201
|
Hamilton S, Terentyeva R, Perger F, Hernández Orengo B, Martin B, Gorr MW, Belevych AE, Clements RT, Györke S, Terentyev D. MCU overexpression evokes disparate dose-dependent effects on mito-ROS and spontaneous Ca 2+ release in hypertrophic rat cardiomyocytes. Am J Physiol Heart Circ Physiol 2021; 321:H615-H632. [PMID: 34415186 PMCID: PMC8794228 DOI: 10.1152/ajpheart.00126.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022]
Abstract
Cardiac dysfunction in heart failure (HF) and diabetic cardiomyopathy (DCM) is associated with aberrant intracellular Ca2+ handling and impaired mitochondrial function accompanied with reduced mitochondrial calcium concentration (mito-[Ca2+]). Pharmacological or genetic facilitation of mito-Ca2+ uptake was shown to restore Ca2+ transient amplitude in DCM and HF, improving contractility. However, recent reports suggest that pharmacological enhancement of mito-Ca2+ uptake can exacerbate ryanodine receptor-mediated spontaneous sarcoplasmic reticulum (SR) Ca2+ release in ventricular myocytes (VMs) from diseased animals, increasing propensity to stress-induced ventricular tachyarrhythmia. To test whether chronic recovery of mito-[Ca2+] restores systolic Ca2+ release without adverse effects in diastole, we overexpressed mitochondrial Ca2+ uniporter (MCU) in VMs from male rat hearts with hypertrophy induced by thoracic aortic banding (TAB). Measurement of mito-[Ca2+] using genetic probe mtRCamp1h revealed that mito-[Ca2+] in TAB VMs paced at 2 Hz under β-adrenergic stimulation is lower compared with shams. Adenoviral 2.5-fold MCU overexpression in TAB VMs fully restored mito-[Ca2+]. However, it failed to improve cytosolic Ca2+ handling and reduce proarrhythmic spontaneous Ca2+ waves. Furthermore, mitochondrial-targeted genetic probes MLS-HyPer7 and OMM-HyPer revealed a significant increase in emission of reactive oxygen species (ROS) in TAB VMs with 2.5-fold MCU overexpression. Conversely, 1.5-fold MCU overexpression in TABs, that led to partial restoration of mito-[Ca2+], reduced mitochondria-derived reactive oxygen species (mito-ROS) and spontaneous Ca2+ waves. Our findings emphasize the key role of elevated mito-ROS in disease-related proarrhythmic Ca2+ mishandling. These data establish nonlinear mito-[Ca2+]/mito-ROS relationship, whereby partial restoration of mito-[Ca2+] in diseased VMs is protective, whereas further enhancement of MCU-mediated Ca2+ uptake exacerbates damaging mito-ROS emission.NEW & NOTEWORTHY Defective intracellular Ca2+ homeostasis and aberrant mitochondrial function are common features in cardiac disease. Here, we directly compared potential benefits of mito-ROS scavenging and restoration of mito-Ca2+ uptake by overexpressing MCU in ventricular myocytes from hypertrophic rat hearts. Experiments using novel mito-ROS and Ca2+ biosensors demonstrated that mito-ROS scavenging rescued both cytosolic and mito-Ca2+ homeostasis, whereas moderate and high MCU overexpression demonstrated disparate effects on mito-ROS emission, with only a moderate increase in MCU being beneficial.
Collapse
MESH Headings
- Adrenergic beta-Agonists/pharmacology
- Animals
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/physiopathology
- Biosensing Techniques
- Calcium/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Cells, Cultured
- Disease Models, Animal
- Heart Rate
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Microscopy, Confocal
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/genetics
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Up-Regulation
- Ventricular Function, Left
- Ventricular Remodeling
- Rats
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Fruzsina Perger
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Benjamín Hernández Orengo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Benjamin Martin
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Matthew W Gorr
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
- College of Nursing, The Ohio State University, Columbus, Ohio
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Richard T Clements
- Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Sandor Györke
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| |
Collapse
|
202
|
Qi Y, Chen S, Lu Y, Zhang Z, Wang S, Chen N, Shen M, Chen F, Chen M, Quan Y, Yang L, Xu Y, Su Y, Hu M, Wang J. Grape seed proanthocyanidin extract ameliorates ionizing radiation-induced hematopoietic stem progenitor cell injury by regulating Foxo1 in mice. Free Radic Biol Med 2021; 174:144-156. [PMID: 34389464 DOI: 10.1016/j.freeradbiomed.2021.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/27/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022]
Abstract
Ionizing radiation (IR)-induced excessive reactive oxygen species (ROS) is an important contributor of the injury of hematopoietic system. Grape seed proanthocyanidin extract (GSPE) is a new type of antioxidant, whereas whether it could ameliorate IR-induced hematopoietic injury remains unclear. Here, we show that GSPE treatment improves the survival of irradiated mice and alleviates IR-induced myelosuppression. Meanwhile, the hematopoietic reconstituting ability of hematopoietic stem cells (HSCs) in mice following irradiation exposure is significantly increased after GSPE treatment. Furthermore, GSPE treatment can reduce IR-induced ROS production and relieve DNA damage and apoptosis in hematopoietic stem progenitor cells (HSPCs). Interestingly, we find that a critical antioxidant-associated gene fokhead box transcription factor O1 (Foxo1) is significantly decreased in HSPCs after irradiation. Consistently, hematopoietic specific deletion of Foxo1 increases the radiosensitivity of mice. Further investigations reveal that GSPE treatment specifically upregulates the expression of Foxo1, as well as its target genes superoxide dismutase 1 (SOD1), superoxide dismutase 2 (SOD2) and catalase (CAT). Importantly, Foxo1 deficiency largely abolishes the radioprotection of GSPE on HSPCs. Collectively, our data demonstrate that GSPE plays an important role in ameliorating IR-induced HSPC injury via the Foxo1-mediated pathway. Therefore, GSPE may be used as a promising radioprotective agent.
Collapse
Affiliation(s)
- Yan Qi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Shilei Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Yukai Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Zihao Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Song Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Naicheng Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Mingqiang Shen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Fang Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Mo Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Yong Quan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Lijing Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Yang Xu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Yongping Su
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Mengjia Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
203
|
Structure, regulation, and biological functions of TIGAR and its role in diseases. Acta Pharmacol Sin 2021; 42:1547-1555. [PMID: 33510458 PMCID: PMC8463536 DOI: 10.1038/s41401-020-00588-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/22/2020] [Indexed: 02/02/2023] Open
Abstract
TIGAR (TP53-induced glycolysis and apoptosis regulator) is the downstream target gene of p53, contains a functional sequence similar to 6-phosphofructose kinase/fructose-2, 6-bisphosphatase (PFKFB) bisphosphatase domain. TIGAR is mainly located in the cytoplasm; in response to stress, TIGAR is translocated to nucleus and organelles, including mitochondria and endoplasmic reticulum to regulate cell function. P53 family members (p53, p63, and p73), some transcription factors (SP1 and CREB), and noncoding miRNAs (miR-144, miR-885-5p, and miR-101) regulate the transcription of TIGAR. TIGAR mainly functions as fructose-2,6-bisphosphatase to hydrolyze fructose-1,6-diphosphate and fructose-2,6-diphosphate to inhibit glycolysis. TIGAR in turn facilitates pentose phosphate pathway flux to produce nicotinamide adenine dinucleotide phosphate (NADPH) and ribose, thereby promoting DNA repair, and reducing intracellular reactive oxygen species. TIGAR thus maintains energy metabolism balance, regulates autophagy and stem cell differentiation, and promotes cell survival. Meanwhile, TIGAR also has a nonenzymatic function and can interact with retinoblastoma protein, protein kinase B, nuclear factor-kappa B, hexokinase 2, and ATP5A1 to mediate cell cycle arrest, inflammatory response, and mitochondrial protection. TIGAR might be a potential target for the prevention and treatment of cardiovascular and neurological diseases, as well as cancers.
Collapse
|
204
|
Susilowati S, Mustofa I, Wurlina W, Triana IN, Utama S, Rimayanti R. Effect of insulin-like growth factor-1 complex of Simmental bull seminal plasma on post-thawed Kacang buck semen fertility. Vet World 2021; 14:2073-2084. [PMID: 34566323 PMCID: PMC8448655 DOI: 10.14202/vetworld.2021.2073-2084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Background and Aim Kacang buck sperm is cryosensitive due to the seminal plasma of semen itself. Meanwhile, bull seminal plasma contains the insulin-like growth factor-1 (IGF-1) complex, which is cryoprotective. The addition of the crude protein of Simmental bull seminal plasma increased the quality of post-thawed semen of Kacang buck. The study was conducted to determine the effects of Simmental bull seminal plasma with IGF-1 on the fertility of post-thawed Kacang buck semen. Materials and Methods Buck semen was diluted in the following skim milk-egg yolk extender preparations: Without the addition of Simmental bull seminal plasma IGF-1 complex protein (T0); with the addition of 12-μg Simmental bull seminal plasma IGF-1 complex protein (T1); and with the addition of 24-μg Simmental bull seminal plasma IGF-1 complex protein (T2). The extended semen was packed in 0.25-mL straws and frozen. Post-thawed semen fertility was evaluated based on the following variables: Sperm motility, viability, intact plasma membrane (IPM), malondialdehyde (MDA) levels, capacitation status, and acrosome reaction. The difference in each variable among the groups was evaluated using analysis of variance, followed by Tukey's honestly significant difference test, at a 95% level of significance. Meanwhile, principal component analysis (PCA) was used to identify the principal component of semen fertility among the seven parameters. Results The T1 group showed the highest sperm motility, viability, IPM, and percentage of incapacitated sperm and the lowest MDA levels, percentage of capacitated sperm, and acrosome reaction. PCA revealed that sperm motility had a moderate to very robust correlation with other variables and is the most crucial parameter, accounting for 80.79% of all variables. Conclusion The IGF-1 complex in Simmental bull seminal plasma was useful for increasing the fertility of post-thawed Kacang buck semen, and sperm motility was the principal component of semen fertility.
Collapse
Affiliation(s)
- Suherni Susilowati
- Laboratory of Veterinary Artificial Insemination, Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Mulyorejo, Surabaya, Indonesia
| | - Imam Mustofa
- Laboratory of Veterinary Obstetrics, Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Mulyorejo, Surabaya, Indonesia
| | - Wurlina Wurlina
- Laboratory of Veterinary Infertility and Sterility, Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Mulyorejo, Surabaya, Indonesia
| | - Indah Norma Triana
- Laboratory of Veterinary Infertility and Sterility, Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Mulyorejo, Surabaya, Indonesia
| | - Suzanita Utama
- Laboratory of Veterinary Obstetrics, Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Mulyorejo, Surabaya, Indonesia
| | - Rimayanti Rimayanti
- Laboratory of Veterinary Infertility and Sterility, Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Mulyorejo, Surabaya, Indonesia
| |
Collapse
|
205
|
Emerging methods for and novel insights gained by absolute quantification of mitochondrial DNA copy number and its clinical applications. Pharmacol Ther 2021; 232:107995. [PMID: 34592204 DOI: 10.1016/j.pharmthera.2021.107995] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023]
Abstract
The past thirty years have seen a surge in interest in pathophysiological roles of mitochondria, and the accurate quantification of mitochondrial DNA copy number (mCN) in cells and tissue samples is a fundamental aspect of assessing changes in mitochondrial health and biogenesis. Quantification of mCN between studies is surprisingly variable due to a combination of physiological variability and diverse protocols being used to measure this endpoint. The advent of novel methods to quantify nucleic acids like digital polymerase chain reaction (dPCR) and high throughput sequencing offer the ability to measure absolute values of mCN. We conducted an in-depth survey of articles published between 1969 -- 2020 to create an overview of mCN values, to assess consensus values of tissue-specific mCN, and to evaluate consistency between methods of assessing mCN. We identify best practices for methods used to assess mCN, and we address the impact of using specific loci on the mitochondrial genome to determine mCN. Current data suggest that clinical measurement of mCN can provide diagnostic and prognostic value in a range of diseases and health conditions, with emphasis on cancer and cardiovascular disease, and the advent of means to measure absolute mCN should improve future clinical applications of mCN measurements.
