palliative medicine, Buffalo, NY 14203
| | - Aref Shahini
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Kenneth L. Seldeen
- Department of Medicine, Division of Geriatrics and Palliative medicine, Buffalo, NY 14203
| | - Bruce Troen
- Department of Medicine, Division of Geriatrics and Palliative medicine, Buffalo, NY 14203
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260,Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14260,Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY 14263,Center for Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, Buffalo, NY 14260,Address for all Correspondence: Stelios T. Andreadis, Ph.D., SUNY Distinguished Professor, Bioengineering Laboratory, 908 Furnas Hall, Department of Chemical and Biological Engineering, Department of Biomedical Engineering, and Center of Excellence in Bioinformatics and Life Sciences, Center for Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA, Tel: (716) 645-1202, Fax: (716) 645-3822,
| |
Collapse
|
33
|
Wang H, Sun Y, Pi C, Yu X, Gao X, Zhang C, Sun H, Zhang H, Shi Y, He X. Nicotinamide Mononucleotide Supplementation Improves Mitochondrial Dysfunction and Rescues Cellular Senescence by NAD +/Sirt3 Pathway in Mesenchymal Stem Cells. Int J Mol Sci 2022; 23:ijms232314739. [PMID: 36499074 PMCID: PMC9738479 DOI: 10.3390/ijms232314739] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
In vitro expansion-mediated replicative senescence has severely limited the clinical applications of mesenchymal stem cells (MSCs). Accumulating studies manifested that nicotinamide adenine dinucleotide (NAD+) depletion is closely related to stem cell senescence and mitochondrial metabolism disorder. Promoting NAD+ level is considered as an effective way to delay aging. Previously, we have confirmed that nicotinamide mononucleotide (NMN), a precursor of NAD+, can alleviate NAD+ deficiency-induced MSC senescence. However, whether NMN can attenuate MSC senescence and its underlying mechanisms are still incompletely clear. The present study herein showed that late passage (LP) MSCs displayed lower NAD+ content, reduced Sirt3 expression and mitochondrial dysfunction. NMN supplementation leads to significant increase in intracellular NAD+ level, NAD+/ NADH ratio, Sirt3 expression, as well as ameliorated mitochondrial function and rescued senescent MSCs. Additionally, Sirt3 over-expression relieved mitochondrial dysfunction, and retrieved senescence-associated phenotypic features in LP MSCs. Conversely, inhibition of Sirt3 activity via a selective Sirt3 inhibitor 3-TYP in early passage (EP) MSCs resulted in aggravated cellular senescence and abnormal mitochondrial function. Furthermore, NMN administration also improves 3-TYP-induced disordered mitochondrial function and cellular senescence in EP MSCs. Collectively, NMN replenishment alleviates mitochondrial dysfunction and rescues MSC senescence through mediating NAD+/Sirt3 pathway, possibly providing a novel mechanism for MSC senescence and a promising strategy for anti-aging pharmaceuticals.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xu He
- Correspondence: ; Tel.: +86-135-0430-7430
| |
Collapse
|
34
|
Liu H, Liu Y, Wang H, Zhao Q, Zhang T, Xie S, Liu Y, Tang Y, Peng Q, Pang W, Yao W, Zhou J. Geometric Constraints Regulate Energy Metabolism and Cellular Contractility in Vascular Smooth Muscle Cells by Coordinating Mitochondrial DNA Methylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203995. [PMID: 36106364 PMCID: PMC9661866 DOI: 10.1002/advs.202203995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Vascular smooth muscle cells (SMCs) can adapt to changes in cellular geometric cues; however, the underlying mechanisms remain elusive. Using 2D micropatterned substrates to engineer cell geometry, it is found that in comparison with an elongated geometry, a square-shaped geometry causes the nuclear-to-cytoplasmic redistribution of DNA methyltransferase 1 (DNMT1), hypermethylation of mitochondrial DNA (mtDNA), repression of mtDNA gene transcription, and impairment of mitochondrial function. Using irregularly arranged versus circumferentially aligned vascular grafts to control cell geometry in 3D growth, it is demonstrated that cell geometry, mtDNA methylation, and vessel contractility are closely related. DNMT1 redistribution is found to be dependent on the phosphoinositide 3-kinase and protein kinase B (AKT) signaling pathways. Cell elongation activates cytosolic phospholipase A2, a nuclear mechanosensor that, when inhibited, hinders AKT phosphorylation, DNMT1 nuclear accumulation, and energy production. The findings of this study provide insights into the effects of cell geometry on SMC function and its potential implications in the optimization of vascular grafts.
