1
|
Buchalska B, Kamińska K, Kowara M, Sobiborowicz-Sadowska A, Cudnoch-Jędrzejewska A. Doxorubicin or Epirubicin Versus Liposomal Doxorubicin Therapy-Differences in Cardiotoxicity. Cardiovasc Toxicol 2025; 25:248-268. [PMID: 39810066 DOI: 10.1007/s12012-024-09952-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Doxorubicin (DOX) is an important drug used in the treatment of many malignancies. Unfortunately DOX causes various side effects, with cardiotoxicity being the most characteristic. Risk factors for DOX induced cardiotoxicity (DIC) include cumulative dose of DOX, preexisting cardiovascular diseases, dyslipidemia, diabetes, smoking, along with the use of other cardiotoxic agents. Development of DIC is associated with many pathological phenomena - increased oxidative stress, as well as upregulation of ferroptosis, apoptosis, necrosis, and autophagy. In DIC expression of many microRNAs is also deregulated. In order to avoid cardiotoxicity and still use DOX effectively DOX derivatives such as epirubicin were synthesized. Nowadays a new liposomal form of DOX (L-DOX) appeared as an alternative to conventional treatment with greatly reduced cardiotoxicity. L-DOX can be divided into two groups of substances - pegylated (PLD) with increased solubility and stability, and non-pegylated (NLPD). Many metaanalyses, clinical along with preclinical studies have shown L-DOX treatment is associated with a smaller decrease of left ventricular ejection fraction (LVEF) and other heart functions, but efficacy of this treatment is comparable to the use of convenctional DOX.
Collapse
Affiliation(s)
- Barbara Buchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Katarzyna Kamińska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Michał Kowara
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Aleksandra Sobiborowicz-Sadowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|
2
|
Guo Q, Liao H, Hao S, Hou D, Liu R, Zhang Y, Tang X, Song R, Tan X, Luo Z, Huang H, Duan C. TSPO exacerbates sepsis-induced cardiac dysfunction by inhibiting p62-Mediated autophagic flux via the ROS-RIP1/RIP3-exosome axis. Free Radic Biol Med 2025; 226:56-69. [PMID: 39542185 DOI: 10.1016/j.freeradbiomed.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Septic cardiomyopathy (SCM) is a critical complication of sepsis, primarily attributed to mitochondrial dysfunction and impaired autophagic flux. This study explores the role of translocator protein (TSPO) in SCM pathogenesis and assesses its potential as a therapeutic target. We identified increased TSPO expression in plasma samples from sepsis patients, with further validation in septic rats and LPS-stimulated H9C2 cardiomyocytes. Elevated TSPO disrupted mitochondrial function, leading to increased reactive oxygen species (ROS) production and activation of the RIP1/RIP3 pathway, which hindered p62-positive autophagosome degradation and promoted inflammation. Moreover, exosome release containing TSPO-positive autophagosomes into plasma may exacerbate systemic inflammation. NADH, identified as a TSPO-binding molecule, restored autophagic flux, improved mitochondrial function, and enhanced cardiac performance and survival in septic rats. These findings suggest that targeting TSPO with NADH could alleviate mitochondrial dysfunction and inflammatory responses in SCM, providing a promising therapeutic strategy for sepsis-induced cardiac injury.
Collapse
Affiliation(s)
- Qiao Guo
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Haitang Liao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, PR China
| | - Shuai Hao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China
| | - Dongyao Hou
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, PR China
| | - Ruixue Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yunting Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xinhao Tang
- Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Rui Song
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xuxin Tan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Zhenchun Luo
- Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, PR China.
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
3
|
Li X, Chen X, Yang FY, Shu T, Jiang L, He B, Tang M, Li X, Fang D, Jose PA, Han Y, Yang Y, Zeng C. Effect of mitochondrial translocator protein TSPO on LPS-induced cardiac dysfunction. J Adv Res 2024:S2090-1232(24)00437-5. [PMID: 39389308 DOI: 10.1016/j.jare.2024.10.004] [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: 05/08/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Sepsis-induced cardiac dysfunction is one of the most serious complications of sepsis. The mitochondrial translocator protein (TSPO), a mitochondrial outer membrane protein, is widely used as a diagnostic marker of inflammation-related diseases and can also lead to the release of inflammatory components. However, whether TSPO has a therapeutic effect on sepsis-induced cardiac dysfunction is unclear. OBJECTIVES The aim of this study is to investigate the involvement of TSPO in the pathogenesis of sepsis-induced cardiac dysfunction and elucidate its underlying mechanism, as well as develop therapeutic strategies targeting TSPO for the prevention and treatment of sepsis-induced cardiac dysfunction. METHODS The sepsis-induced cardiac dysfunction model was established by intraperitoneal injection of lipopolysaccharide (LPS) in C57BL/6 mice (LPS-induced cardiac dysfunction, LICD). TSPO knockout mice were constructed,and the effects of TSPO was detected by survival rate, echocardiography, HE staining, mitochondrial electron microscopy, TUNEL staining. TSPO-binding proteins were identified by co-immunoprecipitation and mass spectrometry. The mechanisms underlying between TSPO and voltage-dependent anion channel (VDAC) was studied through western blot and immunofluorescence. Proteolysis-Targeting Chimeras (PROTAC) technology was used to construct TSPO-PROTAC molecules that can degrade TSPO. RESULTS Our present study found that LPS increased cardiac TSPO expression. Knockout of TSPO in C57BL/6 mice with LICD attenuated the cardiac pathology, mitochondrial dysfunction, and apoptosis of cardiomyocytes and significantly improved cardiac function and survival rate. Co-immunoprecipitation and mass spectrometry identified VDAC as a TSPO binding protein.Down-regulation of TSPO reduced PKA-mediated VDAC phosphorylation and VDAC oligomerization, ameliorated mitochondrial function, and reduced cardiomyocyte apoptosis. The study has clinical translational potential, because administration of TSPO-PROTAC to degrade TSPO improved cardiac function in mice with LICD. CONCLUSION This study elucidated the effect of TSPO in LICD, providing a new therapeutic strategy to down-regulate TSPO by administration of TSPO-PROTAC for the prevention and treatment of LICD.
Collapse
Affiliation(s)
- Xingyue Li
- School of Materials Science and Engineering,SouthwestJiaotong University, Chengdu Sichuan, PR China; Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, PR China
| | - Xiao Chen
- Department of Geriatrics, General Hospital of Western Theater Command, Chengdu, Sichuan, PR China
| | - Feng-Yuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, Sichuan, PR China
| | - Tingting Shu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China
| | - Lintao Jiang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China
| | - Bo He
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China
| | - Ming Tang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China
| | - Xingbing Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China
| | - Dandong Fang
- Department of Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, PR China
| | - Pedro A Jose
- The George Washington University School of Medicine & Health Sciences
| | - Yu Han
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Yongjian Yang
- School of Materials Science and Engineering,SouthwestJiaotong University, Chengdu Sichuan, PR China; Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, PR China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China; Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China; State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University,Chongqing, PR China.
| |
Collapse
|
4
|
Rao RM, El Dhaybi I, Cadet F, Etchebest C, Diharce J. The mutual and dynamic role of TSPO and ligands in their binding process: An example with PK-11195. Biochimie 2024; 224:29-40. [PMID: 38494108 DOI: 10.1016/j.biochi.2024.03.009] [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: 10/30/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/19/2024]
Abstract
Translocator protein (TSPO) is an 18 kDa transmembrane protein, localized primarily on the outer mitochondrial membrane. It has been found to be involved in various physiological processes and pathophysiological conditions. Though studies on its structure have been performed only recently, there is little information on the nature of dynamics and doubts about some structures referenced in the literature, especially the NMR structure of mouse TSPO. In the present work, we thoroughly study the dynamics of mouse TSPO protein by means of atomistic molecular dynamics simulations, in presence as well as in absence of the diagnostic ligand PKA. We considered two starting structures: the NMR structure and a homology model (HM) generated on the basis of X-ray structures from bacterial TSPO. We examine the conformational landscape in both the modes for both starting points, in presence and absence of the ligand, in order to measure its impact for both structures. The analysis highlights high flexibility of the protein globally, but NMR simulations show a surprisingly flexibility even in the presence of the ligand. Interestingly, this is not the case for HM calculations, to the point that the ligand seems not so stable as in the NMR system and an unbinding event process is partially sampled. All those results tend to show that the NMR structure of mTSPO seems not deficient but is just in another portion of the global conformation space of TSPO.
Collapse
Affiliation(s)
- Rajas M Rao
- Data Analytics, Bioinformatics and Structural Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India; Université Paris Cité and Université de la Réunion and Université des Antilles, INSERM, BIGR, DSIMB UMR_S1134, F-74014, Paris, France
| | - Ibaa El Dhaybi
- Université Paris Cité and Université de la Réunion and Université des Antilles, INSERM, BIGR, DSIMB UMR_S1134, F-74014, Paris, France
| | - Frédéric Cadet
- Université Paris Cité and Université de la Réunion and Université des Antilles, INSERM, BIGR, DSIMB UMR_S1134, F-74014, Paris, France; Laboratory of Excellence GR-Ex, Paris, France; Université Paris Cité and Université de la Réunion and Université des Antilles, INSERM, BIGR, DSIMB, F-97715, Saint Denis Messag, France; PEACCEL, Artificial Intelligence Department, Paris, 75013 France
| | - Catherine Etchebest
- Université Paris Cité and Université de la Réunion and Université des Antilles, INSERM, BIGR, DSIMB UMR_S1134, F-74014, Paris, France; Laboratory of Excellence GR-Ex, Paris, France
| | - Julien Diharce
- Université Paris Cité and Université de la Réunion and Université des Antilles, INSERM, BIGR, DSIMB UMR_S1134, F-74014, Paris, France; Laboratory of Excellence GR-Ex, Paris, France.
| |
Collapse
|
5
|
Bréhat J, Issop L, Morin D. History of Tspo deletion and induction in vivo: Phenotypic outcomes under physiological and pathological situations. Biochimie 2024; 224:80-90. [PMID: 38432291 DOI: 10.1016/j.biochi.2024.03.001] [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: 12/21/2023] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/05/2024]
Abstract
The mitochondrial translocator protein (TSPO) is an outer mitochondrial protein membrane with high affinity for cholesterol. It is expressed in most tissues but is more particularly enriched in steroidogenic tissues. TSPO is involved in various biological mechanisms and TSPO regulation has been related to several diseases. However, despite a considerable number of published studies interested in TSPO over the past forty years, the precise function of the protein remains obscure. Most of the functions attributed to TSPO have been identified using pharmacological ligands of this protein, leading to much debate about the accuracy of these findings. In addition, research on the physiological role of TSPO has been hampered by the lack of in vivo deletion models. Studies to perform genetic deletion of Tspo in animal models have long been unsuccessful, which led to the conclusions that the deletion was deleterious and the gene essential to life. During the last decades, thanks to the significant technical advances allowing genome modification, several models of animal genetically modified for TSPO have been developed. These models have modified our view regarding TSPO and profoundly improved our fundamental knowledge on this protein. However, to date, they did not allow to elucidate the precise molecular function of TSPO and numerous questions persist concerning the physiological role of TSPO and its future as a therapeutic target. This article chronologically reviews the development of deletion and induction models of TSPO.