Collapse
|
206
|
Yan T, Zhu S, Shi Y, Xie C, Zhu M, Zhang Y, Wang C, Guo C. Pan-Cancer Analysis of Atrial-Fibrillation-Related Innate Immunity Gene ANXA4. Front Cardiovasc Med 2021; 8:713983. [PMID: 34540918 PMCID: PMC8446278 DOI: 10.3389/fcvm.2021.713983] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/16/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Atrial fibrillation (AF) is the most common tachyarrhythmia around the world. Cancer is one of the main causes of death worldwide. A recent study demonstrated that cancer was associated with an increased incidence of AF. In the present study, we aimed to explore possible mechanisms and potential common therapeutic targets between AF and cancer. Methods: Differentially expressed proteins between AF and sinus rhythm were identified utilizing proteomics analysis. Weighted gene correlation network analysis was applied to cluster proteins into different modules and investigate associations between modules and AF. Hub immune-related genes were selected via InnateDB database and verified using qRT-PCR. RNA sequencing and clinical data of 33 different cancer types were achieved from The Cancer Genome Atlas (TCGA). The correlations between ANXA4 expression and the prognosis were calculated utilizing Cox regression analysis and Kaplan-Meier survival analysis. Spearman's rank correlation test was used to assess associations between ANXA4 and immune infiltration and DNA methylation. Enrichment analysis was performed through gene ontology and gene set enrichment analysis (GSEA). Results:ANXA4 was identified as hub immune-related gene between AF and sinus rhythm. Expression levels of ANXA4 increased in diverse cancer types. Survival analysis suggested prognostic significance of ANXA4 expression levels in various cancer types. Immune correlation analysis indicated that ANXA4 expression levels were associated with tumor immune infiltration in most cancer types. ANXA4 might influence the efficacy of immunotherapy via tumor burden and microsatellite instability. GSEA results indicated that high ANXA4 expression groups were mainly enriched in peroxisome, bile acid biosynthesis, and p53 pathway. Conclusion:ANXA4 was identified as a hub immune-related gene in AF, which has never been reported. Pan-cancer analysis indicated its potential as a novel clinical prognostic marker and therapeutic target in diverse cancer types. ANXA4 might play crucial roles in AF and cancer, and targeted therapy for ANXA4 might reduce the incidence of AF in cancer patients.
Collapse
Affiliation(s)
- Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijie Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Shi
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Changming Xie
- Department of Cardiovascular, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Miao Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangyang Zhang
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changfa Guo
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
207
|
Xie W, Xu R, Fan C, Yang C, Chen H, Cao Y. 900 MHz Radiofrequency Field Induces Mitochondrial Unfolded Protein Response in Mouse Bone Marrow Stem Cells. Front Public Health 2021; 9:724239. [PMID: 34513791 PMCID: PMC8428517 DOI: 10.3389/fpubh.2021.724239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/04/2021] [Indexed: 01/29/2023] Open
Abstract
Objective: To examine whether exposure of mouse bone marrow stromal cells (BMSC) to 900 MHz radiofrequency fields used in mobile communication devices can induce mitochondrial unfolded protein response (UPRmt). Methods: BMSCs were exposed to continuous wave 900 MHz radiofrequency fields (RF) at 120 μW/cm2 power intensity for 4 h/d for 5 consecutive days. Cells in sham group (SH) were cultured in RF exposure system, but without RF radiation. The positive control cells were irradiated with 6 Gy X-ray at a dose rate of 1.103 Gy/min (XR). To inhibit the upstream molecular JNK2 of UPRmt, cells in siRNA + RF, and siRNA + XR group were also pretreated with 100 nM siRNA-JNK2 for 48 h before RF/XR exposure. Thirty minutes, 4 h, and 24 h post-RF/XR exposure, cells were collected, the level of ROS was measured with flow cytometry, the expression levels of UPRmt-related proteins were detected using western blot analysis. Results: Compared with Sham group, the level of ROS in RF and XR group was significantly increased 30 min and 4 h post-RF/XR exposure (P < 0.05), however, the RF/XR-induced increase of ROS level reversed 24 h post-RF/XR exposure. Compared with Sham group, the expression levels of HSP10/HSP60/ClpP proteins in cells of RF and XR group increased significantly 30 min and 4 h post-RF/XR exposure (P < 0.05), however, the RF/XR-induced increase of HSP10/HSP60/ClpP protein levels reversed 24 h post-RF exposure. After interfering with siRNA-JNK2, the RF/XR exposures could not induce the increase of HSP10/HSP60/ClpP protein levels any more. Conclusions: The exposure of 900 MHz RF at 120 μW/cm2 power flux density could increase ROS level and activate a transient UPRmt in BMSC cells. Mitochondrial homeostasis in term of protein folding ability is restored 24 h post-RF exposure. Exposure to RF in our experimental condition did not cause permanent and severe mitochondrial dysfunctions. However, the detailed underlying molecular mechanism of RF-induced UPRmt remains to be further studied.
Collapse
Affiliation(s)
- Wen Xie
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
| | - Rui Xu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
| | - Caiyun Fan
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
| | - Chunyu Yang
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
| | - Haiyan Chen
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
| | - Yi Cao
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
| |
Collapse
|
208
|
Zoccarato A, Nabeebaccus AA, Oexner RR, Santos CXC, Shah AM. The nexus between redox state and intermediary metabolism. FEBS J 2021; 289:5440-5462. [PMID: 34496138 DOI: 10.1111/febs.16191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are not just a by-product of cellular metabolic processes but act as signalling molecules that regulate both physiological and pathophysiological processes. A close connection exists in cells between redox homeostasis and cellular metabolism. In this review, we describe how intracellular redox state and glycolytic intermediary metabolism are closely coupled. On the one hand, ROS signalling can control glycolytic intermediary metabolism by direct regulation of the activity of key metabolic enzymes and indirect regulation via redox-sensitive transcription factors. On the other hand, metabolic adaptation and reprogramming in response to physiological or pathological stimuli regulate intracellular redox balance, through mechanisms such as the generation of reducing equivalents. We also discuss the impact of these intermediary metabolism-redox circuits in physiological and disease settings across different tissues. A better understanding of the mechanisms regulating these intermediary metabolism-redox circuits will be crucial to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Adam A Nabeebaccus
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Rafael R Oexner
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
209
|
Zhao T, Wu W, Sui L, Huang Q, Nan Y, Liu J, Ai K. Reactive oxygen species-based nanomaterials for the treatment of myocardial ischemia reperfusion injuries. Bioact Mater 2021; 7:47-72. [PMID: 34466716 PMCID: PMC8377441 DOI: 10.1016/j.bioactmat.2021.06.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/09/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Interventional coronary reperfusion strategies are widely adopted to treat acute myocardial infarction, but morbidity and mortality of acute myocardial infarction are still high. Reperfusion injuries are inevitable due to the generation of reactive oxygen species (ROS) and apoptosis of cardiac muscle cells. However, many antioxidant and anti-inflammatory drugs are largely limited by pharmacokinetics and route of administration, such as short half-life, low stability, low bioavailability, and side effects for treatment myocardial ischemia reperfusion injury. Therefore, it is necessary to develop effective drugs and technologies to address this issue. Fortunately, nanotherapies have demonstrated great opportunities for treating myocardial ischemia reperfusion injury. Compared with traditional drugs, nanodrugs can effectively increase the therapeutic effect and reduces side effects by improving pharmacokinetic and pharmacodynamic properties due to nanodrugs’ size, shape, and material characteristics. In this review, the biology of ROS and molecular mechanisms of myocardial ischemia reperfusion injury are discussed. Furthermore, we summarized the applications of ROS-based nanoparticles, highlighting the latest achievements of nanotechnology researches for the treatment of myocardial ischemia reperfusion injury. Cardiovascular diseases are the leading cause of death worldwide. Researches of the myocardial infarction pathology and development of new treatments have very important scientific significance in the biomedical field. Many nanomaterials have shown amazing therapeutic effects to reduce myocardial damage by eliminating ROS. Nanomaterials effectively reduced myocardial damage through eliminating ROS from NOXs, M-ETC, M-Ca2+, M-mPTP, and RIRR.
Collapse
Affiliation(s)
- Tianjiao Zhao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410087, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Lihua Sui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Yayun Nan
- Geriatric Medical Center, Ningxia People's Hospital, Yinchuan, 750003, China
| | - Jianhua Liu
- Department of Radiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| |
Collapse
|
210
|
Oppedisano F, Mollace R, Tavernese A, Gliozzi M, Musolino V, Macrì R, Carresi C, Maiuolo J, Serra M, Cardamone A, Volterrani M, Mollace V. PUFA Supplementation and Heart Failure: Effects on Fibrosis and Cardiac Remodeling. Nutrients 2021; 13:nu13092965. [PMID: 34578843 PMCID: PMC8471017 DOI: 10.3390/nu13092965] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) characterized by cardiac remodeling is a condition in which inflammation and fibrosis play a key role. Dietary supplementation with n-3 polyunsaturated fatty acids (PUFAs) seems to produce good results. In fact, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have anti-inflammatory and antioxidant properties and different cardioprotective mechanisms. In particular, following their interaction with the nuclear factor erythropoietin 2 related factor 2 (NRF2), the free fatty acid receptor 4 (Ffar4) receptor, or the G-protein coupled receptor 120 (GPR120) fibroblast receptors, they inhibit cardiac fibrosis and protect the heart from HF onset. Furthermore, n-3 PUFAs increase the left ventricular ejection fraction (LVEF), reduce global longitudinal deformation, E/e ratio (early ventricular filling and early mitral annulus velocity), soluble interleukin-1 receptor-like 1 (sST2) and high-sensitive C Reactive protein (hsCRP) levels, and increase flow-mediated dilation. Moreover, lower levels of brain natriuretic peptide (BNP) and serum norepinephrine (sNE) are reported and have a positive effect on cardiac hemodynamics. In addition, they reduce cardiac remodeling and inflammation by protecting patients from HF onset after myocardial infarction (MI). The positive effects of PUFA supplementation are associated with treatment duration and a daily dosage of 1–2 g. Therefore, both the European Society of Cardiology (ESC) and the American College of Cardiology/American Heart Association (ACC/AHA) define dietary supplementation with n-3 PUFAs as an effective therapy for reducing the risk of hospitalization and death in HF patients. In this review, we seek to highlight the most recent studies related to the effect of PUFA supplementation in HF. For that purpose, a PubMed literature survey was conducted with a focus on various in vitro and in vivo studies and clinical trials from 2015 to 2021.
Collapse
Affiliation(s)
- Francesca Oppedisano
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- Correspondence: (F.O.); (V.M.)
| | - Rocco Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Division of Cardiology, University Hospital Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| | - Annamaria Tavernese
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macrì
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Jessica Maiuolo
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Serra
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
| | - Antonio Cardamone
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
| | | | - Vincenzo Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Graecia, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (R.M.); (C.C.); (J.M.); (M.S.); (A.C.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
- Correspondence: (F.O.); (V.M.)
| |
Collapse
|
211
|
Ilyas F, Jones L, Tee SL, Horsfall M, Swan A, Wollaston F, Hecker T, De Pasquale C, Thomas S, Chong W, Stranks S, Mangoni AA, Selvanayagam JB, Chew DP, De Pasquale CG. Acute pleiotropic effects of dapagliflozin in type 2 diabetic patients with heart failure with reduced ejection fraction: a crossover trial. ESC Heart Fail 2021; 8:4346-4352. [PMID: 34382353 PMCID: PMC8497349 DOI: 10.1002/ehf2.13553] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 11/09/2022] Open
Abstract
Aims This study aimed to explore the rapid effects of dapagliflozin in heart failure with reduced ejection fraction (HFrEF). Methods and results We studied the functional, echocardiographic, electrophysiological, lung ultrasound, ambulatory blood pressure (BP), microvascular and macrovascular function, and biochemical effects of 2 week treatment with dapagliflozin in 19 type 2 diabetic HFrEF patients in a double‐blind, crossover, placebo‐controlled trial. Dapagliflozin had no significant effect on clinical, functional, or quality of life parameters. Dapagliflozin reduced systolic BP [114 (105, 131) vs. 106 (98, 113) mmHg, P < 0.01] and diastolic BP [71 (61, 78) vs. 62 (55, 70) mmHg, P < 0.01]. There was no effect on cardiac chamber size, ventricular systolic function, lung ultrasound, or arterial wave reflection. Dapagliflozin increased creatinine [117 (92, 129) vs. 122 (107, 135) μmol/L, P < 0.05] and haemoglobin [135 (118, 138) vs. 136 (123, 144) g/L, P < 0.05]. There was a reduction in ventricular ectopy [1.4 (0.1, 2.9) vs. 0.2 (0.1, 1.4) %, P < 0.05] and an increase in standard deviation of normal heart beat intervals [70 (58, 90) vs. 74 (62, 103), P < 0.05]. Unexpectedly, dapagliflozin increased high‐sensitivity troponin T [25 (19, 37) vs. 28 (20, 42) ng/L, P < 0.01] and reduced reactive hyperaemia index [1.29 (1.21, 1.56) vs. 1.40 (1.23, 1.84), P < 0.05]. Conclusions After 2 weeks, while multiple parameters supported BP reduction and haemoconcentration with dapagliflozin, reduction in cardiac filling pressure, lung water, and functional improvement was not shown. Reduced ventricular ectopic burden suggests an early antiarrhythmic benefit. The small increase in troponin T and the reduction in the reactive hyperaemia index warrant further mechanistic exploration in this treatment of proven mortality benefit in HFrEF.