Collapse
Affiliation(s)
- Han Liu
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Key Laboratory of Molecular Cardiovascular ScienceMinistry of EducationBeijing100191P. R. China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191P. R. China
| | - Yuefeng Liu
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Key Laboratory of Molecular Cardiovascular ScienceMinistry of EducationBeijing100191P. R. China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191P. R. China
| | - He Wang
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of EducationCollaborative Innovation Center of Chemical Science and Engineering (Tianjin)Nankai UniversityTianjin300071P. R. China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of EducationCollaborative Innovation Center of Chemical Science and Engineering (Tianjin)Nankai UniversityTianjin300071P. R. China
| | - Tao Zhang
- Department of Vascular SurgeryPeking University People's HospitalBeijing100044P. R. China
| | - Si‐an Xie
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Key Laboratory of Molecular Cardiovascular ScienceMinistry of EducationBeijing100191P. R. China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191P. R. China
| | - Yueqi Liu
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Key Laboratory of Molecular Cardiovascular ScienceMinistry of EducationBeijing100191P. R. China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191P. R. China
| | - Yuanjun Tang
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Key Laboratory of Molecular Cardiovascular ScienceMinistry of EducationBeijing100191P. R. China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191P. R. China
| | - Qin Peng
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518132P. R. China
| | - Wei Pang
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Weijuan Yao
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Jing Zhou
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences; Hemorheology CenterSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Key Laboratory of Molecular Cardiovascular ScienceMinistry of EducationBeijing100191P. R. China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191P. R. China
| |
Collapse
|
35
|
Lozhkin A, Vendrov AE, Ramos-Mondragón R, Canugovi C, Stevenson MD, Herron TJ, Hummel SL, Figueroa CA, Bowles DE, Isom LL, Runge MS, Madamanchi NR. Mitochondrial oxidative stress contributes to diastolic dysfunction through impaired mitochondrial dynamics. Redox Biol 2022; 57:102474. [PMID: 36183542 PMCID: PMC9530618 DOI: 10.1016/j.redox.2022.102474] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/11/2022] [Indexed: 11/25/2022] Open
Abstract
Diastolic dysfunction (DD) underlies heart failure with preserved ejection fraction (HFpEF), a clinical syndrome associated with aging that is becoming more prevalent. Despite extensive clinical studies, no effective treatment exists for HFpEF. Recent findings suggest that oxidative stress contributes to the pathophysiology of DD, but molecular mechanisms underpinning redox-sensitive cardiac remodeling in DD remain obscure. Using transgenic mice with mitochondria-targeted NOX4 overexpression (Nox4TG618) as a model, we demonstrate that NOX4-dependent mitochondrial oxidative stress induces DD in mice as measured by increased E/E', isovolumic relaxation time, Tau Glantz and reduced dP/dtmin while EF is preserved. In Nox4TG618 mice, fragmentation of cardiomyocyte mitochondria, increased DRP1 phosphorylation, decreased expression of MFN2, and a higher percentage of apoptotic cells in the myocardium are associated with lower ATP-driven and maximal mitochondrial oxygen consumption rates, a decrease in respiratory reserve, and a decrease in citrate synthase and Complex I activities. Transgenic mice have an increased concentration of TGFβ and osteopontin in LV lysates, as well as MCP-1 in plasma, which correlates with a higher percentage of LV myocardial periostin- and ACTA2-positive cells compared with wild-type mice. Accordingly, the levels of ECM as measured by Picrosirius Red staining as well as interstitial deposition of collagen I are elevated in the myocardium of Nox4TG618 mice. The LV tissue of Nox4TG618 mice also exhibited increased ICaL current, calpain 2 expression, and altered/disrupted Z-disc structure. As it pertains to human pathology, similar changes were found in samples of LV from patients with DD. Finally, treatment with GKT137831, a specific NOX1 and NOX4 inhibitor, or overexpression of mCAT attenuated myocardial fibrosis and prevented DD in the Nox4TG618 mice. Together, our results indicate that mitochondrial oxidative stress contributes to DD by causing mitochondrial dysfunction, impaired mitochondrial dynamics, increased synthesis of pro-inflammatory and pro-fibrotic cytokines, activation of fibroblasts, and the accumulation of extracellular matrix, which leads to interstitial fibrosis and passive stiffness of the myocardium. Further, mitochondrial oxidative stress increases cardiomyocyte Ca2+ influx, which worsens CM relaxation and raises the LV filling pressure in conjunction with structural proteolytic damage.