Collapse
Affiliation(s)
- Juliette Bréhat
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Leeyah Issop
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Didier Morin
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France.
| |
Collapse
|
6
|
Hu B, Yu H, Du S, Li Q. Protoporphyrin IX metabolism mediated via translocator protein (CgTspO) involved in orange shell coloration of pacific oyster (Crassostrea gigas). Int J Biol Macromol 2024; 276:134020. [PMID: 39038584 DOI: 10.1016/j.ijbiomac.2024.134020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/06/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Mollusc shell color polymorphism is influenced by various factors. Pigments secreted in vivo by animals play a critical role in shell coloration. Among the different shell-color hues, orange pigmentation has been partially attributed to porphyrins. However, the detailed causal relationship between porphyrins and orange-shell phenotype in molluscs remains largely unexplored. The various strains of Pacific oyster (Crassostrea gigas) with different shell color provide useful models to study the molecular regulation of mollusc coloration. Accordingly, oysters with orange and gold-shells, exhibiting distinct porphyrin distributions, were selected for analysis of total metabolites and gene expression profile through mantle metabolomic and transcriptomic studies. Translocator protein (TspO) and protoporphyrin IX (PPIX) were identified as potential factors influencing oyster shell-color. The concentration of PPIX was measured using HPLC, while expression profiling of CgTspO was analyzed by qPCR, in situ hybridization, Western blotting, and immunofluorescence techniques. Moreover, the roles of CgTspO in regulating PPIX metabolism and affecting the orange-shell-coloration were investigated in vitro and in vivo. These studies indicate that PPIX and its associated metabolic protein, CgTspO may serve as new regulators of orange-shell-coloration in C. gigas. Data of this study offer new insights into oyster shell coloration and enhancing understandings of mollusc shell color polymorphism.
Collapse
Affiliation(s)
- Biyang Hu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Hong Yu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Shaojun Du
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
7
|
Kong JN, Dipon Ghosh D, Savvidis A, Sando SR, Droste R, Robert Horvitz H. Transcriptional landscape of a hypoxia response identifies cell-specific pathways for adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601765. [PMID: 39005398 PMCID: PMC11245032 DOI: 10.1101/2024.07.02.601765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
How the HIF-1 (Hypoxia-Inducible) transcription factor drives and coordinates distinct responses to low oxygen across diverse cell types is poorly understood. We present a multi-tissue single-cell gene-expression atlas of the hypoxia response of the nematode Caenorhabditis elegans . This atlas highlights how cell-type-specific HIF-1 responses overlap and diverge among and within neuronal, intestinal, and muscle tissues. Using the atlas to guide functional analyses of candidate muscle-specific HIF-1 effectors, we discovered that HIF-1 activation drives downregulation of the tspo-1 ( TSPO, Translocator Protein) gene in vulval muscle cells to modulate a hypoxia-driven change in locomotion caused by contraction of body-wall muscle cells. We further showed that in human cardiomyocytes HIF-1 activation decreases levels of TSPO and thereby alters intracellular cholesterol transport and the mitochondrial network. We suggest that TSPO-1 is an evolutionarily conserved mediator of HIF-1-dependent modulation of muscle and conclude that our gene-expression atlas can help reveal how HIF-1 drives cell-specific adaptations to hypoxia.
Collapse
|
8
|
Bréhat J, Leick S, Musman J, Su JB, Eychenne N, Giton F, Rivard M, Barel LA, Tropeano C, Vitarelli F, Caccia C, Leoni V, Ghaleh B, Pons S, Morin D. Identification of a mechanism promoting mitochondrial sterol accumulation during myocardial ischemia-reperfusion: role of TSPO and STAR. Basic Res Cardiol 2024; 119:481-503. [PMID: 38517482 DOI: 10.1007/s00395-024-01043-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/24/2024]
Abstract
Hypercholesterolemia is a major risk factor for coronary artery diseases and cardiac ischemic events. Cholesterol per se could also have negative effects on the myocardium, independently from hypercholesterolemia. Previously, we reported that myocardial ischemia-reperfusion induces a deleterious build-up of mitochondrial cholesterol and oxysterols, which is potentiated by hypercholesterolemia and prevented by translocator protein (TSPO) ligands. Here, we studied the mechanism by which sterols accumulate in cardiac mitochondria and promote mitochondrial dysfunction. We performed myocardial ischemia-reperfusion in rats to evaluate mitochondrial function, TSPO, and steroidogenic acute regulatory protein (STAR) levels and the related mitochondrial concentrations of sterols. Rats were treated with the cholesterol synthesis inhibitor pravastatin or the TSPO ligand 4'-chlorodiazepam. We used Tspo deleted rats, which were phenotypically characterized. Inhibition of cholesterol synthesis reduced mitochondrial sterol accumulation and protected mitochondria during myocardial ischemia-reperfusion. We found that cardiac mitochondrial sterol accumulation is the consequence of enhanced influx of cholesterol and not of the inhibition of its mitochondrial metabolism during ischemia-reperfusion. Mitochondrial cholesterol accumulation at reperfusion was related to an increase in mitochondrial STAR but not to changes in TSPO levels. 4'-Chlorodiazepam inhibited this mechanism and prevented mitochondrial sterol accumulation and mitochondrial ischemia-reperfusion injury, underlying the close cooperation between STAR and TSPO. Conversely, Tspo deletion, which did not alter cardiac phenotype, abolished the effects of 4'-chlorodiazepam. This study reveals a novel mitochondrial interaction between TSPO and STAR to promote cholesterol and deleterious sterol mitochondrial accumulation during myocardial ischemia-reperfusion. This interaction regulates mitochondrial homeostasis and plays a key role during mitochondrial injury.
Collapse
Affiliation(s)
- Juliette Bréhat
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France
| | - Shirin Leick
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France
| | - Julien Musman
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France
| | - Jin Bo Su
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France
| | | | - Frank Giton
- Pôle Biologie-Pathologie, IMRB U955, Hôpital Henri Mondor, Créteil, France
| | | | | | - Chiara Tropeano
- Laboratory of Clinical Chemistry, ASST-Brianza Department of Medicine and Surgery, Hospital Pio XI Desio, University of Milano Bicocca, Monza, Italy
| | - Frederica Vitarelli
- Laboratory of Clinical Chemistry, ASST-Brianza Department of Medicine and Surgery, Hospital Pio XI Desio, University of Milano Bicocca, Monza, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics, Istituto Neurologico Carlo Besta, Fondazione IRCCS, Milan, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, ASST-Brianza Department of Medicine and Surgery, Hospital Pio XI Desio, University of Milano Bicocca, Monza, Italy
| | - Bijan Ghaleh
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France
| | - Sandrine Pons
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France
| | - Didier Morin
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Faculté de Santé, 8 rue du général Sarrail, 94000, Créteil, France.
| |
Collapse
|
9
|
Casalechi M, Dela Cruz C, Assis WA, Vieira-Lopes M, Lopes FEF, Francisco AMC, Reis FM. Translocator protein expression and localization in human endometrium and endometriosis: A potential target for a noninvasive diagnosis? Cell Biol Int 2024. [PMID: 38511230 DOI: 10.1002/cbin.12157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
The limitations of current imaging methods to detect small or superficial endometriotic lesions prompt the search for new molecular targets. TSPO is an 18 KDa protein located in the outer mitochondrial membrane, which can be traced by positron emission tomography (PET) using specific ligands. TSPO is located mostly in neurons and inflammatory sites outside the brain. We hypothesized that it might also be expressed in the human endometrium and endometrial-like tissue, being a target for molecular imaging of endometriosis. This prospective cross-sectional study included 28 women with endometriosis and 11 endometriosis-free controls. Endometriotic lesions (n = 49) and normal peritoneum (n = 13) from endometriosis patients were obtained during laparoscopy, while samples of eutopic endometrium from patients with endometriosis (n = 28) and from control women (n = 11) were collected in the operating room using a flexible device. TSPO mRNA expression was evaluated by quantitative reverse-transcription real-time PCR while protein expression was evaluated by immunohistochemistry with a monoclonal antibody antihuman TSPO. TSPO mRNA expression was detected in an invariable fashion in all tissue types evaluated; however, TSPO protein was found to be more abundant in the glandular epithelium than in the stroma, both in the endometrium and in the endometriotic lesions. Interestingly, hormone therapies did not alter the expression of TSPO, and its presence was mostly negative in tissues adjacent to endometriotic implants. As a proof of concept, the protein expression pattern of TSPO in endometriotic tissue and along the adjacent areas suggests that TSPO-based molecular imaging might be used for noninvasive endometriosis detection.
Collapse
Affiliation(s)
- Maíra Casalechi
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cynthia Dela Cruz
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Wiviane A Assis
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Millene Vieira-Lopes
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Eduardo F Lopes
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio M C Francisco
- Maternal and Child Department, Universidade do Vale do Sapucaí, Pouso Alegre, Brazil
| | - Fernando M Reis
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
10
|
Yu M, Zhao S. Functional role of translocator protein and its ligands in ocular diseases (Review). Mol Med Rep 2024; 29:33. [PMID: 38186312 PMCID: PMC10804439 DOI: 10.3892/mmr.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
The 18 kDa translocator protein (TSPO) is an essential outer mitochondrial membrane protein that is responsible for mitochondrial transport, maintenance of mitochondrial homeostasis and normal physiological cell function. The role of TSPO in the pathogenesis of ocular diseases is a growing area of interest. More notably, TSPO exerts positive effects in regulating various pathophysiological processes, such as the inflammatory response, oxidative stress, steroid synthesis and modulation of microglial function, in combination with a variety of specific ligands such as 1‑(2‑chlorophenyl‑N‑methylpropyl)‑3‑isoquinolinecarboxamide, 4'‑chlorodiazepam and XBD173. In the present review, the expression of TSPO in ocular tissues and the functional role of TSPO and its ligands in diverse ocular diseases was discussed.