Collapse
Affiliation(s)
- Fahmida Ilyas
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Lynette Jones
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Su Ling Tee
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Matthew Horsfall
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Amy Swan
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Fiona Wollaston
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Tracy Hecker
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | | | - Simeoni Thomas
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - William Chong
- Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Steve Stranks
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Arduino A Mangoni
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Joseph B Selvanayagam
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Derek P Chew
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| | - Carmine G De Pasquale
- Flinders Medical Centre, Bedford Park, South Australia, Australia.,Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
212
|
The Role of Mitochondrial Dysfunction in Atrial Fibrillation: Translation to Druggable Target and Biomarker Discovery. Int J Mol Sci 2021; 22:ijms22168463. [PMID: 34445167 PMCID: PMC8395135 DOI: 10.3390/ijms22168463] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 01/02/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent and progressive cardiac arrhythmia worldwide and is associated with serious complications such as heart failure and ischemic stroke. Current treatment modalities attenuate AF symptoms and are only moderately effective in halting the arrhythmia. Therefore, there is an urgent need to dissect molecular mechanisms that drive AF. As AF is characterized by a rapid atrial activation rate, which requires a high energy metabolism, a role of mitochondrial dysfunction in AF pathophysiology is plausible. It is well known that mitochondria play a central role in cardiomyocyte function, as they produce energy to support the mechanical and electrical function of the heart. Details on the molecular mechanisms underlying mitochondrial dysfunction are increasingly being uncovered as a contributing factor in the loss of cardiomyocyte function and AF. Considering the high prevalence of AF, investigating the role of mitochondrial impairment in AF may guide the path towards new therapeutic and diagnostic targets. In this review, the latest evidence on the role of mitochondria dysfunction in AF is presented. We highlight the key modulators of mitochondrial dysfunction that drive AF and discuss whether they represent potential targets for therapeutic interventions and diagnostics in clinical AF.
Collapse
|
213
|
Wu AJ, Tong BCK, Huang AS, Li M, Cheung KH. Mitochondrial Calcium Signaling as a Therapeutic Target for Alzheimer's Disease. Curr Alzheimer Res 2021; 17:329-343. [PMID: 31820698 DOI: 10.2174/1567205016666191210091302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 10/17/2019] [Accepted: 12/09/2019] [Indexed: 11/22/2022]
Abstract
Mitochondria absorb calcium (Ca2+) at the expense of the electrochemical gradient generated during respiration. The influx of Ca2+ into the mitochondrial matrix helps maintain metabolic function and results in increased cytosolic Ca2+ during intracellular Ca2+ signaling. Mitochondrial Ca2+ homeostasis is tightly regulated by proteins located in the inner and outer mitochondrial membranes and by the cross-talk with endoplasmic reticulum Ca2+ signals. Increasing evidence indicates that mitochondrial Ca2+ overload is a pathological phenotype associated with Alzheimer's Disease (AD). As intracellular Ca2+ dysregulation can be observed before the appearance of typical pathological hallmarks of AD, it is believed that mitochondrial Ca2+ overload may also play an important role in AD etiology. The high mitochondrial Ca2+ uptake can easily compromise neuronal functions and exacerbate AD progression by impairing mitochondrial respiration, increasing reactive oxygen species formation and inducing apoptosis. Additionally, mitochondrial Ca2+ overload can damage mitochondrial recycling via mitophagy. This review will discuss the molecular players involved in mitochondrial Ca2+ dysregulation and the pharmacotherapies that target this dysregulation. As most of the current AD therapeutics are based on amyloidopathy, tauopathy, and the cholinergic hypothesis, they achieve only symptomatic relief. Thus, determining how to reestablish mitochondrial Ca2+ homeostasis may aid in the development of novel AD therapeutic interventions.
Collapse
Affiliation(s)
- Aston J Wu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Benjamin C-K Tong
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Alexis S Huang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - King-Ho Cheung
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| |
Collapse
|
214
|
Weissman D, Maack C. Redox signaling in heart failure and therapeutic implications. Free Radic Biol Med 2021; 171:345-364. [PMID: 34019933 DOI: 10.1016/j.freeradbiomed.2021.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/17/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Heart failure is a growing health burden worldwide characterized by alterations in excitation-contraction coupling, cardiac energetic deficit and oxidative stress. While current treatments are mostly limited to antagonization of neuroendocrine activation, more recent data suggest that also targeting metabolism may provide substantial prognostic benefit. However, although in a broad spectrum of preclinical models, oxidative stress plays a causal role for the development and progression of heart failure, no treatment that targets reactive oxygen species (ROS) directly has entered the clinical arena yet. In the heart, ROS derive from various sources, such as NADPH oxidases, xanthine oxidase, uncoupled nitric oxide synthase and mitochondria. While mitochondria are the primary source of ROS in the heart, communication between different ROS sources may be relevant for physiological signalling events as well as pathologically elevated ROS that deteriorate excitation-contraction coupling, induce hypertrophy and/or trigger cell death. Here, we review the sources of ROS in the heart, the modes of pathological activation of ROS formation as well as therapeutic approaches that may target ROS specifically in mitochondria.
Collapse
Affiliation(s)
- David Weissman
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany; Department of Internal Medicine 1, University Clinic Würzburg, Würzburg, Germany.
| |
Collapse
|
215
|
Kang Y, Liu J, Jiang Y, Yin S, Huang Z, Zhang Y, Wu J, Chen L, Shao L. Understanding the interactions between inorganic-based nanomaterials and biological membranes. Adv Drug Deliv Rev 2021; 175:113820. [PMID: 34087327 DOI: 10.1016/j.addr.2021.05.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/21/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
The interactions between inorganic-based nanomaterials (NMs) and biological membranes are among the most important phenomena for developing NM-based therapeutics and resolving nanotoxicology. Herein, we introduce the structural and functional effects of inorganic-based NMs on biological membranes, mainly the plasma membrane and the endomembrane system, with an emphasis on the interface, which involves highly complex networks between NMs and biomolecules (such as membrane proteins and lipids). Significant efforts have been devoted to categorizing and analyzing the interaction mechanisms in terms of the physicochemical characteristics and biological effects of NMs, which can directly or indirectly influence the effects of NMs on membranes. Importantly, we summarize that the biological membranes act as platforms and thereby mediate NMs-immune system contacts. In this overview, the existing challenges and potential applications in the areas are addressed. A strong understanding of the discussed concepts will promote therapeutic NM designs for drug delivery systems by leveraging the NMs-membrane interactions and their functions.
Collapse
Affiliation(s)
- Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanping Jiang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Suhan Yin
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhendong Huang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
216
|
Lin Y, Fu S, Yao Y, Li Y, Zhao Y, Luo L. Heart failure with preserved ejection fraction based on aging and comorbidities. J Transl Med 2021; 19:291. [PMID: 34229717 PMCID: PMC8259336 DOI: 10.1186/s12967-021-02935-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/10/2021] [Indexed: 12/25/2022] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a leading cause of hospitalizations and mortality when diagnosed at the age of ≥ 65 years. HFpEF represents multifactorial and multisystemic syndrome and has different pathophysiology and phenotypes. Its diagnosis is difficult to be established based on left ventricular ejection fraction and may benefit from individually tailored approaches, underlying age-related changes and frequent comorbidities. Compared with the rapid development in the treatment of heart failure with reduced ejection fraction, HFpEF presents a great challenge and needs to be addressed considering the failure of HF drugs to improve its outcomes. Further extensive studies on the relationships between HFpEF, aging, and comorbidities in carefully phenotyped HFpEF subgroups may help understand the biology, diagnosis, and treatment of HFpEF. The current review summarized the diagnostic and therapeutic development of HFpEF based on the complex relationships between aging, comorbidities, and HFpEF.
Collapse
Affiliation(s)
- Ying Lin
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 572013, China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 572013, China.
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| | - Yao Yao
- Centre for the Study of Ageing and Human Development and Geriatrics Division, Medical School of Duke University, Durham, NC, 27708, USA
- Centre for Healthy Ageing and Development Studies, National School of Development, Peking University, Beijing, 100871, China
| | - Yulong Li
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yali Zhao
- Central Laboratory, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 572013, China.
| | - Leiming Luo
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| |
Collapse
|
217
|
Kassab S, Albalawi Z, Daghistani H, Kitmitto A. Mitochondrial Arrest on the Microtubule Highway-A Feature of Heart Failure and Diabetic Cardiomyopathy? Front Cardiovasc Med 2021; 8:689101. [PMID: 34277734 PMCID: PMC8282893 DOI: 10.3389/fcvm.2021.689101] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/08/2021] [Indexed: 01/16/2023] Open
Abstract
A pathophysiological consequence of both type 1 and 2 diabetes is remodelling of the myocardium leading to the loss of left ventricular pump function and ultimately heart failure (HF). Abnormal cardiac bioenergetics associated with mitochondrial dysfunction occurs in the early stages of HF. Key factors influencing mitochondrial function are the shape, size and organisation of mitochondria within cardiomyocytes, with reports identifying small, fragmented mitochondria in the myocardium of diabetic patients. Cardiac mitochondria are now known to be dynamic organelles (with various functions beyond energy production); however, the mechanisms that underpin their dynamism are complex and links to motility are yet to be fully understood, particularly within the context of HF. This review will consider how the outer mitochondrial membrane protein Miro1 (Rhot1) mediates mitochondrial movement along microtubules via crosstalk with kinesin motors and explore the evidence for molecular level changes in the setting of diabetic cardiomyopathy. As HF and diabetes are recognised inflammatory conditions, with reports of enhanced activation of the NLRP3 inflammasome, we will also consider evidence linking microtubule organisation, inflammation and the association to mitochondrial motility. Diabetes is a global pandemic but with limited treatment options for diabetic cardiomyopathy, therefore we also discuss potential therapeutic approaches to target the mitochondrial-microtubule-inflammatory axis.
Collapse
Affiliation(s)
- Sarah Kassab
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Zainab Albalawi
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Hussam Daghistani
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
218
|
Ping Z, Fan H, Wen C, Ji Z, Liang S. GAPDH siRNA Regulates SH-SY5Y Cell Apoptosis Induced by Exogenous α-Synuclein Protein. Neuroscience 2021; 469:91-102. [PMID: 34216695 DOI: 10.1016/j.neuroscience.2021.06.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 11/15/2022]
Abstract
The transport mechanism of intestinal α-synuclein to the central nervous system has become a new hot topic in Parkinson's disease (PD) research. It is worth noting that the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has been reported to be involved in the pathogenesis of PD. After silencing GAPDH expression by GAPDH siRNA, the normal human intestinal epithelial crypt-like (HIEC) and human SH-SY5Y neuroblastoma cell lines were co-cultured with Escherichia coli cells which were transfected with an α-synuclein overexpression plasmid. The levels of autophagy-related proteins (BECN1, ATG5, LC3A/B and p62) were determined by Western blot analysis. Changes in pro-apoptosis protein levels and flow cytometry analysis were used to assess cell apoptosis and relative intracellular ATP concentration was measured. Oxidative stress was assessed by measuring the levels of reactive oxygen species (ROS) using 2',7'-dichlorofluorescein diacetate (DCFH-DA), thiobarbituric acid-reactive substances (TBARS), and antioxidant capacity was assessed by measuring the glutathione (GSH) levels and superoxide dismutase (SOD) activity. The silencing of the expression of GAPDH pre-knockdown was found to reduce the intracellular levels of ROS and lipid peroxidation, enhance autophagy activity, thereby reducing the cell injury, apoptosis and necrosis induced by exogenous α-synuclein protein in SH-SY5Y cells. This study identifies a new therapeutic target of exogenous α-synuclein protein induced SH-SY5Y cell injury and improves our understanding of the pathophysiological role of GAPDH in vitro.