Collapse
Affiliation(s)
- Andrey Lozhkin
- 1150 West Medical Center Drive, 7200 Medical Science Research Building III, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48019, USA
| | - Aleksandr E Vendrov
- 1150 West Medical Center Drive, 7200 Medical Science Research Building III, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48019, USA
| | - R Ramos-Mondragón
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Chandrika Canugovi
- 1150 West Medical Center Drive, 7200 Medical Science Research Building III, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48019, USA
| | - Mark D Stevenson
- 1150 West Medical Center Drive, 7200 Medical Science Research Building III, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48019, USA
| | - Todd J Herron
- Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI, 48109, USA
| | - Scott L Hummel
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Ann Arbor Veterans Affairs Health System, Ann Arbor, MI, USA
| | - C Alberto Figueroa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Dawn E Bowles
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Marschall S Runge
- 1150 West Medical Center Drive, 7200 Medical Science Research Building III, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48019, USA
| | - Nageswara R Madamanchi
- 1150 West Medical Center Drive, 7200 Medical Science Research Building III, Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48019, USA.
| |
Collapse
|
36
|
Wang M, Pang Y, Guo Y, Tian L, Liu Y, Shen C, Liu M, Meng Y, Cai Z, Wang Y, Zhao W. Metabolic reprogramming: A novel therapeutic target in diabetic kidney disease. Front Pharmacol 2022; 13:970601. [PMID: 36120335 PMCID: PMC9479190 DOI: 10.3389/fphar.2022.970601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common microvascular complications of diabetes mellitus. However, the pathological mechanisms contributing to DKD are multifactorial and poorly understood. Diabetes is characterized by metabolic disorders that can bring about a series of changes in energy metabolism. As the most energy-consuming organs secondary only to the heart, the kidneys must maintain energy homeostasis. Aberrations in energy metabolism can lead to cellular dysfunction or even death. Metabolic reprogramming, a shift from mitochondrial oxidative phosphorylation to glycolysis and its side branches, is thought to play a critical role in the development and progression of DKD. This review focuses on the current knowledge about metabolic reprogramming and the role it plays in DKD development. The underlying etiologies, pathological damages in the involved cells, and potential molecular regulators of metabolic alterations are also discussed. Understanding the role of metabolic reprogramming in DKD may provide novel therapeutic approaches to delay its progression to end-stage renal disease.