Collapse
Affiliation(s)
- Mingyi Yu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 30384, P.R. China
| | - Shaozhen Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 30384, P.R. China
| |
Collapse
|
11
|
Baglini E, Poggetti V, Cavallini C, Petroni D, Forini F, Nicolini G, Barresi E, Salerno S, Costa B, Iozzo P, Neglia D, Menichetti L, Taliani S, Da Settimo F. Targeting the Translocator Protein (18 kDa) in Cardiac Diseases: State of the Art and Future Opportunities. J Med Chem 2024; 67:17-37. [PMID: 38113353 PMCID: PMC10911791 DOI: 10.1021/acs.jmedchem.3c01716] [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: 09/15/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023]
Abstract
Mitochondria dysfunctions are typical hallmarks of cardiac disorders (CDs). The multiple tasks of this energy-producing organelle are well documented, but its pathophysiologic involvement in several manifestations of heart diseases, such as altered electromechanical coupling, excitability, and arrhythmias, is still under investigation. The human 18 kDa translocator protein (TSPO) is a protein located on the outer mitochondrial membrane whose expression is altered in different pathological conditions, including CDs, making it an attractive therapeutic and diagnostic target. Currently, only a few TSPO ligands are employed in CDs and cardiac imaging. In this Perspective, we report an overview of the emerging role of TSPO at the heart level, focusing on the recent literature concerning the development of TSPO ligands used for fighting and imaging heart-related disease conditions. Accordingly, targeting TSPO might represent a successful strategy to achieve novel therapeutic and diagnostic strategies to unravel the fundamental mechanisms and to provide solutions to still unanswered questions in CDs.
Collapse
Affiliation(s)
- Emma Baglini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Valeria Poggetti
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Chiara Cavallini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Debora Petroni
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Francesca Forini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Giuseppina Nicolini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Elisabetta Barresi
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Silvia Salerno
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Barbara Costa
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Patricia Iozzo
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Danilo Neglia
- Fondazione
CNR/Regione Toscana Gabriele Monasterio, Cardiovascular and Imaging
Departments, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Luca Menichetti
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Sabrina Taliani
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Federico Da Settimo
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| |
Collapse
|
12
|
Translocator Protein 18 kDa (TSPO) as a Novel Therapeutic Target for Chronic Pain. Neural Plast 2022; 2022:8057854. [PMID: 36071748 PMCID: PMC9444456 DOI: 10.1155/2022/8057854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/19/2022] [Accepted: 08/13/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic pain is an enormous modern public health problem, with significant numbers of people debilitated by chronic pain from a variety of etiologies. Translocator protein 18 kDa (TSPO) was discovered in 1977 as a peripheral benzodiazepine receptor. It is a five transmembrane domain protein, mainly localized in the outer mitochondrial membrane. Recent and increasing studies have found changes in TSPO and its ligands in various chronic pain models. Reversing their expressions has been shown to alleviate chronic pain in these models, illustrating the effects of TSPO and its ligands. Herein, we review recent evidence and the mechanisms of TSPO in the development of chronic pain associated with peripheral nerve injury, spinal cord injury, cancer, and inflammatory responses. The cumulative evidence indicates that TSPO-based therapy may become an alternative strategy for treating chronic pain.
Collapse
|
13
|
Qu Y, Cao J, Wang D, Wang S, Li Y, Zhu Y. 14,15-Epoxyeicosatrienoic Acid Protect Against Glucose Deprivation and Reperfusion-Induced Cerebral Microvascular Endothelial Cells Injury by Modulating Mitochondrial Autophagy via SIRT1/FOXO3a Signaling Pathway and TSPO Protein. Front Cell Neurosci 2022; 16:888836. [PMID: 35558879 PMCID: PMC9086968 DOI: 10.3389/fncel.2022.888836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Neurovascular system plays a vital role in controlling the blood flow into brain parenchymal tissues. Additionally, it also facilitates the metabolism in neuronal biological activities. Cerebral microvascular endothelial cells (MECs) are involved in mediating progression of the diseases related to cerebral vessels, including stroke. Arachidonic acid can be transformed into epoxyeicosatrienoic acids (EETs) under the catalysis by cytochrome P450 epoxygenase. We have reported that EETs could protect neuronal function. In our research, the further role of 14,15-EET in the protective effects of cerebral MECs and the potential mechanisms involved in oxygen glucose deprivation and reperfusion (OGD/R) were elucidated. In our study, we intervened the SIRT1/FOXO3a pathway and established a TSPO knock down model by using RNA interference technique to explore the cytoprotective role of 14,15-EET in OGD/R injury. Cerebral MECs viability was remarkably reduced after OGD/R treatment, however, 14,15-EET could reverse this effect. To further confirm whether 14,15-EET was mediated by SIRT1/FOXO3a signaling pathway and translocator protein (TSPO) protein, we also detected autophagy-related proteins, mitochondrial membrane potential, apoptosis indicators, oxygen free radicals, etc. It was found that 14,15-EET could regulate the mitophagy induced by OGD/R. SIRT1/FOXO3a signaling pathway and TSPO regulation were related to the protective role of 14,15-EET in cerebral MECs. Moreover, we also explored the potential relationship between SIRT1/FOXO3a signaling pathway and TSPO protein. Our study revealed the protective role and the potential mechanisms of 14,15-EET in cerebral MECs under OGD/R condition.
Collapse
Affiliation(s)
- Youyang Qu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinlu Cao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shu Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yujie Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yulan Zhu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Successful treatment of cancer can be hampered by the attendant risk of cardiotoxicity, manifesting as cardiomyopathy, left ventricle systolic dysfunction and, in some cases, heart failure. This risk can be mitigated if the injury to the heart is detected before the onset to irreversible cardiac impairment. The gold standard for cardiac imaging in cardio-oncology is echocardiography. Despite improvements in the application of this modality, it is not typically sensitive to sub-clinical or early-stage dysfunction. We identify in this review some emerging tracers for detecting incipient cardiotoxicity by positron emission tomography (PET). RECENT FINDINGS Vectors labeled with positron-emitting radionuclides (e.g., carbon-11, fluorine-18, gallium-68) are now available to study cardiac function, metabolism, and tissue repair in preclinical models. Many of these probes are highly sensitive to early damage, thereby potentially addressing the limitations of current imaging approaches, and show promise in preliminary clinical evaluations. The overlapping pathophysiology between cardiotoxicity and heart failure significantly expands the number of imaging tools available to cardio-oncology. This is highlighted by the emergence of radiolabeled probes targeting fibroblast activation protein (FAP) for sensitive detection of dysregulated healing process that underpins adverse cardiac remodeling. The growth of PET scanner technology also creates an opportunity for a renaissance in metabolic imaging in cardio-oncology research.
Collapse
Affiliation(s)
- James M. Kelly
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
| | - John W. Babich
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021 USA
| |
Collapse
|
15
|
Yang C, Zhang Y, Yang B. MIAT, a potent CVD-promoting lncRNA. Cell Mol Life Sci 2021; 79:43. [PMID: 34921634 PMCID: PMC11072732 DOI: 10.1007/s00018-021-04046-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
The initial identification of long non-coding RNA myocardial infarction associated transcript (MIAT) as a genetic risk factor of myocardial infarction has made this lncRNA (designated as lncR-MIAT here) a focus of intensive studies worldwide. Emerging evidence supports that lncR-MIAT is susceptible in its expression to multiple deleterious factors like angiotensin II, isoproterenol, hypoxia, and infection and is anomaly overexpressed in serum, plasma, blood cells and myocardial tissues under a variety of cardiovascular conditions including myocardial infarction, cardiac hypertrophy, diabetic cardiomyopathy, dilated cardiomyopathy, sepsis cardiomyopathy, atrial fibrillation and microvascular dysfunction. Experimental results consistently demonstrated that upregulation of lncR-MIAT plays active roles in the pathological processes of the cardiovascular system and knockdown of this lncRNA effectively ameliorates the adverse conditions. The available data revealed that lncR-MIAT acts through multiple mechanisms such as competitive endogenous RNA, natural antisense RNA and RNA/protein interactions. Moreover, the functional domains of lncR-MIAT accounting for certain specific cellular functions of the full-length transcript have been identified and characterized. These insights will not only tremendously advance our understanding of lncRNA biology and pathophysiology, but also offer good opportunities for more innovative and precise design of agents that have the potential to be developed into new drugs for better therapy of cardiovascular diseases (CVDs) in the future. Herein, we provide an overview of lncR-MIAT, focusing on its roles in cardiovascular diseases, underline the unique cellular/molecular mechanisms for its actions, and speculate the perspectives about the translational studies on the potential diagnostic and therapeutic applications of lncR-MIAT.
Collapse
Affiliation(s)
- Chao Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang, People's Republic of China
| | - Yong Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150086, People's Republic of China
| | - Baofeng Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China.
| |
Collapse
|
16
|
Carbenoxolon Is Capable to Regulate the Mitochondrial Permeability Transition Pore Opening in Chronic Alcohol Intoxication. Int J Mol Sci 2021; 22:ijms221910249. [PMID: 34638588 PMCID: PMC8549702 DOI: 10.3390/ijms221910249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
Background: carbenoxolone, which is a derivative of glyceretic acid, is actively used in pharmacology for the treatment of diseases of various etiologies. In addition, we have shown carbenoxolone as an effective inducer of mitochondrial permeability transition pore in rat brain and liver mitochondria. Methods: in the course of this work, comparative studies were carried out on the effect of carbenoxolone on the parameters of mPTP functioning in mitochondria isolated from the liver of control and alcoholic rats. Results: within the framework of this work, it was found that carbenoxolone significantly increased its effect in the liver mitochondria of rats with chronic intoxication. In particular, this was expressed in a reduction in the lag phase, a decrease in the threshold calcium concentration required to open a pore, an acceleration of high-amplitude cyclosporin-sensitive swelling of mitochondria, as well as an increase in the effect of carbenoxolone on the level of mitochondrial membrane-bound proteins. Thus, as a result of the studies carried out, it was shown that carbenoxolone is involved in the development/modulation of alcohol tolerance and dependence in rats.
Collapse
|
17
|
Xu C, Tao X, Ma X, Zhao R, Cao Z. Cognitive Dysfunction after Heart Disease: A Manifestation of the Heart-Brain Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4899688. [PMID: 34457113 PMCID: PMC8387198 DOI: 10.1155/2021/4899688] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/31/2021] [Indexed: 12/26/2022]
Abstract
The functions of the brain and heart, which are the two main supporting organs of human life, are closely linked. Numerous studies have expounded the mechanisms of the brain-heart axis and its related clinical applications. However, the effect of heart disease on brain function, defined as the heart-brain axis, is less studied even though cognitive dysfunction after heart disease is one of its most frequently reported manifestations. Hypoperfusion caused by heart failure appears to be an important risk factor for cognitive decline. Blood perfusion, the immune response, and oxidative stress are the possible main mechanisms of cognitive dysfunction, indicating that the blood-brain barrier, glial cells, and amyloid-β may play active roles in these mechanisms. Clinicians should pay more attention to the cognitive function of patients with heart disease, especially those with heart failure. In addition, further research elucidating the associated mechanisms would help discover new therapeutic targets to intervene in the process of cognitive dysfunction after heart disease. This review discusses cognitive dysfunction in relation to heart disease and its potential mechanisms.
