1
|
Wang J, Jiang J, Hu H, Chen L. MCU complex: Exploring emerging targets and mechanisms of mitochondrial physiology and pathology. J Adv Res 2025; 68:271-298. [PMID: 38417574 PMCID: PMC11785567 DOI: 10.1016/j.jare.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND Globally, the onset and progression of multiple human diseases are associated with mitochondrial dysfunction and dysregulation of Ca2+ uptake dynamics mediated by the mitochondrial calcium uniporter (MCU) complex, which plays a key role in mitochondrial dysfunction. Despite relevant studies, the underlying pathophysiological mechanisms have not yet been fully elucidated. AIM OF REVIEW This article provides an in-depth analysis of the current research status of the MCU complex, focusing on its molecular composition, regulatory mechanisms, and association with diseases. In addition, we conducted an in-depth analysis of the regulatory effects of agonists, inhibitors, and traditional Chinese medicine (TCM) monomers on the MCU complex and their application prospects in disease treatment. From the perspective of medicinal chemistry, we conducted an in-depth analysis of the structure-activity relationship between these small molecules and MCU and deduced potential pharmacophores and binding pockets. Simultaneously, key structural domains of the MCU complex in Homo sapiens were identified. We also studied the functional expression of the MCU complex in Drosophila, Zebrafish, and Caenorhabditis elegans. These analyses provide a basis for exploring potential treatment strategies targeting the MCU complex and provide strong support for the development of future precision medicine and treatments. KEY SCIENTIFIC CONCEPTS OF REVIEW The MCU complex exhibits varying behavior across different tissues and plays various roles in metabolic functions. It consists of six MCU subunits, an essential MCU regulator (EMRE), and solute carrier 25A23 (SLC25A23). They regulate processes, such as mitochondrial Ca2+ (mCa2+) uptake, mitochondrial adenosine triphosphate (ATP) production, calcium dynamics, oxidative stress (OS), and cell death. Regulation makes it a potential target for treating diseases, especially cardiovascular diseases, neurodegenerative diseases, inflammatory diseases, metabolic diseases, and tumors.
Collapse
Affiliation(s)
- Jin Wang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Jinyong Jiang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou 416000, China
| | - Haoliang Hu
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China; College of Medicine, Hunan University of Arts and Science, Changde 415000, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China.
| |
Collapse
|
2
|
Suzuki I, Xing H, Giblin J, Ashraf A, Chung EJ. Nanoparticle-based therapeutic strategies for mitochondrial dysfunction in cardiovascular disease. J Biomed Mater Res A 2024; 112:895-913. [PMID: 38217313 DOI: 10.1002/jbm.a.37668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/05/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024]
Abstract
Although cardiovascular diseases (CVD) are the leading cause of global mortality, there is a lack of therapies that target and revert underlying pathological processes. Mitochondrial dysfunction is involved in the pathophysiology of CVD, and thus is a potential target for therapeutic development. To target the mitochondria and improve therapeutic efficacy, nanoparticle-based delivery systems have been proposed as promising strategies for the delivery of therapeutic agents to the mitochondria. This review will first discuss how mitochondrial dysfunction is related to the progression of several CVD and then delineate recent progress in mitochondrial targeting using nanoparticle-based delivery systems including peptide-based nanosystems, polymeric nanoparticles, liposomes, and lipid nanoparticles. In addition, we summarize the advantages of these nanocarriers and remaining challenges in targeting the mitochondria as a therapeutic strategy for CVD treatment.