Collapse
|
37
|
Wang T, Cao Y, Zhang H, Wang Z, Man CH, Yang Y, Chen L, Xu S, Yan X, Zheng Q, Wang Y. COVID-19 metabolism: Mechanisms and therapeutic targets. MedComm (Beijing) 2022; 3:e157. [PMID: 35958432 PMCID: PMC9363584 DOI: 10.1002/mco2.157] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dysregulates antiviral signaling, immune response, and cell metabolism in human body. Viral genome and proteins hijack host metabolic network to support viral biogenesis and propagation. However, the regulatory mechanism of SARS-CoV-2-induced metabolic dysfunction has not been elucidated until recently. Multiomic studies of coronavirus disease 2019 (COVID-19) revealed an intensive interaction between host metabolic regulators and viral proteins. SARS-CoV-2 deregulated cellular metabolism in blood, intestine, liver, pancreas, fat, and immune cells. Host metabolism supported almost every stage of viral lifecycle. Strikingly, viral proteins were found to interact with metabolic enzymes in different cellular compartments. Biochemical and genetic assays also identified key regulatory nodes and metabolic dependencies of viral replication. Of note, cholesterol metabolism, lipid metabolism, and glucose metabolism are broadly involved in viral lifecycle. Here, we summarized the current understanding of the hallmarks of COVID-19 metabolism. SARS-CoV-2 infection remodels host cell metabolism, which in turn modulates viral biogenesis and replication. Remodeling of host metabolism creates metabolic vulnerability of SARS-CoV-2 replication, which could be explored to uncover new therapeutic targets. The efficacy of metabolic inhibitors against COVID-19 is under investigation in several clinical trials. Ultimately, the knowledge of SARS-CoV-2-induced metabolic reprogramming would accelerate drug repurposing or screening to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Tianshi Wang
- Shanghai Key Laboratory for Tumor Microenvironment and InflammationDepartment of Biochemistry and Molecular Cell BiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Cao
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Haiyan Zhang
- Bai Jia Obstetrics and Gynecology HospitalShanghaiChina
| | - Zihao Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| | - Cheuk Him Man
- Division of HematologyDepartment of MedicineUniversity of Hong KongPokfulamHong Kong, China
| | - Yunfan Yang
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Lingchao Chen
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersShanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationNeurosurgical Institute of Fudan UniversityShanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Shuangnian Xu
- Department of HematologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Xiaojing Yan
- Department of HematologyThe First Affiliated Hospital of China Medical UniversityShenyangChina
| | - Quan Zheng
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Ping Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| |
Collapse
|
38
|
Chen Y, Qin W, Li L, Wu P, Wei D. Mitophagy: Critical Role in Atherosclerosis Progression. DNA Cell Biol 2022; 41:851-860. [PMID: 36036955 DOI: 10.1089/dna.2022.0249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Autophagy maintains intracellular homeostasis in the cardiovascular system, including in cardiomyocytes, endothelial cells (ECs), and arterial smooth muscle cells. Mitophagy, a selective autophagy that specifically removes damaged and dysfunctional mitochondria, is particularly important for cardiovascular homeostasis. Dysfunctional mitophagy contributes to cardiovascular disease, particularly atherosclerosis (AS). This review focuses on the advances of regulator mechanisms of mitophagy and its potential roles in AS. The findings are beneficial to understanding the pathological processes of atherosclerotic lesions and provide new ideas for the prevention and clinical treatment of AS.
Collapse
Affiliation(s)
- Yanmei Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenhua Qin
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Lu Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Peng Wu
- Yueyang Maternal-Child Medicine Health Hospital Hunan, Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women & Children's Medical Center, Yueyang, China.,Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China.,Yueyang Maternal-Child Medicine Health Hospital Hunan, Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women & Children's Medical Center, Yueyang, China
| |
Collapse
|
39
|
Molaei A, Molaei E, Sadeghnia H, Hayes AW, Karimi G. LKB1: An emerging therapeutic target for cardiovascular diseases. Life Sci 2022; 306:120844. [PMID: 35907495 DOI: 10.1016/j.lfs.2022.120844] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 10/16/2022]
Abstract
Cardiovascular diseases (CVDs) are currently the most common cause of morbidity and mortality worldwide. Experimental studies suggest that liver kinase B1 (LKB1) plays an important role in the heart. Several studies have shown that cardiomyocyte-specific LKB1 deletion leads to hypertrophic cardiomyopathy, left ventricular contractile dysfunction, and an increased risk of atrial fibrillation. In addition, the cardioprotective effects of several medicines and natural compounds, including metformin, empagliflozin, bexarotene, and resveratrol, have been reported to be associated with LKB1 activity. LKB1 limits the size of the damaged myocardial area by modifying cellular metabolism, enhancing the antioxidant system, suppressing hypertrophic signals, and inducing mild autophagy, which are all primarily mediated by the AMP-activated protein kinase (AMPK) energy sensor. LKB1 also improves myocardial efficiency by modulating the function of contractile proteins, regulating the expression of electrical channels, and increasing vascular dilatation. Considering these properties, stimulation of LKB1 signaling offers a promising approach in the prevention and treatment of heart diseases.