Collapse
Affiliation(s)
- Chengyang Xu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Department of Forensic Pathophysiology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Xueshu Tao
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang, China. No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Xiaonan Ma
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Department of Forensic Pathophysiology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Department of Forensic Pathophysiology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Department of Forensic Pathophysiology, School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang 110122, China
| |
Collapse
|
18
|
Balogh V, MacAskill MG, Hadoke PWF, Gray GA, Tavares AAS. Positron Emission Tomography Techniques to Measure Active Inflammation, Fibrosis and Angiogenesis: Potential for Non-invasive Imaging of Hypertensive Heart Failure. Front Cardiovasc Med 2021; 8:719031. [PMID: 34485416 PMCID: PMC8416043 DOI: 10.3389/fcvm.2021.719031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure, which is responsible for a high number of deaths worldwide, can develop due to chronic hypertension. Heart failure can involve and progress through several different pathways, including: fibrosis, inflammation, and angiogenesis. Early and specific detection of changes in the myocardium during the transition to heart failure can be made via the use of molecular imaging techniques, including positron emission tomography (PET). Traditional cardiovascular PET techniques, such as myocardial perfusion imaging and sympathetic innervation imaging, have been established at the clinical level but are often lacking in pathway and target specificity that is important for assessment of heart failure. Therefore, there is a need to identify new PET imaging markers of inflammation, fibrosis and angiogenesis that could aid diagnosis, staging and treatment of hypertensive heart failure. This review will provide an overview of key mechanisms underlying hypertensive heart failure and will present the latest developments in PET probes for detection of cardiovascular inflammation, fibrosis and angiogenesis. Currently, selective PET probes for detection of angiogenesis remain elusive but promising PET probes for specific targeting of inflammation and fibrosis are rapidly progressing into clinical use.
Collapse
Affiliation(s)
- Viktoria Balogh
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G. MacAskill
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A. Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
19
|
Mitochondrion as a Target of Astaxanthin Therapy in Heart Failure. Int J Mol Sci 2021; 22:ijms22157964. [PMID: 34360729 PMCID: PMC8347622 DOI: 10.3390/ijms22157964] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/22/2022] Open
Abstract
Mitochondria are considered to be important organelles in the cell and play a key role in the physiological function of the heart, as well as in the pathogenesis and development of various heart diseases. Under certain pathological conditions, such as cardiovascular diseases, stroke, traumatic brain injury, neurodegenerative diseases, muscular dystrophy, etc., mitochondrial permeability transition pore (mPTP) is formed and opened, which can lead to dysfunction of mitochondria and subsequently to cell death. This review summarizes the results of studies carried out by our group of the effect of astaxanthin (AST) on the functional state of rat heart mitochondria upon direct addition of AST to isolated mitochondria and upon chronic administration of AST under conditions of mPTP opening. It was shown that AST exerted a protective effect under all conditions. In addition, AST treatment was found to prevent isoproterenol-induced oxidative damage to mitochondria and increase mitochondrial efficiency. AST, a ketocarotenoid, may be a potential mitochondrial target in therapy for pathological conditions associated with oxidative damage and mitochondrial dysfunction, and may be a potential mitochondrial target in therapy for pathological conditions.
Collapse
|
20
|
Seidlmayer LK, Hanson BJ, Thai PN, Schaefer S, Bers DM, Dedkova EN. PK11195 Protects From Cell Death Only When Applied During Reperfusion: Succinate-Mediated Mechanism of Action. Front Physiol 2021; 12:628508. [PMID: 34149440 PMCID: PMC8212865 DOI: 10.3389/fphys.2021.628508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Reperfusion after myocardial ischemia causes cellular injury, in part due to changes in mitochondrial Ca2+ handling, oxidative stress, and myocyte energetics. We have previously shown that the 18-kDa translocator protein of the outer mitochondrial membrane (TSPO) can modulate Ca2+ handling. Here, we aim to evaluate the role of the TSPO in ischemia/reperfusion (I/R) injury. Methods: Rabbit ventricular myocytes underwent simulated acute ischemia (20 min) and reperfusion (at 15 min, 1 h, and 3 h) in the absence and presence of 50 μM PK11195, a TSPO inhibitor. Cell death was measured by lactate dehydrogenase (LDH) assay, while changes in mitochondrial Ca2+, membrane potential (ΔΨm), and reactive oxygen species (ROS) generation were monitored using confocal microscopy in combination with fluorescent indicators. Substrate utilization was measured with Biolog mitochondrial plates. Results: Cell death was increased by ~200% following I/R compared to control untreated ventricular myocytes. Incubation with 50 μM PK11195 during both ischemia and reperfusion did not reduce cell death but increased mitochondrial Ca2+ uptake and ROS generation. However, application of 50 μM PK11195 only at the onset and during reperfusion effectively protected against cell death. The large-scale oscillations in ΔΨm observed after ~1 h of reperfusion were significantly delayed by 1 μM cyclosporin A and almost completely prevented by 50 μM PK11195 applied during 3 h of reperfusion. After an initial increase, mitochondrial Ca2+, measured with Myticam, rapidly declined during 3 h of reperfusion after the initial transient increase. This decline was prevented by application of PK11195 at the onset and during reperfusion. PK11195 prevented a significant increase in succinate utilization following I/R and succinate-induced forward-mode ROS generation. Treatment with PK11195 was also associated with a significant increase in glutamate and a decrease in leucine utilization. Conclusion: PK11195 administered specifically at the moment of reperfusion limited ROS-induced ROS release and cell death, likely in part, by a shift from succinate to glutamate utilization. These data demonstrate a unique mechanism to limit cardiac injury after I/R.
Collapse
Affiliation(s)
- Lea K Seidlmayer
- Department of Cardiology, University Hospital Olbenburg, Olbenburg, Germany
| | - Benjamin J Hanson
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Saul Schaefer
- Department of Internal Medicine, Division of Cardiovascular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Donald M Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Elena N Dedkova
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
21
|
Ilic A, Todorovic D, Mutavdzin S, Boricic N, Bozic Nedeljkovic B, Stankovic S, Simic T, Stevanovic P, Celic V, Djuric D. Translocator Protein Modulation by 4'-Chlorodiazepam and NO Synthase Inhibition Affect Cardiac Oxidative Stress, Cardiometabolic and Inflammatory Markers in Isoprenaline-Induced Rat Myocardial Infarction. Int J Mol Sci 2021; 22:2867. [PMID: 33799869 PMCID: PMC8000569 DOI: 10.3390/ijms22062867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022] Open
Abstract
The possible cardioprotective effects of translocator protein (TSPO) modulation with its ligand 4'-Chlorodiazepam (4'-ClDzp) in isoprenaline (ISO)-induced rat myocardial infarction (MI) were evaluated, alone or in the presence of L-NAME. Wistar albino male rats (b.w. 200-250 g, age 6-8 weeks) were divided into 4 groups (10 per group, total number N = 40), and certain substances were applied: 1. ISO 85 mg/kg b.w. (twice), 2. ISO 85 mg/kg b.w. (twice) + L-NAME 50 mg/kg b.w., 3. ISO 85 mg/kg b.w. (twice) + 4'-ClDzp 0.5 mg/kg b.w., 4. ISO 85 mg/kg b.w. (twice) + 4'-ClDzp 0.5 mg/kg b.w. + L-NAME 50 mg/kg b.w. Blood and cardiac tissue were sampled for myocardial injury and other biochemical markers, cardiac oxidative stress, and for histopathological evaluation. The reduction of serum levels of high-sensitive cardiac troponin T hs cTnT and tumor necrosis factor alpha (TNF-α), then significantly decreased levels of serum homocysteine Hcy, urea, and creatinine, and decreased levels of myocardial injury enzymes activities superoxide dismutase (SOD) and glutathione peroxidase (GPx) as well as lower grades of cardiac ischemic changes were demonstrated in ISO-induced MI treated with 4'-ClDzp. It has been detected that co-treatment with 4'-ClDzp + L-NAME changed the number of registered parameters in comparison to 4'-ClDzp group, indicating that NO (nitric oxide) should be important in the effects of 4'-ClDzp.
Collapse
Affiliation(s)
- Ana Ilic
- Department of Cardiology, University Clinical Hospital Center “Dr. Dragisa Misovic—Dedinje”, 11000 Belgrade, Serbia; (A.I.); (V.C.)
| | - Dusan Todorovic
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.T.); (S.M.)
| | - Slavica Mutavdzin
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.T.); (S.M.)
| | - Novica Boricic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Biljana Bozic Nedeljkovic
- Institute for Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sanja Stankovic
- Center for Medical Biochemistry, Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Predrag Stevanovic
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, University Clinical Hospital Center “Dr. Dragisa Misovic—Dedinje”, 11000 Belgrade, Serbia;
| | - Vera Celic
- Department of Cardiology, University Clinical Hospital Center “Dr. Dragisa Misovic—Dedinje”, 11000 Belgrade, Serbia; (A.I.); (V.C.)
| | - Dragan Djuric
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.T.); (S.M.)
| |
Collapse
|
22
|
Cui Y, Liang Y, Ip MSM, Mak JCW. Cigarette smoke induces apoptosis via 18 kDa translocator protein in human bronchial epithelial cells. Life Sci 2021; 265:118862. [PMID: 33301812 DOI: 10.1016/j.lfs.2020.118862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/20/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
AIMS The 18 kDa translocator protein (TSPO) - also known as peripheral benzodiazepine receptor, is found to be expressed in lung epithelium and pneumocytes, which is closely associated with the mitochondrial permeability transition pore (mPTP) and apoptosis. Cigarette smoking, a key risk factor for the development of chronic obstructive pulmonary disease (COPD), is known to induce apoptosis. We aimed to investigate TSPO subcellular localization and to examine whether cigarette smoke medium (CSM) induce apoptosis via TSPO in airway epithelial cells. MAIN METHODS TSPO subcellular localization and expression were evaluated using immunofluorescent staining and Western blot analysis respectively. TSPO ligands either PK 11195 (a specific antagonist) or AC-5216 (a specific agonist) were pre-incubated in human bronchial epithelial cells before treating with 2% CSM for measurements of apoptotic cells, mitochondrial membrane potential (ΔΨm), cytoplasmic/mitochondrial reactive oxygen species (ROS) and inflammatory marker interleukin (IL)-8 respectively. KEY FINDINGS TSPO was localized around the nucleus and overlapped with mitochondria in BEAS-2B cells. CSM caused an increase in apoptotic cells along with elevation of TSPO protein expression. Pretreatment of PK 11195 suppressed while AC-5216 potentiated CSM-induced apoptosis, collapse of ΔΨm, elevation of cytoplasmic/mitochondrial ROS levels and IL-8 release. In support, knockdown of TSPO caused a significant suppression of CSM-induced IL-8 release in BEAS-2B cells. SIGNIFICANCE The findings suggest that TSPO may play a crucial role in the regulation of cigarette smoke-induced mitochondrial dysfunction via mPTP. Therefore, the development of specific TSPO antagonists like PK11195 may be beneficial to combat smoking-related diseases, such as COPD.