Collapse
Affiliation(s)
- Zhang Ping
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China; Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China
| | - Hu Fan
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China; Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chai Wen
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China; Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China
| | - Zhang Ji
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China; Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China
| | - Shao Liang
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China; Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, China; The people's Hospital of Yu Du County, Jiangxi, China.
| |
Collapse
|
219
|
Morciano G, Naumova N, Koprowski P, Valente S, Sardão VA, Potes Y, Rimessi A, Wieckowski MR, Oliveira PJ. The mitochondrial permeability transition pore: an evolving concept critical for cell life and death. Biol Rev Camb Philos Soc 2021; 96:2489-2521. [PMID: 34155777 DOI: 10.1111/brv.12764] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
In this review, we summarize current knowledge of perhaps one of the most intriguing phenomena in cell biology: the mitochondrial permeability transition pore (mPTP). This phenomenon, which was initially observed as a sudden loss of inner mitochondrial membrane impermeability caused by excessive calcium, has been studied for almost 50 years, and still no definitive answer has been provided regarding its mechanisms. From its initial consideration as an in vitro artifact to the current notion that the mPTP is a phenomenon with physiological and pathological implications, a long road has been travelled. We here summarize the role of mitochondria in cytosolic calcium control and the evolving concepts regarding the mitochondrial permeability transition (mPT) and the mPTP. We show how the evolving mPTP models and mechanisms, which involve many proposed mitochondrial protein components, have arisen from methodological advances and more complex biological models. We describe how scientific progress and methodological advances have allowed milestone discoveries on mPTP regulation and composition and its recognition as a valid target for drug development and a critical component of mitochondrial biology.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera 1, Cotignola, Ravenna, 48033, Italy.,Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 70, Ferrara, 44121, Italy
| | - Natalia Naumova
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua Medical School, Via Giustiniani 2, Padova, 35128, Italy
| | - Piotr Koprowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Sara Valente
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| | - Yaiza Potes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Alessandro Rimessi
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 70, Ferrara, 44121, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| |
Collapse
|
220
|
Lin M, Hu X, Chang S, Chang Y, Bian W, Hu R, Wang J, Zhu Q, Qiu J. Advances of Antisense Oligonucleotide Technology in the Treatment of Hereditary Neurodegenerative Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6678422. [PMID: 34211575 PMCID: PMC8211492 DOI: 10.1155/2021/6678422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/13/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022]
Abstract
Antisense nucleic acids are single-stranded oligonucleotides that have been specially chemically modified, which can bind to RNA expressed by target genes through base complementary pairing and affect protein synthesis at the level of posttranscriptional processing or protein translation. In recent years, the application of antisense nucleic acid technology in the treatment of neuromuscular diseases has made remarkable progress. In 2016, the US FDA approved two antisense nucleic acid drugs for the treatment of Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA), and the development to treat other neurodegenerative diseases has also entered the clinical stage. Therefore, ASO represents a treatment with great potential. The article will summarize ASO therapies in terms of mechanism of action, chemical modification, and administration methods and analyze their role in several common neurodegenerative diseases, such as SMA, DMD, and amyotrophic lateral sclerosis (ALS). This article systematically summarizes the great potential of antisense nucleic acid technology in the treatment of hereditary neurodegenerative diseases.
Collapse
Affiliation(s)
- Mengsi Lin
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xinyi Hu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Shiyi Chang
- School of Medicine, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Chang
- School of Life Sciences, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Wenjun Bian
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ruikun Hu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jing Wang
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Qingwen Zhu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jiaying Qiu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| |
Collapse
|
221
|
Williams JL, Hall CL, Meimaridou E, Metherell LA. Loss of Nnt Increases Expression of Oxidative Phosphorylation Complexes in C57BL/6J Hearts. Int J Mol Sci 2021; 22:6101. [PMID: 34198873 PMCID: PMC8201288 DOI: 10.3390/ijms22116101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/26/2022] Open
Abstract
Nicotinamide nucleotide transhydrogenase (NNT) is a proton pump in the inner mitochondrial membrane that generates reducing equivalents in the form of NAPDH, which can be used for anabolic pathways or to remove reactive oxygen species (ROS). A number of studies have linked NNT dysfunction to cardiomyopathies and increased risk of atherosclerosis; however, biallelic mutations in humans commonly cause a phenotype of adrenal insufficiency, with rare occurrences of cardiac dysfunction and testicular tumours. Here, we compare the transcriptomes of the hearts, adrenals and testes from three mouse models: the C57BL/6N, which expresses NNT; the C57BL/6J, which lacks NNT; and a third mouse, expressing the wild-type NNT sequence on the C57BL/6J background. We saw enrichment of oxidative phosphorylation genes in the C57BL/B6J in the heart and adrenal, possibly indicative of an evolved response in this substrain to loss of Nnt. However, differential gene expression was mainly driven by mouse background with some changes seen in all three tissues, perhaps reflecting underlying genetic differences between the C57BL/B6J and -6N substrains.
Collapse
Affiliation(s)
- Jack L. Williams
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.L.W.); (C.L.H.)
| | - Charlotte L. Hall
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.L.W.); (C.L.H.)
| | - Eirini Meimaridou
- School of Human Sciences, London Metropolitan University, London N7 8DB, UK;
| | - Lou A. Metherell
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.L.W.); (C.L.H.)
| |
Collapse
|
222
|
Cantoni O, Zito E, Fiorani M, Guidarelli A. Arsenite impinges on endoplasmic reticulum-mitochondria crosstalk to elicit mitochondrial ROS formation and downstream toxicity. Semin Cancer Biol 2021; 76:132-138. [PMID: 34089843 DOI: 10.1016/j.semcancer.2021.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023]
Abstract
Arsenite is an important carcinogen and toxic compound, causing various deleterious effects through multiple mechanisms. In this review, we focused on mitochondrial ROS (mitoROS) and discussed on the mechanisms mediating their formation. The metalloid promotes direct effects in mitochondria, resulting in superoxide formation only under conditions of increased mitochondrial Ca2+ concentration ([Ca2+]m). In this perspective, the time of exposure and concentration requirements for arsenite were largely conditioned by other effects of the metalloid in specific sites of the endoplasmic reticulum (ER). Arsenite induced a slow and limited mobilization of Ca2+ from IP3R via a saturable mechanism, failing to increase the [Ca2+]m. This effect was however associated with the triggering of an intraluminal crosstalk between the IP3R and the ryanodine receptor (RyR), causing a large and concentration dependent release of Ca2+ from RyR and a parallel increase in [Ca2+]m. Thus, the Ca2+-dependent mitoO2-. formation appears to be conditioned by the spatial/functional organization of the ER/mitochondria network and RyR expression. We also speculate on the possibility that the ER stress response might regulate the above effects on the intraluminal crosstalk between the IP3R and the RyR via oxidation of critical thiols mediated by the H2O2 locally released by oxidoreductin 1α.
Collapse
Affiliation(s)
- Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
223
|
Implications of SGLT Inhibition on Redox Signalling in Atrial Fibrillation. Int J Mol Sci 2021; 22:ijms22115937. [PMID: 34073033 PMCID: PMC8198069 DOI: 10.3390/ijms22115937] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained (atrial) arrhythmia, a considerable global health burden and often associated with heart failure. Perturbations of redox signalling in cardiomyocytes provide a cellular substrate for the manifestation and maintenance of atrial arrhythmias. Several clinical trials have shown that treatment with sodium-glucose linked transporter inhibitors (SGLTi) improves mortality and hospitalisation in heart failure patients independent of the presence of diabetes. Post hoc analysis of the DECLARE-TIMI 58 trial showed a 19% reduction in AF in patients with diabetes mellitus (hazard ratio, 0.81 (95% confidence interval: 0.68-0.95), n = 17.160) upon treatment with SGLTi, regardless of pre-existing AF or heart failure and independent from blood pressure or renal function. Accordingly, ongoing experimental work suggests that SGLTi not only positively impact heart failure but also counteract cellular ROS production in cardiomyocytes, thereby potentially altering atrial remodelling and reducing AF burden. In this article, we review recent studies investigating the effect of SGLTi on cellular processes closely interlinked with redox balance and their potential effects on the onset and progression of AF. Despite promising insight into SGLTi effect on Ca2+ cycling, Na+ balance, inflammatory and fibrotic signalling, mitochondrial function and energy balance and their potential effect on AF, the data are not yet conclusive and the importance of individual pathways for human AF remains to be established. Lastly, an overview of clinical studies investigating SGLTi in the context of AF is provided.
Collapse
|
224
|
Generation of somatic mitochondrial DNA-replaced cells for mitochondrial dysfunction treatment. Sci Rep 2021; 11:10897. [PMID: 34035362 PMCID: PMC8149667 DOI: 10.1038/s41598-021-90316-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial diseases currently have no cure regardless of whether the cause is a nuclear or mitochondrial genome mutation. Mitochondrial dysfunction notably affects a wide range of disorders in aged individuals, including neurodegenerative diseases, cancers, and even senescence. Here, we present a procedure to generate mitochondrial DNA-replaced somatic cells with a combination of a temporal reduction in endogenous mitochondrial DNA and coincubation with exogeneous isolated mitochondria. Heteroplasmy in mitochondrial disease patient-derived fibroblasts in which the mutant genotype was dominant over the wild-type genotype was reversed. Mitochondrial disease patient-derived fibroblasts regained respiratory function and showed lifespan extension. Mitochondrial membranous components were utilized as a vehicle to deliver the genetic materials into endogenous mitochondria-like horizontal genetic transfer in prokaryotes. Mitochondrial DNA-replaced cells could be a resource for transplantation to treat maternal inherited mitochondrial diseases.
Collapse
|
225
|
Survival upon Staphylococcus aureus mediated wound infection in Caenorhabditis elegans and the mechanism entailed. Microb Pathog 2021; 157:104952. [PMID: 34022354 DOI: 10.1016/j.micpath.2021.104952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/31/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
Infection following injury is one of the major threats which causes huge economic burden in wound care management all over the world. Injury often results with poor healing when coupled by following infection. In contrast to this, we observed enhanced survival of wound infected worms compared to wounded worms in Caenorhabditis elegans wound model while infecting with Staphylococcus aureus. Hence, the study was intended to identify the mechanism for the enhanced survival of wound infected worms through LCMS/MS based high throughput proteomic analysis. Bioinformatics analyses of the identified protein players indicated differential enrichment of several pathways including MAPK signaling, oxidative phosphorylation and phosphatidylinositol signaling. Inhibition of oxidative phosphorylation and phosphatidylinositol signaling through chemical treatment showed complete reversal of the enhanced survival during wound infection nevertheless mutant of MAPK pathway did not reverse the same. Consequently, it was delineated that oxidative phosphorylation and phosphatidylinositol signaling are crucial for the survival. In this regard, elevated calcium signals and ROS including O- and H2O2 were observed in wounded and wound infected worms. Consequently, it was insinuated that presence of pathogen stress could have incited survival in wound infected worms with the aid of elevated ROS and calcium signals.
Collapse
|
226
|
Emmerzaal TL, Nijkamp G, Veldic M, Rahman S, Andreazza AC, Morava E, Rodenburg RJ, Kozicz T. Effect of neuropsychiatric medications on mitochondrial function: For better or for worse. Neurosci Biobehav Rev 2021; 127:555-571. [PMID: 34000348 DOI: 10.1016/j.neubiorev.2021.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/12/2021] [Accepted: 05/04/2021] [Indexed: 01/22/2023]
Abstract
Individuals with mitochondrial disease often present with psychopathological comorbidity, and mitochondrial dysfunction has been proposed as the underlying pathobiology in various psychiatric disorders. Several studies have suggested that medications used to treat neuropsychiatric disorders could directly influence mitochondrial function. This review provides a comprehensive overview of the effect of these medications on mitochondrial function. We collected preclinical information on six major groups of antidepressants and other neuropsychiatric medications and found that the majority of these medications either positively influenced mitochondrial function or showed mixed effects. Only amitriptyline, escitalopram, and haloperidol were identified as having exclusively adverse effects on mitochondrial function. In the absence of formal clinical trials, and until such trials are completed, the data from preclinical studies reported and discussed here could inform medication prescribing practices for individuals with psychopathology and impaired mitochondrial function in the underlying pathology.