Collapse
Affiliation(s)
- Ali Molaei
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hamidreza Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran..
| |
Collapse
|
40
|
Role of Oxidative Stress in the Pathogenesis of Atherothrombotic Diseases. Antioxidants (Basel) 2022; 11:antiox11071408. [PMID: 35883899 PMCID: PMC9312358 DOI: 10.3390/antiox11071408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress is generated by the imbalance between reactive oxygen species (ROS) formation and antioxidant scavenger system’s activity. Increased ROS, such as superoxide anion, hydrogen peroxide, hydroxyl radical and peroxynitrite, likely contribute to the development and complications of atherosclerotic cardiovascular diseases (ASCVD). In genetically modified mouse models of atherosclerosis, the overexpression of ROS-generating enzymes and uncontrolled ROS formation appear to be associated with accelerated atherosclerosis. Conversely, the overexpression of ROS scavenger systems reduces or stabilizes atherosclerotic lesions, depending on the genetic background of the mouse model. In humans, higher levels of circulating biomarkers derived from the oxidation of lipids (8-epi-prostaglandin F2α, and malondialdehyde), as well as proteins (oxidized low-density lipoprotein, nitrotyrosine, protein carbonyls, advanced glycation end-products), are increased in conditions of high cardiovascular risk or overt ASCVD, and some oxidation biomarkers have been reported as independent predictors of ASCVD in large observational cohorts. In animal models, antioxidant supplementation with melatonin, resveratrol, Vitamin E, stevioside, acacetin and n-polyunsaturated fatty acids reduced ROS and attenuated atherosclerotic lesions. However, in humans, evidence from large, placebo-controlled, randomized trials or prospective studies failed to show any athero-protective effect of antioxidant supplementation with different compounds in different CV settings. However, the chronic consumption of diets known to be rich in antioxidant compounds (e.g., Mediterranean and high-fish diet), has shown to reduce ASCVD over decades. Future studies are needed to fill the gap between the data and targets derived from studies in animals and their pathogenetic and therapeutic significance in human ASCVD.
Collapse
|
41
|
Karnewar S, Pulipaka S, Katta S, Panuganti D, Neeli PK, Thennati R, Jerald MK, Kotamraju S. Mitochondria-targeted esculetin mitigates atherosclerosis in the setting of aging via the modulation of SIRT1-mediated vascular cell senescence and mitochondrial function in Apoe mice. Atherosclerosis 2022; 356:28-40. [DOI: 10.1016/j.atherosclerosis.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
|
42
|
Riascos-Bernal DF, Maira A, Sibinga NES. The Atypical Cadherin FAT1 Limits Mitochondrial Respiration and Proliferation of Vascular Smooth Muscle Cells. Front Cardiovasc Med 2022; 9:905717. [PMID: 35647082 PMCID: PMC9130956 DOI: 10.3389/fcvm.2022.905717] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/19/2022] [Indexed: 12/23/2022] Open
Abstract
Smooth muscle cells contribute to cardiovascular disease, the leading cause of death worldwide. The capacity of these cells to undergo phenotypic switching in mature arteries of the systemic circulation underlies their pathogenic role in atherosclerosis and restenosis, among other vascular diseases. Growth factors and cytokines, extracellular matrix components, regulation of gene expression, neuronal influences, and mechanical forces contribute to smooth muscle cell phenotypic switching. Comparatively little is known about cell metabolism in this process. Studies of cancer and endothelial cell biology have highlighted the importance of cellular metabolic processes for phenotypic transitions that accompany tumor growth and angiogenesis. However, the understanding of cell metabolism during smooth muscle cell phenotypic modulation is incipient. Studies of the atypical cadherin FAT1, which is strongly upregulated in smooth muscle cells in response to arterial injury, suggest that it has important and distinctive functions in this context, mediating control of both smooth muscle cell mitochondrial metabolism and cell proliferation. Here we review the progress made in understanding how FAT1 affects the smooth muscle cell phenotype, highlighting the significance of FAT1 as a processed protein and unexpected regulator of mitochondrial respiration. These mechanisms suggest how a transmembrane protein may relay signals from the extracellular milieu to mitochondria to control metabolic activity during smooth muscle cell phenotypic switching.