Collapse
Affiliation(s)
- Yuting Cui
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingmin Liang
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Mary S M Ip
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Judith C W Mak
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology & Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
23
|
Cui Y, Pan M, Ma J, Song X, Cao W, Zhang P. Recent progress in the use of mitochondrial membrane permeability transition pore in mitochondrial dysfunction-related disease therapies. Mol Cell Biochem 2021; 476:493-506. [PMID: 33000352 DOI: 10.1007/s11010-020-03926-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria have various cellular functions, including ATP synthesis, calcium homeostasis, cell senescence, and death. Mitochondrial dysfunction has been identified in a variety of disorders correlated with human health. Among the many underlying mechanisms of mitochondrial dysfunction, the opening up of the mitochondrial permeability transition pore (mPTP) is one that has drawn increasing interest in recent years. It plays an important role in apoptosis and necrosis; however, the molecular structure and function of the mPTP have still not been fully elucidated. In recent years, the abnormal opening up of the mPTP has been implicated in the development and pathogenesis of diverse diseases including ischemia/reperfusion injury (IRI), neurodegenerative disorders, tumors, and chronic obstructive pulmonary disease (COPD). This review provides a systematic introduction to the possible molecular makeup of the mPTP and summarizes the mitochondrial dysfunction-correlated diseases and highlights possible underlying mechanisms. Since the mPTP is an important target in mitochondrial dysfunction, this review also summarizes potential treatments, which may be used to inhibit pore opening up via the molecules composing mPTP complexes, thus suppressing the progression of mitochondrial dysfunction-related diseases.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Science, Shandong University of Technology, Zibo, Shandong Province, China
| | - Mingyue Pan
- Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong Province, China
| | - Jing Ma
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong Province, China
| | - Xinhua Song
- School of Life Science, Shandong University of Technology, Zibo, Shandong Province, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong Province, China.
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong Province, China.
| |
Collapse
|
24
|
Gyöngyösi M, Lukovic D, Zlabinger K, Spannbauer A, Gugerell A, Pavo N, Traxler D, Pils D, Maurer G, Jakab A, Riesenhuber M, Pircher A, Winkler J, Bergler-Klein J. Liposomal doxorubicin attenuates cardiotoxicity via induction of interferon-related DNA damage resistance. Cardiovasc Res 2020; 116:970-982. [PMID: 31346605 DOI: 10.1093/cvr/cvz192] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/17/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022] Open
Abstract
AIMS The clinical application of doxorubicin (DOX) is severely compromised by its cardiotoxic effects, which limit the therapeutic index and the cumulative dose. Liposomal encapsulation of DOX (Myocet®) provides a certain protective effect against cardiotoxicity by reducing myocardial drug accumulation. We aimed to evaluate transcriptomic responses to anthracyclines with different cardiotoxicity profiles in a translational large animal model for identifying potential alleviation strategies. METHODS AND RESULTS We treated domestic pigs with either DOX, epirubicin (EPI), or liposomal DOX and compared the cardiac, laboratory, and haemodynamic effects with saline-treated animals. Cardiotoxicity was encountered in all groups, reflected by an increase of plasma markers N-terminal pro-brain-natriuretic peptide and Troponin I and an impact on body weight. High morbidity of EPI-treated animals impeded further evaluation. Cardiac magnetic resonance imaging with gadolinium late enhancement and transthoracic echocardiography showed stronger reduction of the left and right ventricular systolic function and stronger myocardial fibrosis in DOX-treated animals than in those treated with the liposomal formulation. Gene expression profiles of the left and right ventricles were analysed by RNA-sequencing and validated by qPCR. Interferon-stimulated genes (ISGs), linked to DNA damage repair and cell survival, were downregulated by DOX, but upregulated by liposomal DOX in both the left and right ventricle. The expression of cardioprotective translocator protein (TSPO) was inhibited by DOX, but not its liposomal formulation. Cardiac fibrosis with activation of collagen was found in all treatment groups. CONCLUSIONS All anthracycline-derivatives resulted in transcriptional activation of collagen synthesis and processing. Liposomal packaging of DOX-induced ISGs in association with lower cardiotoxicity, which is of high clinical importance in anticancer treatment. Our study identified potential mechanisms for rational development of strategies to mitigate anthracycline-induced cardiomyopathy.
Collapse
Affiliation(s)
- Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Andreas Spannbauer
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Alfred Gugerell
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Dietmar Pils
- Center for Medical Statistics, Informatics, and Intelligent Systems (CeMSIIS), and Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Gerald Maurer
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Andras Jakab
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.,Center for MR-Research, University Children's Hospital Zurich, Steinwiesstraße 75, 8032 Zurich, Switzerland
| | - Martin Riesenhuber
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Andreas Pircher
- Division of Hematology and Oncology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Johannes Winkler
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Jutta Bergler-Klein
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| |
Collapse
|
25
|
Gliozzi M, Scarano F, Musolino V, Carresi C, Scicchitano M, Ruga S, Zito MC, Nucera S, Bosco F, Maiuolo J, Macrì R, Guarnieri L, Mollace R, Coppoletta AR, Nicita C, Tavernese A, Palma E, Muscoli C, Mollace V. Role of TSPO/VDAC1 Upregulation and Matrix Metalloproteinase-2 Localization in the Dysfunctional Myocardium of Hyperglycaemic Rats. Int J Mol Sci 2020; 21:ijms21207432. [PMID: 33050121 PMCID: PMC7587933 DOI: 10.3390/ijms21207432] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/06/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Clinical management of diabetic cardiomyopathy represents an unmet need owing to insufficient knowledge about the molecular mechanisms underlying the dysfunctional heart. The aim of this work is to better clarify the role of matrix metalloproteinase 2 (MMP-2) isoforms and of translocator protein (TSPO)/voltage-dependent anion-selective channel 1 (VDAC1) modulation in the development of hyperglycaemia-induced myocardial injury. Hyperglycaemia was induced in Sprague-Dawley rats through a streptozocin injection (35 mg/Kg, i.p.). After 60 days, cardiac function was analysed by echocardiography. Nicotinamide Adenine Dinucleotide Phosphate NADPH oxidase and TSPO expression was assessed by immunohistochemistry. MMP-2 activity was detected by zymography. Superoxide anion production was estimated by MitoSOX™ staining. Voltage-dependent anion-selective channel 1 (VDAC-1), B-cell lymphoma 2 (Bcl-2), and cytochrome C expression was assessed by Western blot. Hyperglycaemic rats displayed cardiac dysfunction; this response was characterized by an overexpression of NADPH oxidase, accompanied by an increase of superoxide anion production. Under hyperglycaemia, increased expression of TSPO and VDAC1 was detected. MMP-2 downregulated activity occurred under hyperglycemia and this profile of activation was accompanied by the translocation of intracellular N-terminal truncated isoform of MMP-2 (NT-MMP-2) from mitochondria-associated membrane (MAM) into mitochondria. In the onset of diabetic cardiomyopathy, mitochondrial impairment in cardiomyocytes is characterized by the dysregulation of the different MMP-2 isoforms. This can imply the generation of a “frail” myocardial tissue unable to adapt itself to stress.
Collapse
Affiliation(s)
- Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
- Correspondence: ; Tel.: +39-0961-3694301
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Maria Caterina Zito
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Roberta Macrì
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Rocco Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
| | - Caterina Nicita
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
| | - Annamaria Tavernese
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
- Division of Cardiology, University Hospital Policlinico Tor Vergata, 00133 Rome, Italy
| | - Ernesto Palma
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
| | - Carolina Muscoli
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
- IRCCS San Raffaele Pisana, Via di Valcannuta, 00163 Rome, Italy
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (F.S.); (V.M.); (C.C.); (M.S.); (S.R.); (M.C.Z.); (S.N.); (F.B.); (J.M.); (R.M.); (L.G.); (R.M.); (A.R.C.); (C.N.); (E.P.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Presso Fondazione Terina, 88046 Lamezia Terme (CZ), Italy;
- IRCCS San Raffaele Pisana, Via di Valcannuta, 00163 Rome, Italy
| |
Collapse
|
26
|
Gliozzi M, Musolino V, Bosco F, Scicchitano M, Scarano F, Nucera S, Zito MC, Ruga S, Carresi C, Macrì R, Guarnieri L, Maiuolo J, Tavernese A, Coppoletta AR, Nicita C, Mollace R, Palma E, Muscoli C, Belzung C, Mollace V. Cholesterol homeostasis: Researching a dialogue between the brain and peripheral tissues. Pharmacol Res 2020; 163:105215. [PMID: 33007421 DOI: 10.1016/j.phrs.2020.105215] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Cholesterol homeostasis is a highly regulated process in human body because of its several functions underlying the biology of cell membranes, the synthesis of all steroid hormones and bile acids and the need of trafficking lipids destined to cell metabolism. In particular, it has been recognized that peripheral and central nervous system cholesterol metabolism are separated by the blood brain barrier and are regulated independently; indeed, peripherally, it depends on the balance between dietary intake and hepatic synthesis on one hand and its degradation on the other, whereas in central nervous system it is synthetized de novo to ensure brain physiology. In view of this complex metabolism and its relevant functions in mammalian, impaired levels of cholesterol can induce severe cellular dysfunction leading to metabolic, cardiovascular and neurodegenerative diseases. The aim of this review is to clarify the role of cholesterol homeostasis in health and disease highlighting new intriguing aspects of the cross talk between its central and peripheral metabolism.