Collapse
Affiliation(s)
- Tim L Emmerzaal
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Imaging, Anatomy, Nijmegen, The Netherlands; Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA
| | - Gerben Nijkamp
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Imaging, Anatomy, Nijmegen, The Netherlands
| | - Marin Veldic
- Mayo Clinic, Department of Psychiatry, Rochester, MN, USA
| | - Shamima Rahman
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Ana Cristina Andreazza
- University of Toronto, Temerty Faculty of Medicine, Department of Pharmacology & Toxicology and Psychiatry, Toronto, Canada
| | - Eva Morava
- Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA; Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Richard J Rodenburg
- Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Department of Pediatrics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tamas Kozicz
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Imaging, Anatomy, Nijmegen, The Netherlands; Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA; Mayo Clinic, Department of Biochemistry and Molecular Biology, Rochester, MN, USA.
| |
Collapse
|
227
|
Tan X, Huang J, Wang Y, He S, Jia L, Zhu Y, Pu K, Zhang Y, Yang X. Transformable Nanosensitizer with Tumor Microenvironment-Activated Sonodynamic Process and Calcium Release for Enhanced Cancer Immunotherapy. Angew Chem Int Ed Engl 2021; 60:14051-14059. [PMID: 33797161 DOI: 10.1002/anie.202102703] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/31/2021] [Indexed: 12/15/2022]
Abstract
Despite the promise of sonodynamic processes in cancer therapy, existing sonosensitizers often fail to regulate the generation of reactive oxygen species (ROS) against tumors, potentially leading to off-target toxicity to normal tissues. We report a transformable core-shell nanosonosensitizer (TiO2 @CaP) that reinvigorates ROS generation and dissolves its CaP shell to release Ca2+ in an acidic tumor microenvironment (TME) under ultrasound activation. Thus, TiO2 @CaP acts as a smart nanosonosensitizer that specifically induces mitochondrial dysfunction via overloading intracellular Ca2+ ions to synergize with the sonodynamic process in the TME. TiO2 @CaP substantially enhances immunogenic cell death, resulting in enhanced T-cell recruitment and infiltration into the immunogenic cold tumor (4T1). In conjunction with checkpoint blockade therapy (anti-PD 1), TiO2 @CaP-mediated sonodynamic therapy elicits systemic antitumor immunity, leading to regression of non-treated distant tumors and inhibition of lung metastasis. This work paves the way to development of "smart" TME-activatable sonosensitizers with temporospatial control over antitumor responses.
Collapse
Affiliation(s)
- Xuan Tan
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| | - Jingzhao Huang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| | - Yiqian Wang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| | - Shasha He
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Le Jia
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| | - Yanhong Zhu
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| | - Xiangliang Yang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430074, P. R. China
| |
Collapse
|
228
|
Tan X, Huang J, Wang Y, He S, Jia L, Zhu Y, Pu K, Zhang Y, Yang X. Transformable Nanosensitizer with Tumor Microenvironment‐Activated Sonodynamic Process and Calcium Release for Enhanced Cancer Immunotherapy. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102703] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Xuan Tan
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| | - Jingzhao Huang
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| | - Yiqian Wang
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| | - Shasha He
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Le Jia
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| | - Yanhong Zhu
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| | - Xiangliang Yang
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology 1037 Luoyu Road Wuhan 430074 P. R. China
| |
Collapse
|
229
|
Modulations of Cardiac Functions and Pathogenesis by Reactive Oxygen Species and Natural Antioxidants. Antioxidants (Basel) 2021; 10:antiox10050760. [PMID: 34064823 PMCID: PMC8150787 DOI: 10.3390/antiox10050760] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 01/11/2023] Open
Abstract
Homeostasis in the level of reactive oxygen species (ROS) in cardiac myocytes plays a critical role in regulating their physiological functions. Disturbance of balance between generation and removal of ROS is a major cause of cardiac myocyte remodeling, dysfunction, and failure. Cardiac myocytes possess several ROS-producing pathways, such as mitochondrial electron transport chain, NADPH oxidases, and nitric oxide synthases, and have endogenous antioxidation mechanisms. Cardiac Ca2+-signaling toolkit proteins, as well as mitochondrial functions, are largely modulated by ROS under physiological and pathological conditions, thereby producing alterations in contraction, membrane conductivity, cell metabolism and cell growth and death. Mechanical stresses under hypertension, post-myocardial infarction, heart failure, and valve diseases are the main causes for stress-induced cardiac remodeling and functional failure, which are associated with ROS-induced pathogenesis. Experimental evidence demonstrates that many cardioprotective natural antioxidants, enriched in foods or herbs, exert beneficial effects on cardiac functions (Ca2+ signal, contractility and rhythm), myocytes remodeling, inflammation and death in pathological hearts. The review may provide knowledge and insight into the modulation of cardiac pathogenesis by ROS and natural antioxidants.
Collapse
|
230
|
Lai HL, Fan XX, Li RZ, Wang YW, Zhang J, Liu L, Neher E, Yao XJ, Leung ELH. Roles of Ion Fluxes, Metabolism, and Redox Balance in Cancer Therapy. Antioxid Redox Signal 2021; 34:1108-1127. [PMID: 33115253 DOI: 10.1089/ars.2020.8125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent Advances: The 2019 Nobel Prize awarded to the mechanisms for oxygen sensing and adaptation according to oxygen availability, highlighting the fundamental importance of gaseous molecules. Gaseous molecules, including reactive oxygen species (ROS), can interact with different cations generated during metabolic and redox dysregulation in cancer cells. Cross talk between calcium signaling and metabolic/redox pathways leads to network-based dyregulation in cancer. Significance: Recent discovery on using small molecules targeting the ion channels, redox signaling, and protein modification on metabolic enzymes can effectively inhibit cancer growth. Several FDA-approved drugs and clinical trials are ongoing to target the calcium channels, such as TRPV6 and TRPM8. Multiple small molecules from natural products target metablic and redox enzymes to exert an anticancer effect. Critical Issues: Small molecules targeting key ion channels, metabolic enzymes that control key aspects of metabolism, and redox proteins are promising, but their action mechanisms of the target are needed to be elucidated with advanced-omic technologies, which can give network-based and highly dimensioal data. In addition, small molecules that can directly modify the protein residues have emerged as a novel anticancer strategy. Future Directions: Advanced technology accelerates the detection of ions and metabolic and redox changes in clinical samples for diagnosis and informs the decision of cancer treatment. The improvement of ROS detection, ROS target identification, and computational-aid drug discovery also improves clincal outcome.Overall, network-based or holistic regulations of cancer via ion therapy and metabolic and redox intervention are promising as new anticancer strategies. Antioxid. Redox Signal. 34, 1108-1127.
Collapse
Affiliation(s)
- Huan-Ling Lai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Run-Ze Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Yu-Wei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Junmin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.,School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Erwin Neher
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.,Membrane Biophysics Emeritus Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| |
Collapse
|
231
|
Li Z, Zhao H, Wang J. Metabolism and Chronic Inflammation: The Links Between Chronic Heart Failure and Comorbidities. Front Cardiovasc Med 2021; 8:650278. [PMID: 34026868 PMCID: PMC8131678 DOI: 10.3389/fcvm.2021.650278] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) patients often suffer from multiple comorbidities, such as diabetes, atrial fibrillation, depression, chronic obstructive pulmonary disease, and chronic kidney disease. The coexistance of comorbidities usually leads to multi morbidity and poor prognosis. Treatments for HF patients with multi morbidity are still an unmet clinical need, and finding an effective therapy strategy is of great value. HF can lead to comorbidity, and in return, comorbidity may promote the progression of HF, creating a vicious cycle. This reciprocal correlation indicates there may be some common causes and biological mechanisms. Metabolism remodeling and chronic inflammation play a vital role in the pathophysiological processes of HF and comorbidities, indicating metabolism and inflammation may be the links between HF and comorbidities. In this review, we comprehensively discuss the major underlying mechanisms and therapeutic implications for comorbidities of HF. We first summarize the potential role of metabolism and inflammation in HF. Then, we give an overview of the linkage between common comorbidities and HF, from the perspective of epidemiological evidence to the underlying metabolism and inflammation mechanisms. Moreover, with the help of bioinformatics, we summarize the shared risk factors, signal pathways, and therapeutic targets between HF and comorbidities. Metabolic syndrome, aging, deleterious lifestyles (sedentary behavior, poor dietary patterns, smoking, etc.), and other risk factors common to HF and comorbidities are all associated with common mechanisms. Impaired mitochondrial biogenesis, autophagy, insulin resistance, and oxidative stress, are among the major mechanisms of both HF and comorbidities. Gene enrichment analysis showed the PI3K/AKT pathway may probably play a central role in multi morbidity. Additionally, drug targets common to HF and several common comorbidities were found by network analysis. Such analysis has already been instrumental in drug repurposing to treat HF and comorbidity. And the result suggests sodium-glucose transporter-2 (SGLT-2) inhibitors, IL-1β inhibitors, and metformin may be promising drugs for repurposing to treat multi morbidity. We propose that targeting the metabolic and inflammatory pathways that are common to HF and comorbidities may provide a promising therapeutic strategy.
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Hongmei Zhao
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
232
|
Ding R, Yin YL, Jiang LH. Reactive Oxygen Species-Induced TRPM2-Mediated Ca 2+ Signalling in Endothelial Cells. Antioxidants (Basel) 2021; 10:antiox10050718. [PMID: 34063677 PMCID: PMC8147627 DOI: 10.3390/antiox10050718] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells form the innermost layer of blood vessels with a fundamental role as the physical barrier. While regulation of endothelial cell function by reactive oxygen species (ROS) is critical in physiological processes such as angiogenesis, endothelial function is a major target for interruption by oxidative stress resulting from generation of high levels of ROS in endothelial cells by various pathological factors and also release of ROS by neutrophils. TRPM2 is a ROS-sensitive Ca2+-permeable channel expressed in endothelial cells of various vascular beds. In this review, we provide an overview of the TRPM2 channel and its role in mediating ROS-induced Ca2+ signaling in endothelial cells. We discuss the TRPM2-mediated Ca2+ signaling in vascular endothelial growth factor-induced angiogenesis and in post-ischemic neovascularization. In particular, we examine the accumulative evidence that supports the role of TRPM2-mediated Ca2+ signaling in endothelial cell dysfunction caused by various oxidative stress-inducing factors that are associated with tissue inflammation, obesity and diabetes, as well as air pollution. These findings provide new, mechanistic insights into ROS-mediated regulation of endothelial cells in physiology and diseases.
Collapse
Affiliation(s)
- Ran Ding
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang 453003, China; (R.D.); (Y.-L.Y.)
| | - Ya-Ling Yin
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang 453003, China; (R.D.); (Y.-L.Y.)
| | - Lin-Hua Jiang
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang 453003, China; (R.D.); (Y.-L.Y.)
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: ; Tel.: +44-113-3434-231
| |
Collapse
|
233
|
Bou-Teen D, Kaludercic N, Weissman D, Turan B, Maack C, Di Lisa F, Ruiz-Meana M. Mitochondrial ROS and mitochondria-targeted antioxidants in the aged heart. Free Radic Biol Med 2021; 167:109-124. [PMID: 33716106 DOI: 10.1016/j.freeradbiomed.2021.02.043] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/14/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Excessive mitochondrial ROS production has been causally linked to the pathophysiology of aging in the heart and other organs, and plays a deleterious role in several age-related cardiac pathologies, including myocardial ischemia-reperfusion injury and heart failure, the two worldwide leading causes of death and disability in the elderly. However, ROS generation is also a fundamental mitochondrial function that orchestrates several signaling pathways, some of them exerting cardioprotective effects. In cardiac myocytes, mitochondria are particularly abundant and are specialized in subcellular populations, in part determined by their relationships with other organelles and their cyclic calcium handling activity necessary for adequate myocardial contraction/relaxation and redox balance. Depending on their subcellular location, mitochondria can themselves be differentially targeted by ROS and display distinct age-dependent functional decline. Thus, precise mitochondria-targeted therapies aimed at counteracting unregulated ROS production are expected to have therapeutic benefits in certain aging-related heart conditions. However, for an adequate design of such therapies, it is necessary to unravel the complex and dynamic interactions between mitochondria and other cellular processes.