Collapse
Affiliation(s)
- Dario F Riascos-Bernal
- Department of Medicine (Cardiology) and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alishba Maira
- Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Nicholas E S Sibinga
- Department of Medicine (Cardiology) and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
43
|
Becker PH, Le Guillou E, Duque M, Blondel A, Gons C, Ben Souna H, Imbard A, Fournier N, Gaignard P, Thérond P. Cholesterol accumulation induced by acetylated LDL exposure modifies the enzymatic activities of the TCA cycle without impairing the respiratory chain functionality in macrophages. Biochimie 2022; 200:87-98. [PMID: 35618159 DOI: 10.1016/j.biochi.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/31/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022]
Abstract
The unregulated uptake of modified low-density lipoproteins (LDL) by macrophages leads to foam cell formation, promoting atherosclerotic plaque progression. The cholesterol efflux capacity of macrophages by the ATP-Binding Cassette transporters depends on the ATP mitochondrial production. Therefore, the mitochondrial function maintenance is crucial in limiting foam cell formation. Thus, we aimed to investigate the mechanisms involved in the mitochondrial dysfunction that may occur in cholesterol-laden macrophages. We incubated THP-1 macrophages with acetylated LDL (acLDL) to obtain cholesterol-laden cells or with mildly oxidized LDL (oxLDL) to generate cholesterol- and oxidized lipids-laden cells. Cellular cholesterol content was measured in each condition. Mitochondrial function was evaluated by measurement of several markers of energetic metabolism, oxidative phosphorylation, oxidative stress, mitochondrial biogenesis and dynamics. OxLDL-exposed macrophages exhibited a significantly reduced mitochondrial respiration and complexes I and III activities, associated to an oxidative stress state and a reduced mitochondrial DNA copy number. Meanwhile, acLDL-exposed macrophages featured an efficient oxidative phosphorylation despite the decreased activities of aconitase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase. Our study revealed that mitochondrial function was differently impacted according to the nature of modified LDL. Exposure to cholesterol and oxidized lipids carried by oxLDL leads to a mitochondrial dysfunction in macrophages, affecting the mitochondrial respiratory chain functional capacity, whereas the cellular cholesterol enrichment induced by acLDL exposure results in a tricarboxylic acid cycle shunt while maintaining mitochondrial energetic production, reflecting a metabolic adaptation to cholesterol intake. These new mechanistic insights are of direct relevance to the understanding of the mitochondrial dysfunction in foam cells.
Collapse
Affiliation(s)
- Pierre-Hadrien Becker
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France.