Collapse
Affiliation(s)
- Micaela Gliozzi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Francesca Bosco
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Federica Scarano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Saverio Nucera
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Stefano Ruga
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Cristina Carresi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Roberta Macrì
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Annamaria Tavernese
- Division of Cardiology, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Caterina Nicita
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Rocco Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Ernesto Palma
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| | | | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| |
Collapse
|
27
|
Baburina Y, Odinokova I, Krestinina O. The Effects of PK11195 and Protoporphyrin IX Can Modulate Chronic Alcohol Intoxication in Rat Liver Mitochondria under the Opening of the Mitochondrial Permeability Transition Pore. Cells 2020; 9:cells9081774. [PMID: 32722345 PMCID: PMC7463720 DOI: 10.3390/cells9081774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Decades of active research have shown that mitochondrial dysfunction, the associated oxidative stress, impaired anti-stress defense mechanisms, and the activation of the proapoptotic signaling pathways underlie pathological changes in organs and tissues. Pathologies caused by alcohol primarily affect the liver. Alcohol abuse is the cause of many liver diseases, such as steatosis, alcoholic steatohepatitis, fibrosis, cirrhosis, and, potentially, hepatocellular cancer. In this study, the effect of chronic alcohol exposure on rat liver mitochondria was investigated. We observed an ethanol-induced increase in sensitivity to calcium, changes in the level of protein kinase Akt and GSK-3β phosphorylation, an induction of the mitochondrial permeability transition pore (mPTP), and strong alterations in the expression of mPTP regulators. Moreover, we also showed an enhanced effect of PK11195 and PPIX, on the parameters of the mPTP opening in rat liver mitochondria (RLM) isolated from ethanol-treated rats compared to the RLM from control rats. We suggest that the results of this study could help elucidate the mechanisms of chronic ethanol action on the mitochondria and contribute to the development of new therapeutic strategies for treating the effects of ethanol-related diseases.
Collapse
|
28
|
Adameova A, Shah AK, Dhalla NS. Role of Oxidative Stress in the Genesis of Ventricular Arrhythmias. Int J Mol Sci 2020; 21:E4200. [PMID: 32545595 PMCID: PMC7349053 DOI: 10.3390/ijms21124200] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Ventricular arrhythmias, mainly lethal arrhythmias, such as ventricular tachycardia and fibrillation, may lead to sudden cardiac death. These are triggered as a result of cardiac injury due to chronic ischemia, acute myocardial infarction and various stressful conditions associated with increased levels of circulating catecholamines and angiotensin II. Several mechanisms have been proposed to underlie electrical instability of the heart promoting ventricular arrhythmias; however, oxidative stress which adversely affects ion homeostasis due to changes in the ion channel structure and function, seems to play a critical role in eliciting different types of ventricular arrhythmias. Prevention or mitigation of the severity of ventricular arrhythmias due to antioxidants has been indicated as the fundamental contribution in the field of preventive cardiology; however, novel interventions have to be developed for greater effectiveness and specificity in attenuating the adverse effects of oxidative stress. In this review, we have attempted to discuss proarrhythmic effects of oxidative stress differing in time and concentration dependence and highlight a molecular and cellular concept how it alters cardiac cell automaticity and conduction velocity sensitizing the probability of ventricular arrhythmias with resultant sudden cardiac death due to ischemic heart disease and other stressful situations. It is concluded that pharmacological approaches targeting multiple mechanisms besides oxidative stress might be more effective in the treatment of ventricular arrhythmias than current antiarrhythmic therapy.
Collapse
Affiliation(s)
- Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, and Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Anureet K. Shah
- Department of Kinesiology, Nutrition and Food Science, California State University, Los Angeles, CA 90032, USA;
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada;
| |
Collapse
|
29
|
Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, Papadopoulos V, Troadec JD, Leprince J. Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther 2020; 208:107386. [DOI: 10.1016/j.pharmthera.2019.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
|
30
|
Jurkiewicz P, Senicourt L, Ayeb H, Lequin O, Lacapere JJ, Batoko H. A Plant-Specific N-terminal Extension Reveals Evolutionary Functional Divergence within Translocator Proteins. iScience 2020; 23:100889. [PMID: 32087576 PMCID: PMC7033594 DOI: 10.1016/j.isci.2020.100889] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/30/2019] [Accepted: 02/03/2020] [Indexed: 12/13/2022] Open
Abstract
Conserved translocator proteins (TSPOs) mediate cell stress responses possibly in a cell-type-specific manner. This work reports on the molecular function of plant TSPO and their possible evolutionary divergence. Arabidopsis thaliana TSPO (AtTSPO) is stress induced and has a conserved polybasic, plant-specific N-terminal extension. AtTSPO reduces water loss by depleting aquaporin PIP2;7 in the plasma membrane. Herein, AtTSPO was found to bind phosphoinositides in vitro, but only full-length AtTSPO or chimeric mouse TSPO with an AtTSPO N-terminus bound PI(4,5)P2in vitro and modified PIP2;7 levels in vivo. Expression of AtTSPO but not its N-terminally truncated variant enhanced phospholipase C activity and depleted PI(4,5)P2 from the plasma membrane and its enrichment in Golgi membranes. Deletion or point mutations within the AtTSPO N-terminus affected PI(4,5)P2 binding and almost prevented AtTSPO-PIP2;7 interaction in vivo. The findings imply functional divergence of plant TSPOs from bacterial and animal counterparts via evolutionary acquisition of the phospholipid-interacting N-terminus.
Collapse
Affiliation(s)
- Pawel Jurkiewicz
- Louvain Institute of Biomolecular Science and Technology (LIBST), University of Louvain (UCLouvain), Croix du Sud 4-5, L7.07.14, 1348 Louvain-la-Neuve, Belgium
| | - Lucile Senicourt
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, 4 Place Jussieu, 75005 Paris, France
| | - Haitham Ayeb
- Louvain Institute of Biomolecular Science and Technology (LIBST), University of Louvain (UCLouvain), Croix du Sud 4-5, L7.07.14, 1348 Louvain-la-Neuve, Belgium
| | - Olivier Lequin
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, 4 Place Jussieu, 75005 Paris, France
| | - Jean-Jacques Lacapere
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, 4 Place Jussieu, 75005 Paris, France
| | - Henri Batoko
- Louvain Institute of Biomolecular Science and Technology (LIBST), University of Louvain (UCLouvain), Croix du Sud 4-5, L7.07.14, 1348 Louvain-la-Neuve, Belgium.
| |
Collapse
|
31
|
Meng Y, Tian M, Yin S, Lai S, Zhou Y, Chen J, He M, Liao Z. Downregulation of TSPO expression inhibits oxidative stress and maintains mitochondrial homeostasis in cardiomyocytes subjected to anoxia/reoxygenation injury. Biomed Pharmacother 2020; 121:109588. [DOI: 10.1016/j.biopha.2019.109588] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/15/2019] [Accepted: 10/26/2019] [Indexed: 02/06/2023] Open
|
32
|
Fantin SM, Parson KF, Niu S, Liu J, Polasky DA, Dixit SM, Ferguson-Miller SM, Ruotolo BT. Collision Induced Unfolding Classifies Ligands Bound to the Integral Membrane Translocator Protein. Anal Chem 2019; 91:15469-15476. [PMID: 31743004 DOI: 10.1021/acs.analchem.9b03208] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Membrane proteins represent most current therapeutic targets, yet remain understudied due to their insolubility in aqueous solvents and generally low yields during purification and expression. Ion mobility-mass spectrometry and collision induced unfolding experiments have recently garnered attention as methods capable of directly detecting and quantifying ligand binding within a wide range of membrane protein systems. Despite prior success, ionized surfactant often creates chemical noise patterns resulting in significant challenges surrounding the study of small membrane protein-ligand complexes. Here, we present a new data analysis workflow that overcomes such chemical noise and then utilize this approach to quantify and classify ligand binding associated with the 36 kDa dimer of translocator protein (TSPO). Following our denoising protocol, we detect separate gas-phase unfolding signatures for lipid and protoporphyrin TSPO binders, molecular classes that likely interact with separate regions of the protein surface. Further, a detailed classification analysis reveals that lipid alkyl chain saturation levels can be detected within our gas-phase protein unfolding data. We combine these data and classification schemes with mass spectra acquired directly from liquid-liquid extracts to propose an identity for a previously unknown endogenous TSPO ligand.
Collapse
Affiliation(s)
- Sarah M Fantin
- Department of Chemistry , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Kristine F Parson
- Department of Chemistry , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Shuai Niu
- Department of Chemistry , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Jian Liu
- Department of Biochemistry and Molecular Biology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Daniel A Polasky
- Department of Chemistry , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Sugyan M Dixit
- Department of Chemistry , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Shelagh M Ferguson-Miller
- Department of Biochemistry and Molecular Biology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Brandon T Ruotolo
- Department of Chemistry , University of Michigan , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
33
|
Astaxanthin Inhibits Mitochondrial Permeability Transition Pore Opening in Rat Heart Mitochondria. Antioxidants (Basel) 2019; 8:antiox8120576. [PMID: 31766490 PMCID: PMC6943429 DOI: 10.3390/antiox8120576] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022] Open
Abstract
The mitochondrion is the main organelle of oxidative stress in cells. Increased permeability of the inner mitochondrial membrane is a key phenomenon in cell death. Changes in membrane permeability result from the opening of the mitochondrial permeability transition pore (mPTP), a large-conductance channel that forms after the overload of mitochondria with Ca2+ or in response to oxidative stress. The ketocarotenoid astaxanthin (AST) is a potent antioxidant that is capable of maintaining the integrity of mitochondria by preventing oxidative stress. In the present work, the effect of AST on the functioning of mPTP was studied. It was found that AST was able to inhibit the opening of mPTP, slowing down the swelling of mitochondria by both direct addition to mitochondria and administration. AST treatment changed the level of mPTP regulatory proteins in isolated rat heart mitochondria. Consequently, AST can protect mitochondria from changes in the induced permeability of the inner membrane. AST inhibited serine/threonine protein kinase B (Akt)/cAMP-responsive element-binding protein (CREB) signaling pathways in mitochondria, which led to the prevention of mPTP opening. Since AST improves the resistance of rat heart mitochondria to Ca2+-dependent stress, it can be assumed that after further studies, this antioxidant will be considered an effective tool for improving the functioning of the heart muscle in general under normal and medical conditions.
Collapse
|
34
|
Malaguti M, Cardenia V, Rodriguez-Estrada MT, Hrelia S. Nutraceuticals and physical activity: Their role on oxysterols-mediated neurodegeneration. J Steroid Biochem Mol Biol 2019; 193:105430. [PMID: 31325497 DOI: 10.1016/j.jsbmb.2019.105430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/07/2023]
Abstract
Over the past few years, the contribution of oxysterols to the onset and development of some of the major neurodegenerative diseases (such as Alzheimer's and Parkinson's diseases) has been scientifically asserted, being mainly related to altered brain cholesterol homeostasis. To counteract oxysterol induced inflammation at neuronal level, one possible intervention approach is the administration of some nutrients and/or plant secondary metabolites. On the other hand, the pleiotropic beneficial effects of physical activity seem to play an important role on prevention and counteraction of neurodegenerative diseases, through the modulation of oxysterol homeostasis and the prevention of demyelination. The present review provides a picture of the promising role of nutraceuticals and physical activity on oxysterol-mediated neurodegeneration, pointing out also the different in vitro and in vivo aspects that need to be further investigated for a better understanding of the association of these three counterparts and their overall effect on people at increased risk for neurodegenerative diseases.