Collapse
Affiliation(s)
- Diana Bou-Teen
- Hospital Universitari Vall d'Hebron, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR),Universitat Autonoma de Barcelona, 08035, Barcelona, Spain
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), via Ugo Bassi 58/B, 35131, Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35129, Padova, Italy
| | - David Weissman
- Comprehensive Heart Failure Center, University Clinic Würzburg, 97080, Würzburg, Germany
| | - Belma Turan
- Departments of Biophysics, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, 97080, Würzburg, Germany
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), via Ugo Bassi 58/B, 35131, Padova, Italy; Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Marisol Ruiz-Meana
- Hospital Universitari Vall d'Hebron, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR),Universitat Autonoma de Barcelona, 08035, Barcelona, Spain; Centro de Investigación Biomédica en Red-CV, CIBER-CV, Spain.
| |
Collapse
|
234
|
Geisberger S, Bartolomaeus H, Neubert P, Willebrand R, Zasada C, Bartolomaeus T, McParland V, Swinnen D, Geuzens A, Maifeld A, Krampert L, Vogl M, Mähler A, Wilck N, Markó L, Tilic E, Forslund SK, Binger KJ, Stegbauer J, Dechend R, Kleinewietfeld M, Jantsch J, Kempa S, Müller DN. Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes. Circulation 2021; 144:144-158. [PMID: 33906377 PMCID: PMC8270232 DOI: 10.1161/circulationaha.120.052788] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Dietary high salt (HS) is a leading risk factor for mortality and morbidity. Serum sodium transiently increases postprandially but can also accumulate at sites of inflammation affecting differentiation and function of innate and adaptive immune cells. Here, we focus on how changes in extracellular sodium, mimicking alterations in the circulation and tissues, affect the early metabolic, transcriptional, and functional adaption of human and murine mononuclear phagocytes. Methods: Using Seahorse technology, pulsed stable isotope-resolved metabolomics, and enzyme activity assays, we characterize the central carbon metabolism and mitochondrial function of human and murine mononuclear phagocytes under HS in vitro. HS as well as pharmacological uncoupling of the electron transport chain under normal salt is used to analyze mitochondrial function on immune cell activation and function (as determined by Escherichiacoli killing and CD4+ T cell migration capacity). In 2 independent clinical studies, we analyze the effect of a HS diet during 2 weeks (URL: http://www.clinicaltrials.gov. Unique identifier: NCT02509962) and short-term salt challenge by a single meal (URL: http://www.clinicaltrials.gov. Unique identifier: NCT04175249) on mitochondrial function of human monocytes in vivo. Results: Extracellular sodium was taken up into the intracellular compartment, followed by the inhibition of mitochondrial respiration in murine and human macrophages. Mechanistically, HS reduces mitochondrial membrane potential, electron transport chain complex II activity, oxygen consumption, and ATP production independently of the polarization status of macrophages. Subsequently, cell activation is altered with improved bactericidal function in HS-treated M1-like macrophages and diminished CD4+ T cell migration in HS-treated M2-like macrophages. Pharmacological uncoupling of the electron transport chain under normal salt phenocopies HS-induced transcriptional changes and bactericidal function of human and murine mononuclear phagocytes. Clinically, also in vivo, rise in plasma sodium concentration within the physiological range reversibly reduces mitochondrial function in human monocytes. In both a 14-day and single meal HS challenge, healthy volunteers displayed a plasma sodium increase of and respectively, that correlated with decreased monocytic mitochondrial oxygen consumption. Conclusions: Our data identify the disturbance of mitochondrial respiration as the initial step by which HS mechanistically influences immune cell function. Although these functional changes might help to resolve bacterial infections, a shift toward proinflammation could accelerate inflammatory cardiovascular disease.
Collapse
Affiliation(s)
- Sabrina Geisberger
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Germany (S.G., C.Z., S.K.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Patrick Neubert
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Germany (P.N., L.K., M.V., J.J.)
| | - Ralf Willebrand
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, UHasselt, Campus Diepenbeek, Belgium (R.W., D.S., A.G., M.K.)
| | - Christin Zasada
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Germany (S.G., C.Z., S.K.)
| | | | - Victoria McParland
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Dries Swinnen
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, UHasselt, Campus Diepenbeek, Belgium (R.W., D.S., A.G., M.K.)
| | - Anneleen Geuzens
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, UHasselt, Campus Diepenbeek, Belgium (R.W., D.S., A.G., M.K.)
| | - András Maifeld
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Luka Krampert
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Germany (P.N., L.K., M.V., J.J.)
| | - Marion Vogl
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Germany (P.N., L.K., M.V., J.J.)
| | - Anja Mähler
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Nicola Wilck
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (N.W.).,Department of Nephrology and Internal Intensive Care Medicine (N.W.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Lajos Markó
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Ekin Tilic
- Institute of Evolutionary Biology, University of Bonn, Germany (T.B., E.T.)
| | - Sofia K Forslund
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| | - Katrina J Binger
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia (K.J.B.)
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany (J.S.)
| | - Ralf Dechend
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Department of Cardiology and Nephrology, HELIOS-Klinikum, Berlin, Germany (R.D.)
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, UHasselt, Campus Diepenbeek, Belgium (R.W., D.S., A.G., M.K.)
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Germany (P.N., L.K., M.V., J.J.)
| | - Stefan Kempa
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Germany (S.G., C.Z., S.K.)
| | - Dominik N Müller
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,German Center for Cardiovascular Research, partner site Berlin (S.G., H.B., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (H.B., V.M., A. Maifeld, A. Mähler, L.M., S.K.F., R.D., D.N.M.).,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.G., H.B., V.M., A. Maifeld, A. Mähler, N.W., L.M., S.K.F., R.D., D.N.M.)
| |
Collapse
|
235
|
Xue XJ, Li FR, Yu J. Mitochondrial pathway of the lysine demethylase 5C inhibitor CPI-455 in the Eca-109 esophageal squamous cell carcinoma cell line. World J Gastroenterol 2021; 27:1805-1815. [PMID: 33967558 PMCID: PMC8072195 DOI: 10.3748/wjg.v27.i16.1805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/14/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Esophageal cancer is a malignant tumor of the digestive tract that is difficult to diagnose early. CPI-455 has been reported to inhibit various cancers, but its role in esophageal squamous cell carcinoma (ESCC) is unknown.
AIM To investigate the effects and mechanism of the lysine demethylase 5C inhibitor, CPI-455, on ESCC cells.
METHODS A methyl tetrazolium assay was used to detect the inhibitory effect of CPI-455 on the proliferation of Eca-109 cells. Apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential were assessed by flow cytometry. Laser confocal scanning and transmission electron microscopy were used to observe changes in Eca-109 cell morphology. The protein expression of P53, Bax, lysine-specific demethylase 5C (KDM5C), cleaved Caspase-9, and cleaved Caspase-3 were assayed by western blotting.
RESULTS Compared with the control group, CPI-455 significantly inhibited Eca-109 cell proliferation. Gemcitabine inhibited Eca-109 cell proliferation in a concentration- and time-dependent manner. CPI-455 caused extensive alteration of the mitochondria, which appeared to have become atrophied. The cell membrane was weakly stained and the cytoplasmic structures were indistinct and disorganized, with serious cavitation when viewed by transmission electron microscopy. The flow cytometry and western blot results showed that, compared with the control group, the mitochondrial membrane potential was decreased and depolarized in Eca-109 cells treated with CPI-455. CPI-455 significantly upregulated the ROS content, P53, Bax, Caspase-9, and Caspase-3 protein expression in Eca-109 cells, whereas KDM5C expression was downregulated.
CONCLUSION CPI-455 inhibited Eca-109 cell proliferation via mitochondrial apoptosis by regulating the expression of related genes.
Collapse
Affiliation(s)
- Xiao-Jie Xue
- Department of Clinical Laboratory, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi 435000, Hubei Province, China
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi 435000, Hubei Province, China
- Medical College, Wuhan University of Science and Technology, Wuhan 430081, Hubei Province, China
| | - Fei-Rong Li
- Department of Clinical Laboratory, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi 435000, Hubei Province, China
| | - Jing Yu
- Department of Laboratory Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, Hubei Province, China
| |
Collapse
|
236
|
Hamilton S, Terentyeva R, Clements RT, Belevych AE, Terentyev D. Sarcoplasmic reticulum-mitochondria communication; implications for cardiac arrhythmia. J Mol Cell Cardiol 2021; 156:105-113. [PMID: 33857485 DOI: 10.1016/j.yjmcc.2021.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/15/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022]
Abstract
Sudden cardiac death due to ventricular tachyarrhythmias remains the major cause of mortality in the world. Heart failure, diabetic cardiomyopathy, old age-related cardiac dysfunction and inherited disorders are associated with enhanced propensity to malignant cardiac arrhythmias. Both defective mitochondrial function and abnormal intracellular Ca2+ homeostasis have been established as the key contributing factors in the pathophysiology and arrhythmogenesis in these conditions. This article reviews current advances in understanding of bidirectional control of ryanodine receptor-mediated sarcoplasmic reticulum Ca2+ release and mitochondrial function, and how defects in crosstalk between these two organelles increase arrhythmic risk in cardiac disease.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America
| | - Richard T Clements
- Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, United States of America
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America.
| |
Collapse
|
237
|
Affiliation(s)
- Joanne F. Garbincius
- Center for Translational Medicine, Lewis Katz School of
Medicine at Temple University, Philadelphia, PA 19140
| | - John W. Elrod
- Center for Translational Medicine, Lewis Katz School of
Medicine at Temple University, Philadelphia, PA 19140
| |
Collapse
|
238
|
Ma G, Yang Y, Chen Y, Wei X, Ding J, Zhou RP, Hu W. Blockade of TRPM7 Alleviates Chondrocyte Apoptosis and Articular Cartilage Damage in the Adjuvant Arthritis Rat Model Through Regulation of the Indian Hedgehog Signaling Pathway. Front Pharmacol 2021; 12:655551. [PMID: 33927631 PMCID: PMC8076952 DOI: 10.3389/fphar.2021.655551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
Articular cartilage damage with subsequent impairment of joint function is a common feature of articular diseases, in particular, rheumatoid arthritis and osteoarthritis. While articular cartilage injury mediated by chondrocyte apoptosis is a known major pathological feature of arthritis, the specific mechanisms remain unclear at present. Transient receptor potential melastatin-like seven channel (TRPM7) is reported to play an important regulatory role in apoptosis. This study focused on the effects of TRPM7 on arthritic chondrocyte injury and its underlying mechanisms of action. Sodium nitroprusside (SNP)-induced rat primary chondrocyte apoptosis and rat adjuvant arthritis (AA) were used as in vitro and in vivo models, respectively. Blockage of TRPM7 with 2-APB or specific siRNA resulted in increased chondrocyte viability and reduced toxicity of SNP. Moreover, treatment with 2-APB enhanced the Bcl-2/Bax ratio and reduced cleaved PARP and IL-6, MMP-13 and ADAMTS-5 expression in SNP-treated chondrocytes. Activation of Indian Hedgehog with purmorphamine reversed the protective effects of 2-APB on SNP-induced chondrocyte apoptosis. Blockage of TRPM7 with 2-APB relieved the clinical signs of AA in the rat model and reduced the arthritis score and paw swelling. Similar to findings in SNP-treated chondrocytes, 2-APB treatment increased the Bcl-2/Bax ratio and suppressed cleaved PARP, IL-6, MMP-13, ADAMTS-5, TRPM7, and Indian hedgehog expression in articular cartilage of AA rats. Our collective findings suggest that blockade of TRPM7 could effectively reduce chondrocyte apoptosis and articular cartilage damage in rats with adjuvant arthritis through regulation of the Indian Hedgehog signaling pathway.
Collapse
Affiliation(s)
- Ganggang Ma
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yang Yang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Xin Wei
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
| |
Collapse
|
239
|
Zhao H, Pan X. Mitochondrial Ca 2+ and cell cycle regulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:171-207. [PMID: 34253295 DOI: 10.1016/bs.ircmb.2021.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It has been demonstrated for more than 40 years that intracellular calcium (Ca2+) controls a variety of cellular functions, including mitochondrial metabolism and cell proliferation. Cytosolic Ca2+ fluctuation during key stages of the cell cycle can lead to mitochondrial Ca2+ uptake and subsequent activation of mitochondrial oxidative phosphorylation and a range of signaling. However, the relationship between mitochondrial Ca2+ and cell cycle progression has long been neglected because the molecule responsible for Ca2+ uptake has been unknown. Recently, the identification of the mitochondrial Ca2+ uniporter (MCU) has led to key advances. With improved Ca2+ imaging and detection, effects of MCU-mediated mitochondrial Ca2+ have been observed at different stages of the cell cycle. Elevated Ca2+ signaling boosts ATP and ROS production, remodels cytosolic Ca2+ pathways and reprograms cell fate-determining networks. These findings suggest that manipulating mitochondrial Ca2+ signaling may serve as a potential strategy in the control of many crucial biological events, such as tumor development and cell division in hematopoietic stem cells (HSCs). In this review, we summarize the current understanding of the role of mitochondrial Ca2+ signaling during different stages of the cell cycle and highlight the potential physiological and pathological significance of mitochondrial Ca2+ signaling.