| | - Edouard Le Guillou
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Mathilde Duque
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Amélie Blondel
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Camille Gons
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Hajar Ben Souna
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Apolline Imbard
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Necker-Enfants Malades, AP-HP, Laboratoire de Biochimie Métabolique, Paris, 75015, France
| | - Natalie Fournier
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Européen Georges Pompidou, AP-HP, Laboratoire de Biochimie, Paris, 75015, France
| | - Pauline Gaignard
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Patrice Thérond
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| |
Collapse
|
44
|
Wu Y, Jiang T, Hua J, Xiong Z, Dai K, Chen H, Li L, Peng J, Peng X, Zheng Z, Xiong W. PINK1/Parkin-mediated mitophagy in cardiovascular disease: From pathogenesis to novel therapy. Int J Cardiol 2022; 361:61-69. [PMID: 35594994 DOI: 10.1016/j.ijcard.2022.05.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease(CVD)is one of the predominant causes of death and morbidity. Mitochondria play a key role in maintaining cardiac energy metabolism. However, mitochondrial dysfunction leads to excessive production of ROS, resulting in oxidative damage to cardiomyocytes and contributing to a variety of cardiovascular diseases. In such a case, the clearance of impaired mitochondria is necessary. Currently, most studies have indicated an essential role for mitophagy in maintaining cardiac homeostasis and regulating CVD-related metabolic transition. Recent studies have implicated that PTEN-induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy has been implicated in maintaining cardiomyocyte homeostasis. Here, we discuss the physiological and pathological roles of PINK1/Parkin-mediated mitophagy in the cardiovascular system, as well as potential therapeutic strategies based on PINK1/Parkin-mediated mitophagy modulation, which are of great significance for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yanze Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ting Jiang
- Department of Hospital Infection Control, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jinghai Hua
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhiping Xiong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Kai Dai
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hui Chen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lei Li
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jingtian Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenjun Xiong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
45
|
Yang J, Guo Q, Feng X, Liu Y, Zhou Y. Mitochondrial Dysfunction in Cardiovascular Diseases: Potential Targets for Treatment. Front Cell Dev Biol 2022; 10:841523. [PMID: 35646910 PMCID: PMC9140220 DOI: 10.3389/fcell.2022.841523] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are serious public health issues and are responsible for nearly one-third of global deaths. Mitochondrial dysfunction is accountable for the development of most CVDs. Mitochondria produce adenosine triphosphate through oxidative phosphorylation and inevitably generate reactive oxygen species (ROS). Excessive ROS causes mitochondrial dysfunction and cell death. Mitochondria can protect against these damages via the regulation of mitochondrial homeostasis. In recent years, mitochondria-targeted therapy for CVDs has attracted increasing attention. Various studies have confirmed that clinical drugs (β-blockers, angiotensin-converting enzyme inhibitors/angiotensin receptor-II blockers) against CVDs have mitochondrial protective functions. An increasing number of cardiac mitochondrial targets have shown their cardioprotective effects in experimental and clinical studies. Here, we briefly introduce the mechanisms of mitochondrial dysfunction and summarize the progression of mitochondrial targets against CVDs, which may provide ideas for experimental studies and clinical trials.
Collapse
|
46
|
Li A, Gao M, Liu B, Qin Y, Chen L, Liu H, Wu H, Gong G. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis 2022; 13:444. [PMID: 35534453 PMCID: PMC9085840 DOI: 10.1038/s41419-022-04906-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic organelles that participate in ATP generation and involve calcium homeostasis, oxidative stress response, and apoptosis. Dysfunctional or damaged mitochondria could cause serious consequences even lead to cell death. Therefore, maintaining the homeostasis of mitochondria is critical for cellular functions. Mitophagy is a process of selectively degrading damaged mitochondria under mitochondrial toxicity conditions, which plays an essential role in mitochondrial quality control. The abnormal mitophagy that aggravates mitochondrial dysfunction is closely related to the pathogenesis of many diseases. As the myocardium is a highly oxidative metabolic tissue, mitochondria play a central role in maintaining optimal performance of the heart. Dysfunctional mitochondria accumulation is involved in the pathophysiology of cardiovascular diseases, such as myocardial infarction, cardiomyopathy and heart failure. This review discusses the most recent progress on mitophagy and its role in cardiovascular disease.