Collapse
Affiliation(s)
- Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Rimini, 47921, Italy.
| | - Vladimiro Cardenia
- Department of Agricultural, Forest and Food Sciences DISAFA, University of Turin, Largo Braccini 2, 10095, Grugliasco, Italy
| | | | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Rimini, 47921, Italy
| |
Collapse
|
35
|
Naryzhnaya NV, Maslov LN, Oeltgen PR. Pharmacology of mitochondrial permeability transition pore inhibitors. Drug Dev Res 2019; 80:1013-1030. [PMID: 31823411 DOI: 10.1002/ddr.21593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Natalia V. Naryzhnaya
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Leonid N. Maslov
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Peter R. Oeltgen
- Department of PathologyUniversity of Kentucky College of Medicine Lexington Kentucky
| |
Collapse
|
36
|
Myocardial Inflammation Predicts Remodeling and Neuroinflammation After Myocardial Infarction. J Am Coll Cardiol 2019; 71:263-275. [PMID: 29348018 DOI: 10.1016/j.jacc.2017.11.024] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/01/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The local inflammatory tissue response after acute myocardial infarction (MI) determines subsequent healing. Systemic interaction may induce neuroinflammation as a precursor to neurodegeneration. OBJECTIVES This study sought to assess the influence of MI on cardiac and brain inflammation using noninvasive positron emission tomography (PET) of the heart-brain axis. METHODS After coronary artery ligation or sham surgery, mice (n = 49) underwent serial whole-body PET imaging of the mitochondrial translocator protein (TSPO) as a marker of activated macrophages and microglia. Patients after acute MI (n = 3) were also compared to healthy controls (n = 9). RESULTS Infarct mice exhibited elevated myocardial TSPO signal at 1 week versus sham (percent injected dose per gram: 8.0 ± 1.6 vs. 4.8 ± 0.9; p < 0.001), localized to activated CD68+ inflammatory cells in the infarct. Early TSPO signal predicted subsequent left ventricular remodeling at 8 weeks (rpartial = -0.687; p = 0.001). In parallel, brain TSPO signal was elevated at 1 week (1.7 ± 0.2 vs. 1.4 ± 0.2 for sham; p = 0.017), localized to activated microglia. After interval decline at 4 weeks, progressive heart failure precipitated a second wave of neuroinflammation (1.8 ± 0.2; p = 0.005). TSPO was concurrently up-regulated in remote cardiomyocytes at 8 weeks (8.8 ± 1.7, p < 0.001) without inflammatory cell infiltration, suggesting mitochondrial impairment. Angiotensin-converting enzyme inhibitor treatment lowered acute inflammation in the heart (p = 0.003) and brain (p = 0.06) and improved late cardiac function (p = 0.05). Patients also demonstrated elevation of cardiac TSPO signal in the infarct territory, paralleled by neuroinflammation versus controls. CONCLUSIONS The brain is susceptible to acute MI and chronic heart failure. Immune activation may interconnect heart and brain dysfunction, a finding that provides a foundation for strategies to improve heart and brain outcomes.
Collapse
|
37
|
Morin D, Long R, Panel M, Laure L, Taranu A, Gueguen C, Pons S, Leoni V, Caccia C, Vatner SF, Vatner DE, Qiu H, Depre C, Berdeaux A, Ghaleh B. Hsp22 overexpression induces myocardial hypertrophy, senescence and reduced life span through enhanced oxidative stress. Free Radic Biol Med 2019; 137:194-200. [PMID: 31047988 DOI: 10.1016/j.freeradbiomed.2019.04.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/12/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
Abstract
H11 kinase/Hsp22 (Hsp22) is a small heat shock protein, which, when overexpressed cardiac specifically in transgenic (TG) mice, induces stable left ventricular (LV) hypertrophy. Hsp22 also increases oxidative phosphorylation and mitochondrial reactive oxygen species (ROS) production, mechanisms mediating LV hypertrophy, senescence and reduced lifespan. Therefore, we investigated whether ROS production mediates LV hypertrophy, senescence and reduced life span in Hsp22 TG mice. Survival curves revealed that TG mice had a 48% reduction in their mean life span compared to wild type (WT) mice. This was associated with a significant increase in senescence markers, such as p16, p19 mRNA levels as well as the percentage of β-galactosidase positive cells and telomerase activity. Oxidized (GSSG)/reduced (GSH) glutathione ratio, an indicator of oxidative stress, and ROS production from 3 major cellular sources was measured in cardiac tissue. Hearts from TG mice exhibited a decrease in GSH/GSSG ratio together with increased ROS production from all sources. To study the role of ROS, mice were treated with the antioxidant Tempol from weaning to their sacrifice. Chronic Tempol treatment abolished oxidative stress and overproduction of ROS, and reduced myocardial hypertrophy and Akt phosphorylation in TG mice. Tempol also significantly extended life span and prevented aging markers in TG mice. Taken together these results show that overexpression of Hsp22 increases oxidative stress responsible for the induction of hypertrophy and senescence and ultimately reduction in life span.
Collapse
Affiliation(s)
- Didier Morin
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France.
| | - Romain Long
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Mathieu Panel
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Lydie Laure
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Adela Taranu
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Cindy Gueguen
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Sandrine Pons
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Valerio Leoni
- Laboratory Medicine, Desio Hospital, University of Milano Bicocca, Milan, Italy
| | - Claudio Caccia
- Laboratory of Clinical Pathology and Medical Genetics, Institute Neurologico IRCCS Carlo Besta, Milano, Italy
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Hongyu Qiu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Christophe Depre
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Alain Berdeaux
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Bijan Ghaleh
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| |
Collapse
|
38
|
Novel Molecular Targets Participating in Myocardial Ischemia-Reperfusion Injury and Cardioprotection. Cardiol Res Pract 2019; 2019:6935147. [PMID: 31275641 PMCID: PMC6558612 DOI: 10.1155/2019/6935147] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Worldwide morbidity and mortality from acute myocardial infarction (AMI) and related heart failure remain high. While effective early reperfusion of the criminal coronary artery after a confirmed AMI is the typical treatment at present, collateral myocardial ischemia-reperfusion injury (MIRI) and pertinent cardioprotection are still challenging to address and have inadequately understood mechanisms. Therefore, unveiling the related novel molecular targets and networks participating in triggering and resisting the pathobiology of MIRI is a promising and valuable frontier. The present study specifically focuses on the recent MIRI advances that are supported by sophisticated bio-methodology in order to bring the poorly understood interrelationship among pro- and anti-MIRI participant molecules up to date, as well as to identify findings that may facilitate the further investigation of novel targets.
Collapse
|
39
|
Torrezan-Nitao E, Figueiredo RCBQ, Marques-Santos LF. Mitochondrial permeability transition pore in sea urchin female gametes. Mech Dev 2018; 154:208-218. [DOI: 10.1016/j.mod.2018.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/21/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022]
|
40
|
TSPO Ligands Promote Cholesterol Efflux and Suppress Oxidative Stress and Inflammation in Choroidal Endothelial Cells. Int J Mol Sci 2018; 19:ijms19123740. [PMID: 30477223 PMCID: PMC6321017 DOI: 10.3390/ijms19123740] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023] Open
Abstract
Choroidal endothelial cells supply oxygen and nutrients to retinal pigment epithelial (RPE) cells and photoreceptors, recycle metabolites, and dispose of metabolic waste through the choroidal blood circulation. Death of the endothelial cells of the choroid may cause abnormal deposits including unesterified and esterified cholesterol beneath RPE cells and within Bruch’s membrane that contribute to the progression of age-related macular degeneration (AMD), the most prevalent cause of blindness in older people. Translocator protein (TSPO) is a cholesterol-binding protein that is involved in mitochondrial cholesterol transport and other cellular functions. We have investigated the role of TSPO in choroidal endothelial cells. Immunocytochemistry showed that TSPO was localized to the mitochondria of choroidal endothelial cells. Choroidal endothelial cells exposed to TSPO ligands (Etifoxine or XBD-173) had significantly increased cholesterol efflux, higher expression of cholesterol homeostasis genes (LXRα, CYP27A1, CYP46A1, ABCA1 and ABCG1), and reduced biosynthesis of cholesterol and phospholipids from [14C]acetate, when compared to untreated controls. Treatment with TSPO ligands also resulted in reduced production of reactive oxygen species (ROS), increased antioxidant capacity, and reduced release of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α and VEGF) induced by oxidized LDL. These data suggest TSPO ligands may offer promise for the treatment of AMD.
Collapse
|
41
|
Svensson F, Zoufir A, Mahmoud S, Afzal AM, Smit I, Giblin KA, Clements PJ, Mettetal JT, Pointon A, Harvey JS, Greene N, Williams RV, Bender A. Information-Derived Mechanistic Hypotheses for Structural Cardiotoxicity. Chem Res Toxicol 2018; 31:1119-1127. [PMID: 30350600 DOI: 10.1021/acs.chemrestox.8b00159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adverse events resulting from drug therapy can be a cause of drug withdrawal, reduced and or restricted clinical use, as well as a major economic burden for society. To increase the safety of new drugs, there is a need to better understand the mechanisms causing the adverse events. One way to derive new mechanistic hypotheses is by linking data on drug adverse events with the drugs' biological targets. In this study, we have used data mining techniques and mutual information statistical approaches to find associations between reported adverse events collected from the FDA Adverse Event Reporting System and assay outcomes from ToxCast, with the aim to generate mechanistic hypotheses related to structural cardiotoxicity (morphological damage to cardiomyocytes and/or loss of viability). Our workflow identified 22 adverse event-assay outcome associations. From these associations, 10 implicated targets could be substantiated with evidence from previous studies reported in the literature. For two of the identified targets, we also describe a more detailed mechanism, forming putative adverse outcome pathways associated with structural cardiotoxicity. Our study also highlights the difficulties deriving these type of associations from the very limited amount of data available.