Collapse
Affiliation(s)
- Haixin Zhao
- State Key Laboratory of Experimental Haematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xin Pan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.
| |
Collapse
|
240
|
Xiang L, Shao Y, Chen Y. Mitochondrial dysfunction and mitochondrion-targeted therapeutics in liver diseases. J Drug Target 2021; 29:1080-1093. [PMID: 33788656 DOI: 10.1080/1061186x.2021.1909051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is a vital metabolic and detoxifying organ and suffers diverse endogenous or exogenous damage. Hepatocyte mitochondria experience various structural and functional defects from liver injury, bearing oxidative stress, metabolic dysregulation, and the disturbance of mitochondrial quality control (MQC) mechanisms. Mitochondrial malfunction initiates the mitochondria-mediated apoptotic pathways and the release of damage signals, aggravating liver damage and disease progression via inflammation and reparative fibrogenesis. Removal of mitochondrial impairment or the improvement of MQC mechanisms restore mitochondrial homeostasis and benefit liver health. This review discusses the association of mitochondrial disorders with hepatic pathophysiological processes and the resultant potential of mitochondrion-targeting therapeutics for hepatic disorders. The recent advances in the MQC mechanisms and the mitochondrial-derived damage-associated molecular patterns (DAMPs) in the pathology and treatment of liver disease are particularly focussed.
Collapse
Affiliation(s)
- Li Xiang
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, China
| | - Yaru Shao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China.,Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, China
| | - Yuping Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China.,Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, China
| |
Collapse
|
241
|
Lodde V, Morandini P, Costa A, Murgia I, Ezquer I. cROStalk for Life: Uncovering ROS Signaling in Plants and Animal Systems, from Gametogenesis to Early Embryonic Development. Genes (Basel) 2021; 12:525. [PMID: 33916807 PMCID: PMC8067062 DOI: 10.3390/genes12040525] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
This review explores the role of reactive oxygen species (ROS)/Ca2+ in communication within reproductive structures in plants and animals. Many concepts have been described during the last years regarding how biosynthesis, generation products, antioxidant systems, and signal transduction involve ROS signaling, as well as its possible link with developmental processes and response to biotic and abiotic stresses. In this review, we first addressed classic key concepts in ROS and Ca2+ signaling in plants, both at the subcellular, cellular, and organ level. In the plant science field, during the last decades, new techniques have facilitated the in vivo monitoring of ROS signaling cascades. We will describe these powerful techniques in plants and compare them to those existing in animals. Development of new analytical techniques will facilitate the understanding of ROS signaling and their signal transduction pathways in plants and mammals. Many among those signaling pathways already have been studied in animals; therefore, a specific effort should be made to integrate this knowledge into plant biology. We here discuss examples of how changes in the ROS and Ca2+ signaling pathways can affect differentiation processes in plants, focusing specifically on reproductive processes where the ROS and Ca2+ signaling pathways influence the gametophyte functioning, sexual reproduction, and embryo formation in plants and animals. The study field regarding the role of ROS and Ca2+ in signal transduction is evolving continuously, which is why we reviewed the recent literature and propose here the potential targets affecting ROS in reproductive processes. We discuss the opportunities to integrate comparative developmental studies and experimental approaches into studies on the role of ROS/ Ca2+ in both plant and animal developmental biology studies, to further elucidate these crucial signaling pathways.
Collapse
Affiliation(s)
- Valentina Lodde
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety (VESPA), Università degli Studi di Milano, 20133 Milan, Italy;
| | - Piero Morandini
- Department of Environmental Science and Policy, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Alex Costa
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.C.); (I.M.)
| | - Irene Murgia
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.C.); (I.M.)
| | - Ignacio Ezquer
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.C.); (I.M.)
| |
Collapse
|
242
|
Zhang K, Zhou X, Wang J, Zhou Y, Qi W, Chen H, Nie S, Xie M. Dendrobium officinale polysaccharide triggers mitochondrial disorder to induce colon cancer cell death via ROS-AMPK-autophagy pathway. Carbohydr Polym 2021; 264:118018. [PMID: 33910741 DOI: 10.1016/j.carbpol.2021.118018] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
The homeostasis between mitochondrial function and autophagy is crucial to the physiological activity of cancer cells, and its mechanism is conducive to the development of anti-tumor drugs. Here, we aimed to explore the effect and mechanism of Dendrobium officinale polysaccharide (DOP) on colon cancer cell line CT26. Our data showed that DOP significantly inhibited the proliferation of CT26 cells and elevated autophagy level. Moreover, DOP disrupted mitochondrial function through increasing reactive oxygen species (ROS) and reducing mitochondrial membrane potential (MMP), thereby impairing ATP biosynthesis, which activated AMPK/mTOR autophagy signaling. Intriguingly, the further experiments demonstrated that DOP-induced cytotoxicity, excessive autophagy and mitochondrial dysfunction were reversed after CT26 cells pretreated with antioxidant (N-acetyl-l-cysteine). Herein, these findings implied that DOP-induced mitochondrial dysfunction and cytotoxic autophagy repressed the propagation of CT26 cells via ROS-ATP-AMPK signaling, providing a new opinion for the study of antineoplastic drugs.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Xingtao Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China.
| | - Junqiao Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Yujia Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Wucheng Qi
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Haihong Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China.
| |
Collapse
|
243
|
Li B, Xiong W, Liang WM, Chiou JS, Lin YJ, Chang ACY. Targeting of CAT and VCAM1 as Novel Therapeutic Targets for DMD Cardiomyopathy. Front Cell Dev Biol 2021; 9:659177. [PMID: 33869226 PMCID: PMC8047121 DOI: 10.3389/fcell.2021.659177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) related cardiomyopathy is the leading cause of early mortality in DMD patients. There is an urgent need to gain a better understanding of the disease molecular pathogenesis and develop effective therapies to prevent the onset of heart failure. In the present study, we used DMD human induced pluripotent stem cells (DMD-hiPSCs) derived cardiomyocytes (CMs) as a platform to explore the active compounds in commonly used Chinese herbal medicine (CHM) herbs. Single CHM herb (DaH, ZK, and CQZ) reduced cell beating rate, decreased cellular ROS accumulation, and improved structure of DMD hiPSC-CMs. Cross-comparison of transcriptomic profiling data and active compound library identified nine active chemicals targeting ROS neutralizing Catalase (CAT) and structural protein vascular cell adhesion molecule 1 (VCAM1). Treatment with Quecetin, Kaempferol, and Vitamin C, targeting CAT, conferred ROS protection and improved contraction; treatment with Hesperidin and Allicin, targeting VCAM1, induced structure enhancement via induction of focal adhesion. Lastly, overexpression of CAT or VCAM1 in DMD hiPSC-CMs reconstituted efficacious effects and conferred increase in cardiomyocyte function. Together, our results provide a new insight in treating DMD cardiomyopathy via targeting of CAT and VCAM1, and serves as an example of translating Bed to Bench back to Bed using a muti-omics approach.
Collapse
Affiliation(s)
- Bin Li
- Department of Cardiology and Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiyao Xiong
- Department of Cardiology and Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Miin Liang
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Jian-Shiun Chiou
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Ying-Ju Lin
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Alex C Y Chang
- Department of Cardiology and Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
244
|
Lkhagva B, Lin Y, Chen Y, Cheng W, Higa S, Kao Y, Chen Y. ZFHX3 knockdown dysregulates mitochondrial adaptations to tachypacing in atrial myocytes through enhanced oxidative stress and calcium overload. Acta Physiol (Oxf) 2021; 231:e13604. [PMID: 33332716 DOI: 10.1111/apha.13604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 01/02/2023]
Abstract
AIM To investigate the role of zinc finger homeobox 3 gene (ZFHX3) in tachypacing-induced mitochondrial dysfunction and explore its molecular mechanisms and potential as a therapeutic target in atrial fibrillation (AF). METHODS Through a bioluminescent assay, a patch clamp, confocal fluorescence and fluorescence microscopy, microplate enzyme activity assays and Western blotting, we studied ATP and ADP production, mitochondrial electron transfer chain complex activities, ATP-sensitive potassium channels (IKATP ), mitochondrial oxidative stress, Ca2+ content, and protein expression in control and ZFHX3 knockdown (KD) HL-1 cells subjected to 1 and 5-Hz pacing for 24 hours. RESULTS Compared with 1-Hz pacing, 5-Hz pacing increased ATP and ADP production, IKATP , phosphorylated adenosine monophosphate-activated protein kinase and inositol 1,4,5-triphosphate (IP3 ) receptor (IP3 R) protein expression. Tachypacing induced mitochondrial oxidative stress and Ca2+ overload in both cell types. Furthermore, under 1- and 5-Hz pacing, ZFHX3 KD cells showed higher IKATP , ATP and ADP production, mitochondrial oxidative stress and Ca2+ content than control cells. Under 5-Hz pacing, 2-aminoethoxydiphenyl borate (2-APB; 3 μmol/L, an IP3 R inhibitor) and MitoTEMPO (10 µmol/L, a mitochondria-targeted antioxidant) reduced ADP and increased ATP production in both cell types; however, only 2-APB significantly reduced mitochondrial Ca2+ overload in control cells. Under 5-Hz pacing, mitochondrial oxidative stress was significantly reduced by both MitoTEMPO and 2-APB and only by 2-APB in control and ZFHX3 KD cells respectively. CONCLUSION ZFHX3 KD cells modulate mitochondrial adaptations to tachypacing in HL-1 cardiomyocytes through Ca2+ overload, oxidative stress and metabolic disorder. Targeting IP3 R signalling or oxidative stress could reduce AF.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Yung‐Kuo Lin
- Division of Cardiology Department of Internal Medicine School of Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Yao‐Chang Chen
- Department of Biomedical Engineering National Defense Medical Center Taipei Taiwan
| | - Wan‐Li Cheng
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory Division of Cardiovascular Medicine Makiminato Central Hospital Okinawa Japan
| | - Yu‐Hsun Kao
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
- Department of Medical Education and Research Wan‐Fang Hospital Taipei Medical University Taipei Taiwan
| | - Yi‐Jen Chen
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
- Cardiovascular Research CenterWan‐Fang HospitalTaipei Medical University Taipei Taiwan
| |
Collapse
|
245
|
Alevriadou BR, Patel A, Noble M, Ghosh S, Gohil VM, Stathopulos PB, Madesh M. Molecular nature and physiological role of the mitochondrial calcium uniporter channel. Am J Physiol Cell Physiol 2021; 320:C465-C482. [PMID: 33296287 PMCID: PMC8260355 DOI: 10.1152/ajpcell.00502.2020] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Calcium (Ca2+) signaling is critical for cell function and cell survival. Mitochondria play a major role in regulating the intracellular Ca2+ concentration ([Ca2+]i). Mitochondrial Ca2+ uptake is an important determinant of cell fate and governs respiration, mitophagy/autophagy, and the mitochondrial pathway of apoptosis. Mitochondrial Ca2+ uptake occurs via the mitochondrial Ca2+ uniporter (MCU) complex. This review summarizes the present knowledge on the function of MCU complex, regulation of MCU channel, and the role of MCU in Ca2+ homeostasis and human disease pathogenesis. The channel core consists of four MCU subunits and essential MCU regulators (EMRE). Regulatory proteins that interact with them include mitochondrial Ca2+ uptake 1/2 (MICU1/2), MCU dominant-negative β-subunit (MCUb), MCU regulator 1 (MCUR1), and solute carrier 25A23 (SLC25A23). In addition to these proteins, cardiolipin, a mitochondrial membrane-specific phospholipid, has been shown to interact with the channel core. The dynamic interplay between the core and regulatory proteins modulates MCU channel activity after sensing local changes in [Ca2+]i, reactive oxygen species, and other environmental factors. Here, we highlight the structural details of the human MCU heteromeric assemblies and their known roles in regulating mitochondrial Ca2+ homeostasis. MCU dysfunction has been shown to alter mitochondrial Ca2+ dynamics, in turn eliciting cell apoptosis. Changes in mitochondrial Ca2+ uptake have been implicated in pathological conditions affecting multiple organs, including the heart, skeletal muscle, and brain. However, our structural and functional knowledge of this vital protein complex remains incomplete, and understanding the precise role for MCU-mediated mitochondrial Ca2+ signaling in disease requires further research efforts.