Collapse
Affiliation(s)
- Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hanyu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Huayan Wu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
47
|
Susser LI, Rayner KJ. Through the layers: how macrophages drive atherosclerosis across the vessel wall. J Clin Invest 2022; 132:157011. [PMID: 35499077 PMCID: PMC9057606 DOI: 10.1172/jci157011] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Leah I. Susser
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
48
|
Moturi S, Ghosh-Choudhary SK, Finkel T. Cardiovascular disease and the biology of aging. J Mol Cell Cardiol 2022; 167:109-117. [PMID: 35421400 DOI: 10.1016/j.yjmcc.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
Abstract
The incidence and prevalence of a wide range of cardiovascular diseases increases as a function of age. This well-established epidemiological relationship suggests that chronological aging might contribute or increase susceptibility to varied conditions such as atherosclerosis, vascular stiffening or heart failure. Here, we explore the mechanistic links that connect both rare and common cardiovascular conditions to the basic biology of aging. These links provide a rational basis to begin to develop a new set of therapeutics targeting the fundamental mechanisms underlying the aging process and suggest that in the near future, age itself might become a modifiable cardiovascular risk factor.
Collapse
Affiliation(s)
- Shria Moturi
- Aging Institute, University of Pittsburgh School of Medicine/UPMC, Pittsburgh, PA 15219, United States of America
| | - Shohini K Ghosh-Choudhary
- Aging Institute, University of Pittsburgh School of Medicine/UPMC, Pittsburgh, PA 15219, United States of America
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine/UPMC, Pittsburgh, PA 15219, United States of America.
| |
Collapse
|
49
|
Oxidative phosphorylation promotes vascular calcification in chronic kidney disease. Cell Death Dis 2022; 13:229. [PMID: 35277475 PMCID: PMC8917188 DOI: 10.1038/s41419-022-04679-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 01/20/2023]
Abstract
Metabolism has been reported to associate with the progression of vascular diseases. However, how vascular calcification in chronic kidney disease (CKD) is regulated by metabolic status remains poorly understood. Using a model of 5/6 nephrectomy, we demonstrated that the aortic tissues of CKD mice had a preference for using oxidative phosphorylation (OXPHOS). Both high phosphate and human uremic serum-stimulated vascular smooth muscle cells (VSMCs) had enhanced mitochondrial respiration capacity, while the glycolysis level was not significantly different. Besides, 2-deoxy-d-glucose (2-DG) exacerbated vascular calcification by upregulating OXPHOS. The activity of cytochrome c oxidase (COX) was higher in the aortic tissue of CKD mice than those of sham-operated mice. Moreover, the expression levels of COX15 were higher in CKD patients with aortic arch calcification (AAC) than those without AAC, and the AAC scores were correlated with the expression level of COX15. Suppressing COX sufficiently attenuated vascular calcification. Our findings verify the relationship between OXPHOS and calcification, and may provide potential therapeutic approaches for vascular calcification in CKD.
Collapse
|
50
|
Przybylska S, Tokarczyk G. Lycopene in the Prevention of Cardiovascular Diseases. Int J Mol Sci 2022; 23:1957. [PMID: 35216071 PMCID: PMC8880080 DOI: 10.3390/ijms23041957] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of human mortality worldwide. Oxidative stress and inflammation are pathophysiological processes involved in the development of CVD. That is why bioactive food ingredients, including lycopene, are so important in their prevention, which seems to be a compound increasingly promoted in the diet of people with cardiovascular problems. Lycopene present in tomatoes and tomato products is responsible not only for their red color but also for health-promoting properties. It is characterized by a high antioxidant potential, the highest among carotenoid pigments. Mainly for this reason, epidemiological studies show a number of favorable properties between the consumption of lycopene in the diet and a reduced risk of cardiovascular disease. While there is also some controversy in research into its protective effects on the cardiovascular system, growing evidence supports its beneficial role for the heart, endothelium, blood vessels, and health. The mechanisms of action of lycopene are now being discovered and may explain some of the contradictions observed in the literature. This review aims to present the current knowledge in recent years on the preventive role of lycopene cardiovascular disorders.
Collapse
Affiliation(s)
- Sylwia Przybylska
- Department of Fish, Plant and Gastronomy Technology, West Pomeranian University of Technology in Szczecin, 71-459 Szczecin, Poland;
| | | |
Collapse
|