Collapse
Affiliation(s)
- Fredrik Svensson
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| | - Azedine Zoufir
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| | - Samar Mahmoud
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| | - Avid M Afzal
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| | - Ines Smit
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| | - Kathryn A Giblin
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| | - Peter J Clements
- GlaxoSmithKline R&D Ltd , Park Road , Ware, Hertfordshire SG12 0DP , United Kingdom
| | - Jerome T Mettetal
- Drug Safety and Metabolism , AstraZeneca , 35 Gatehouse Drive , Waltham , Massachusetts 02451 , United States
| | - Amy Pointon
- Safety and ADME Translational Sciences , AstraZeneca , Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - James S Harvey
- GlaxoSmithKline R&D Ltd , Park Road , Ware, Hertfordshire SG12 0DP , United Kingdom
| | - Nigel Greene
- Predictive Compound Safety and ADME , AstraZeneca , 35 Gatehouse Drive , Waltham , Massachusetts 02451 , United States
| | - Richard V Williams
- Lhasa Limited , Granary Wharf House, 2 Canal Wharf , Leeds LS11 5PS , United Kingdom
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , United Kingdom
| |
Collapse
|
42
|
Cardiac-specific Conditional Knockout of the 18-kDa Mitochondrial Translocator Protein Protects from Pressure Overload Induced Heart Failure. Sci Rep 2018; 8:16213. [PMID: 30385779 PMCID: PMC6212397 DOI: 10.1038/s41598-018-34451-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 10/18/2018] [Indexed: 01/17/2023] Open
Abstract
Heart failure (HF) is characterized by abnormal mitochondrial calcium (Ca2+) handling, energy failure and impaired mitophagy resulting in contractile dysfunction and myocyte death. We have previously shown that the 18-kDa mitochondrial translocator protein of the outer mitochondrial membrane (TSPO) can modulate mitochondrial Ca2+ uptake. Experiments were designed to test the role of the TSPO in a murine pressure-overload model of HF induced by transverse aortic constriction (TAC). Conditional, cardiac-specific TSPO knockout (KO) mice were generated using the Cre-loxP system. TSPO-KO and wild-type (WT) mice underwent TAC for 8 weeks. TAC-induced HF significantly increased TSPO expression in WT mice, associated with a marked reduction in systolic function, mitochondrial Ca2+ uptake, complex I activity and energetics. In contrast, TSPO-KO mice undergoing TAC had preserved ejection fraction, and exhibited fewer clinical signs of HF and fibrosis. Mitochondrial Ca2+ uptake and energetics were restored in TSPO KO mice, associated with decreased ROS, improved complex I activity and preserved mitophagy. Thus, HF increases TSPO expression, while preventing this increase limits the progression of HF, preserves ATP production and decreases oxidative stress, thereby preventing metabolic failure. These findings suggest that pharmacological interventions directed at TSPO may provide novel therapeutics to prevent or treat HF.
Collapse
|
43
|
Ilkan Z, Akar FG. The Mitochondrial Translocator Protein and the Emerging Link Between Oxidative Stress and Arrhythmias in the Diabetic Heart. Front Physiol 2018; 9:1518. [PMID: 30416455 PMCID: PMC6212558 DOI: 10.3389/fphys.2018.01518] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023] Open
Abstract
The mitochondrial translocator protein (TSPO) is a key outer mitochondrial membrane protein that regulates the activity of energy-dissipating mitochondrial channels in response to oxidative stress. In this article, we provide an overview of the role of TSPO in the systematic amplification of reactive oxygen species (ROS) through an autocatalytic process known as ROS-induced ROS-release (RIRR). We describe how this TSPO-driven process destabilizes the mitochondrial membrane potential leading to electrical instability at the cellular and whole heart levels. Finally, we provide our perspective on the role of TSPO in the pathophysiology of diabetes, in general and diabetes-related arrhythmias, in particular.
Collapse
Affiliation(s)
- Zeki Ilkan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fadi G Akar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
44
|
Panel M, Ghaleh B, Morin D. Mitochondria and aging: A role for the mitochondrial transition pore? Aging Cell 2018; 17:e12793. [PMID: 29888494 PMCID: PMC6052406 DOI: 10.1111/acel.12793] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
The cellular mechanisms responsible for aging are poorly understood. Aging is considered as a degenerative process induced by the accumulation of cellular lesions leading progressively to organ dysfunction and death. The free radical theory of aging has long been considered the most relevant to explain the mechanisms of aging. As the mitochondrion is an important source of reactive oxygen species (ROS), this organelle is regarded as a key intracellular player in this process and a large amount of data supports the role of mitochondrial ROS production during aging. Thus, mitochondrial ROS, oxidative damage, aging, and aging-dependent diseases are strongly connected. However, other features of mitochondrial physiology and dysfunction have been recently implicated in the development of the aging process. Here, we examine the potential role of the mitochondrial permeability transition pore (mPTP) in normal aging and in aging-associated diseases.
Collapse
Affiliation(s)
- Mathieu Panel
- INSERM U955, équipe 3; Créteil France
- Université Paris-Est, UMR_S955, DHU A-TVB, UPEC; Créteil France
| | - Bijan Ghaleh
- INSERM U955, équipe 3; Créteil France
- Université Paris-Est, UMR_S955, DHU A-TVB, UPEC; Créteil France
| | - Didier Morin
- INSERM U955, équipe 3; Créteil France
- Université Paris-Est, UMR_S955, DHU A-TVB, UPEC; Créteil France
| |
Collapse
|
45
|
Zhu H, Sun A. Programmed necrosis in heart disease: Molecular mechanisms and clinical implications. J Mol Cell Cardiol 2018; 116:125-134. [DOI: 10.1016/j.yjmcc.2018.01.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/24/2017] [Accepted: 01/31/2018] [Indexed: 02/05/2023]
|
46
|
Notter T, Coughlin JM, Gschwind T, Weber-Stadlbauer U, Wang Y, Kassiou M, Vernon AC, Benke D, Pomper MG, Sawa A, Meyer U. Translational evaluation of translocator protein as a marker of neuroinflammation in schizophrenia. Mol Psychiatry 2018; 23:323-334. [PMID: 28093569 DOI: 10.1038/mp.2016.248] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/14/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) imaging with radiotracers that target translocator protein 18 kDa (TSPO) has become a popular approach to assess putative neuroinflammatory processes and associated microglia activation in psychotic illnesses. It remains unclear, however, whether TSPO imaging can accurately capture low-grade inflammatory processes such as those present in schizophrenia and related disorders. Therefore, we evaluated the validity of TSPO as a disease-relevant marker of inflammation using a translational approach, which combined neurodevelopmental and neurodegenerative mouse models with PET imaging in patients with recent-onset schizophrenia and matched controls. Using an infection-mediated neurodevelopmental mouse model, we show that schizophrenia-relevant behavioral abnormalities and increased inflammatory cytokine expression are associated with reduced prefrontal TSPO levels. On the other hand, TSPO was markedly upregulated in a mouse model of acute neurodegeneration and reactive gliosis, which was induced by intrahippocampal injection of kainic acid. In both models, the changes in TSPO levels were not restricted to microglia but emerged in various cell types, including microglia, astrocytes and vascular endothelial cells. Human PET imaging using the second-generation TSPO radiotracer [11C]DPA-713 revealed a strong trend towards reduced TSPO binding in the middle frontal gyrus of patients with recent-onset schizophrenia, who were previously shown to display increased levels of inflammatory cytokines in peripheral and central tissues. Together, our findings challenge the common assumption that central low-grade inflammation in schizophrenia is mirrored by increased TSPO expression or ligand binding. Our study further underscores the need to interpret altered TSPO binding in schizophrenia with caution, especially when measures of TSPO are not complemented with other markers of inflammation. Unless more selective microglial markers are available for PET imaging, quantification of cytokines and other inflammatory biomarkers, along with their molecular signaling pathways, may be more accurate in attempts to characterize inflammatory profiles in schizophrenia and other mental disorders that lack robust reactive gliosis.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T Gschwind
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - U Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Y Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - M Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW, Australia
- Discipline of Medical Radiation Sciences, The University of Sydney, Sydney, NSW, Australia
| | - A C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - D Benke
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - M G Pomper
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
47
|
Coupling between physiological TSPO expression in brain and myocardium allows stabilization of late-phase cerebral [18F]GE180 PET quantification. Neuroimage 2018; 165:83-91. [DOI: 10.1016/j.neuroimage.2017.10.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 01/31/2023] Open
|
48
|
Regulation of Mitochondrial, Cellular, and Organismal Functions by TSPO. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:103-136. [PMID: 29413517 DOI: 10.1016/bs.apha.2017.09.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In 1999, the enigma of the 18kDa mitochondrial translocator protein (TSPO), also known as the peripheral-type benzodiazepine receptor, was the seeming disparity of the many functions attributed to TSPO, ranging from the potential of TSPO acting as a housekeeping gene at molecular biological levels to adaptations to stress, and even involvement in higher emotional and cognitive functioning, such as anxiety and depression. In the years since then, knowledge regarding the many functions modulated by TSPO has expanded, and understanding has deepened. In addition, new functions could be firmly associated with TSPO, such as regulation of programmed cell death and modulation of gene expression. Interestingly, control by the mitochondrial TSPO over both of these life and death functions appears to include Ca++ homeostasis, generation of reactive oxygen species (ROS), and ATP production. Other mitochondrial functions under TSPO control are considered to be steroidogenesis and tetrapyrrole metabolism. As TSPO effects on gene expression and on programmed cell death can be related to the wide range of functions that can be associated with TSPO, several of these five elements of Ca++, ROS, ATP, steroids, and tetrapyrroles may indeed form the basis of TSPO's capability to operate as a multifunctional housekeeping gene to maintain homeostasis of the cell and of the whole multicellular organism.
Collapse
|
49
|
The macrophage marker translocator protein (TSPO) is down-regulated on pro-inflammatory 'M1' human macrophages. PLoS One 2017; 12:e0185767. [PMID: 28968465 PMCID: PMC5624624 DOI: 10.1371/journal.pone.0185767] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/19/2017] [Indexed: 12/14/2022] Open
Abstract
The translocator protein (TSPO) is a mitochondrial membrane protein, of as yet uncertain function. Its purported high expression on activated macrophages, has lent utility to TSPO targeted molecular imaging in the form of positron emission tomography (PET), as a means to detect and quantify inflammation in vivo. However, existing literature regarding TSPO expression on human activated macrophages is lacking, mostly deriving from brain tissue studies, including studies of brain malignancy, and inflammatory diseases such as multiple sclerosis. Here, we utilized three human sources of monocyte derived macrophages (MDM), from THP-1 monocytes, healthy peripheral blood monocytes and synovial fluid monocytes from patients with rheumatoid arthritis, to undertake a detailed investigation of TSPO expression in activated macrophages. In this work, we demonstrate a consistent down-regulation of TSPO mRNA and protein in macrophages activated to a pro-inflammatory, or ‘M1’ phenotype. Conversely, stimulation of macrophages to an M2 phenotype with IL-4, dexamethasone or TGF-β1 did not alter TSPO expression, regardless of MDM source. The reasons for this are uncertain, but our study findings add some supporting evidence for recent investigations concluding that TSPO may be involved in negative regulation of inflammatory responses in macrophages.
Collapse
|
50
|
Musman J, Paradis S, Panel M, Pons S, Barau C, Caccia C, Leoni V, Ghaleh B, Morin D. A TSPO ligand prevents mitochondrial sterol accumulation and dysfunction during myocardial ischemia-reperfusion in hypercholesterolemic rats. Biochem Pharmacol 2017. [DOI: 10.1016/j.bcp.2017.06.125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|