Collapse
Affiliation(s)
- B Rita Alevriadou
- Department of Biomedical Engineering, Jacobs School of Medicine and Biomedical Sciences and School of Engineering and Applied Sciences, University at Buffalo-State University of New York, Buffalo, New York
| | - Akshar Patel
- Department of Biomedical Engineering, Jacobs School of Medicine and Biomedical Sciences and School of Engineering and Applied Sciences, University at Buffalo-State University of New York, Buffalo, New York
| | - Megan Noble
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sagnika Ghosh
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Vishal M Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Muniswamy Madesh
- Department of Medicine/Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
246
|
Liu CJ, Cheng CW, Tsai YS, Huang HS. Crosstalk between Renal and Vascular Calcium Signaling: The Link between Nephrolithiasis and Vascular Calcification. Int J Mol Sci 2021; 22:ijms22073590. [PMID: 33808324 PMCID: PMC8036726 DOI: 10.3390/ijms22073590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Calcium (Ca2+) is an important mediator of multicellular homeostasis and is involved in several diseases. The interplay among the kidney, bone, intestine, and parathyroid gland in Ca2+ homeostasis is strictly modulated by numerous hormones and signaling pathways. The calcium-sensing receptor (CaSR) is a G protein–coupled receptor, that is expressed in calcitropic tissues such as the parathyroid gland and the kidney, plays a pivotal role in Ca2+ regulation. CaSR is important for renal Ca2+, as a mutation in this receptor leads to hypercalciuria and calcium nephrolithiasis. In addition, CaSR is also widely expressed in the vascular system, including vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) and participates in the process of vascular calcification. Aberrant Ca2+ sensing by the kidney and VSMCs, owing to altered CaSR expression or function, is associated with the formation of nephrolithiasis and vascular calcification. Based on emerging epidemiological evidence, patients with nephrolithiasis have a higher risk of vascular calcification, but the exact mechanism linking the two conditions is unclear. However, a dysregulation in Ca2+ homeostasis and dysfunction in CaSR might be the connection between the two. This review summarizes renal calcium handling and calcium signaling in the vascular system, with a special focus on the link between nephrolithiasis and vascular calcification.
Collapse
Affiliation(s)
- Chan-Jung Liu
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan; (C.-J.L.); (C.-W.C.)
| | - Chia-Wei Cheng
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan; (C.-J.L.); (C.-W.C.)
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan;
- Center for Clinical Medicine Research, National Cheng Kung University Hospital, Tainan 704302, Taiwan
| | - Ho-Shiang Huang
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan; (C.-J.L.); (C.-W.C.)
- Correspondence: ; Tel.: +886-6-2353535 (ext. 5251); Fax: +886-6-2766179
| |
Collapse
|
247
|
Zheng J, Chen P, Zhong J, Cheng Y, Chen H, He Y, Chen C. HIF‑1α in myocardial ischemia‑reperfusion injury (Review). Mol Med Rep 2021; 23:352. [PMID: 33760122 PMCID: PMC7974458 DOI: 10.3892/mmr.2021.11991] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a severe injury to the ischemic myocardium following the recovery of blood flow. Currently, there is no effective treatment for MIRI in clinical practice. Over the past two decades, biological studies of hypoxia and hypoxia-inducible factor-1α (HIF-1α) have notably improved understanding of oxygen homeostasis. HIF-1α is an oxygen-sensitive transcription factor that mediates adaptive metabolic responses to hypoxia and serves a pivotal role in MIRI. In particular, previous studies have demonstrated that HIF-1α improves mitochondrial function, decreases cellular oxidative stress, activates cardioprotective signaling pathways and downstream protective genes and interacts with non-coding RNAs. The present review summarizes the roles and associated mechanisms of action of HIF-1α in MIRI. In addition, HIF-1α-associated MIRI intervention, including natural compounds, exosomes, ischemic preconditioning and ischemic post-processing are presented. The present review provides evidence for the roles of HIF-1α activation in MIRI and supports its use as a therapeutic target.
Collapse
Affiliation(s)
- Jie Zheng
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Peier Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jianfeng Zhong
- Guangdong Key Laboratory of Age‑related Cardiac and Cerebral Diseases, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yu Cheng
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Hao Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Can Chen
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524003, P.R. China
| |
Collapse
|
248
|
Godsman N, Kohlhaas M, Nickel A, Cheyne L, Mingarelli M, Schweiger L, Hepburn C, Munts C, Welch A, Delibegovic M, Van Bilsen M, Maack C, Dawson DK. Metabolic alterations in a rat model of Takotsubo syndrome. Cardiovasc Res 2021; 118:1932-1946. [PMID: 33711093 PMCID: PMC9239582 DOI: 10.1093/cvr/cvab081] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Cardiac energetic impairment is a major finding in takotsubo patients. We investigate specific metabolic adaptations to direct future therapies. METHODS AND RESULTS An isoprenaline-injection female rat model (versus sham) was studied at day-3; recovery assessed at day-7. Substrate uptake, metabolism, inflammation and remodelling were investigated by 18F-FDG-PET, metabolomics, qPCR and WB. Isolated cardiomyocytes were patch-clamped during stress protocols for redox states of NAD(P)H/FAD or [Ca2+]c, [Ca2+]m and sarcomere length. Mitochondrial respiration was assessed by seahorse/Clark electrode (glycolytic and β-oxidation substrates).Cardiac 18F-FDG metabolic rate was increased in takotsubo (p = 0.006), as were expression of GLUT4-RNA/GLUT1/HK2-RNA and HK activity (all p < 0.05), with concomitant accumulation of glucose- and fructose-6-phosphates (p > 0.0001). Both lactate and pyruvate were lower (p < 0.05) despite increases in LDH-RNA and PDH (p < 0.05 both). β-oxidation enzymes CPT1b-RNA and 3KAT were increased (p < 0.01) but malonyl-CoA (CPT-1 regulator) was upregulated (p = 0.01) with decreased fatty acids and acyl-carnitines levels (p = 0.0001-0.02). Krebs cycle intermediates α-ketoglutarate and succinyl-carnitine were reduced (p < 0.05) as was cellular ATP reporter dihydroorotate (p = 0.003). Mitochondrial Ca2+ uptake during high workload was impaired on day-3 (p < 0.0001), inducing oxidation of NAD(P)H and FAD (p = 0.03) but resolved by day-7. There were no differences in mitochondrial respiratory function, sarcomere shortening or [Ca2+] transients of isolated cardiomyocytes, implying preserved integrity of both mitochondria and cardiomyocyte. Inflammation and remodelling were upregulated - increased CD68-RNA, collagen RNA/protein and skeletal actin RNA (all p < 0.05). CONCLUSION Dys-regulation of glucose and lipid metabolic pathways with decreases in final glycolytic and β-oxidation metabolites and reduced availability of Krebs intermediates characterises takotsubo myocardium. The energetic deficit accompanies defective Ca2+ handling, inflammation and upregulation of remodelling pathways, with preservation of sarcomeric and mitochondrial integrity. TRANSLATIONAL PERSPECTIVE The simultaneous dysregulation in the glycolytic and beta-oxidation pathways which underlies the energetic deficit of the takotsubo heart supports further testing of currently available metabolic modulators as possible candidates for successful therapy, as well as targeting the inflammatory and remodelling pathways.
Collapse
Affiliation(s)
- Nadine Godsman
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, United Kingdom
| | | | | | - Lesley Cheyne
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, United Kingdom
| | - Marco Mingarelli
- Biomedical physics, University of Aberdeen, Aberdeen, United Kingdom
| | - Lutz Schweiger
- John Mallard Scottish P.E.T. Centre, University of Aberdeen, Aberdeen, United Kingdom
| | - Claire Hepburn
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, United Kingdom
| | - Chantal Munts
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences Maastricht University, Netherlands
| | - Andy Welch
- Biomedical physics, University of Aberdeen, Aberdeen, United Kingdom
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, United Kingdom
| | - Marc Van Bilsen
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences Maastricht University, Netherlands
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), Würzburg, Germany
| | - Dana K Dawson
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
249
|
Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T, Chen D, Piao HL, Liu HX. The double-edged roles of ROS in cancer prevention and therapy. Theranostics 2021; 11:4839-4857. [PMID: 33754031 PMCID: PMC7978298 DOI: 10.7150/thno.56747] [Citation(s) in RCA: 282] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/31/2021] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) serve as cell signaling molecules generated in oxidative metabolism and are associated with a number of human diseases. The reprogramming of redox metabolism induces abnormal accumulation of ROS in cancer cells. It has been widely accepted that ROS play opposite roles in tumor growth, metastasis and apoptosis according to their different distributions, concentrations and durations in specific subcellular structures. These double-edged roles in cancer progression include the ROS-dependent malignant transformation and the oxidative stress-induced cell death. In this review, we summarize the notable literatures on ROS generation and scavenging, and discuss the related signal transduction networks and corresponding anticancer therapies. There is no doubt that an improved understanding of the sophisticated mechanism of redox biology is imperative to conquer cancer.
Collapse
|
250
|
Kreitmeier KG, Tarnowski D, Nanadikar MS, Baier MJ, Wagner S, Katschinski DM, Maier LS, Sag CM. CaMKII δ Met281/282 oxidation is not required for recovery of calcium transients during acidosis. Am J Physiol Heart Circ Physiol 2021; 320:H1199-H1212. [PMID: 33449853 DOI: 10.1152/ajpheart.00040.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/15/2020] [Accepted: 01/11/2021] [Indexed: 12/31/2022]
Abstract
CaMKII is needed for the recovery of Ca2+ transients during acidosis but also mediates postacidic arrhythmias. CaMKIIδ can sustain its activity following Met281/282 oxidation. Increasing cytosolic Na+ during acidosis as well as postacidic pH normalization should result in prooxidant conditions within the cell favoring oxidative CaMKIIδ activation. We tested whether CaMKIIδ activation through Met281/282 oxidation is involved in recovery of Ca2+ transients during acidosis and promotes cellular arrhythmias post-acidosis. Single cardiac myocytes were isolated from a well-established mouse model in which CaMKIIδ was made resistant to oxidative activation by knock-in replacement of two oxidant-sensitive methionines (Met281/282) with valines (MM-VV). MM-VV myocytes were exposed to extracellular acidosis (pHo 6.5) and compared to wild type (WT) control cells. Full recovery of Ca2+ transients was observed in both WT and MM-VV cardiac myocytes during late-phase acidosis. This was associated with comparably enhanced sarcoplasmic reticulum Ca2+ load and preserved CaMKII specific phosphorylation of phospholamban at Thr17 in MM-VV myocytes. CaMKII was phosphorylated at Thr287, but not Met281/282 oxidized. In line with this, postacidic cellular arrhythmias occurred to a similar extent in WT and MM-VV cells, whereas inhibition of CaMKII using AIP completely prevented recovery of Ca2+ transients during acidosis and attenuated postacidic arrhythmias in MM-VV cells. Using genetically altered cardiomyocytes with cytosolic expression of redox-sensitive green fluorescent protein-2 coupled to glutaredoxin 1, we found that acidosis has a reductive effect within the cytosol of cardiac myocytes despite a significant acidosis-related increase in cytosolic Na+. Our study shows that activation of CaMKIIδ through Met281/282 oxidation is neither required for recovery of Ca2+ transients during acidosis nor relevant for postacidic arrhythmogenesis in isolated cardiac myocytes. Acidosis reduces the cytosolic glutathione redox state of isolated cardiac myocytes despite a significant increase in cytosolic Na+. Pharmacological inhibition of global CaMKII activity completely prevents recovery of Ca2+ transients and protects from postacidic arrhythmias in MM-VV myocytes, which confirms the relevance of CaMKII in the context of acidosis.NEW & NOTEWORTHY The current study shows that activation of CaMKIIδ through Met281/282 oxidation is neither required for CaMKII-dependent recovery of Ca2+ transients during acidosis nor relevant for the occurrence of postacidic cellular arrhythmias. Despite a usually prooxidant increase in cytosolic Na+, acidosis reduces the cytosolic glutathione redox state within cardiac myocytes. This novel finding suggests that oxidation of cytosolic proteins is less likely to occur during acidosis.
Collapse
Affiliation(s)
- K G Kreitmeier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
- Department of Internal Medicine III, University Medical Center Regensburg, Germany
| | - D Tarnowski
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - M S Nanadikar
- Institute for Cardiovascular Physiology, Georg August University, Göttingen, Germany
| | - M J Baier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - S Wagner
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - D M Katschinski
- Institute for Cardiovascular Physiology, Georg August University, Göttingen, Germany
| | - L S Maier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| | - C M Sag
- Department of Internal Medicine II, University Medical Center Regensburg, Germany
| |
Collapse
|