Collapse
Affiliation(s)
- Isabella Suzuki
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Huihua Xing
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Joshua Giblin
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Anisa Ashraf
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Eun Ji Chung
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
- Bridge Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
3
|
Danylovych H, Danylovych Y, Chunikhin A, Cherenok S, Kalchenko V, Kosterin S. Use of thiacalix[4]arene C-1193 for a directed influence on the functional activity of mitochondria and simulation of this process using a Petri nets. BIOTECHNOLOGIA 2024; 105:69-81. [PMID: 38633893 PMCID: PMC11020151 DOI: 10.5114/bta.2024.135643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 04/19/2024] Open
Abstract
In molecular biological studies, considerable attention is paid to macrocyclic nanoscale compounds known as calix[4]arenes. An imperative concern in biochemical membranology and molecular biotechnology is the exploration of effectors capable of modifying the intensity of redox reactions within the inner mitochondrial membrane and influencing the activity of its Ca2+ transport systems. The simulation model development is relevant to formalize and generalize the experimental data and assess the conformity of experimental results with theoretical predictions. Experiments were carried out on a suspension of isolated rat myometrial mitochondria. The synthesized thiacalix[4]arene C-1193, containing four sulfur atoms, was employed. Demonstrations of time-dependent and concentration-dependent (0.01-10 μM) inhibition of Ca2+ accumulation and reactive oxygen species (ROS) formation by mitochondria in the presence of C-1193 were observed. While C-1193 inhibited the oxidation of NADH and FADH2, it did not induce mitochondrial swelling. The thiacalix[4]arene also inhibited the synthesis of nitric oxide, with a Ki of 5.5 ± 1.7 nM, positioning it as a high-affinity blocker of endogenous NO generation in mitochondria. These results are the basis for the possible application of the synthesized thiacalix[4]arene as a tool in researching biochemical processes in mitochondria. A simulation model employing functional hybrid Petri nets was developed, reproducing the functional activity of mitochondria, including simultaneous NADH oxidation, ROS formation, NO synthesis, and Ca2+ accumulation. The derived equations formalize and describe the time dependencies of the listed processes in the medium under the influence of thiacalix[4]arene C-1193.
Collapse
Affiliation(s)
- Hanna Danylovych
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Yurii Danylovych
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Alexander Chunikhin
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Sergiy Cherenok
- Institute of Organic Chemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Vitaly Kalchenko
- Institute of Organic Chemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Sergiy Kosterin
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| |
Collapse
|
4
|
Shen C, Ding X, Ruan J, Ruan F, Hu W, Huang J, He C, Yu Y, Zuo Z. Black phosphorus quantum dots induce myocardial inflammatory responses and metabolic disorders in mice. J Environ Sci (China) 2024; 137:53-64. [PMID: 37980037 DOI: 10.1016/j.jes.2023.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 11/20/2023]
Abstract
As an ultrasmall derivative of black phosphorus (BP) sheets, BP quantum dots (BP-QDs) have been effectively used in many fields. Currently, information on the cardiotoxicity induced by BP-QDs remains limited. We aimed to evaluate BP-QD-induced cardiac toxicity in mice. Histopathological examination of heart tissue sections was performed. Transcriptome sequencing, real-time quantitative PCR (RT‒qPCR), western blotting, and enzyme-linked immunosorbent assay (ELISA) assays were used to detect the mRNA and/or protein expression of proinflammatory cytokines, nuclear factor kappa B (NF-κB), phosphatidylinositol 3 kinase-protein kinase B (PI3K-AKT), peroxisome proliferator-activated receptor gamma (PPARγ), and glucose/lipid metabolism pathway-related genes. We found that heart weight and heart/body weight index (HBI) were significantly reduced in mice after intragastric administration of 0.1 or 1 mg/kg BP-QDs for 28 days. In addition, obvious inflammatory cell infiltration and increased cardiomyocyte diameter were observed in the BP-QD-treated groups. Altered expression of proinflammatory cytokines and genes related to the NF-κB signaling pathway further confirmed that BP-QD exposure induced inflammatory responses. In addition, BP-QD treatment also affected the PI3K-AKT, PPARγ, thermogenesis, oxidative phosphorylation, and cardiac muscle contraction signaling pathways. The expression of genes related to glucose/lipid metabolism signaling pathways was dramatically affected by BP-QD exposure, and the effect was primarily mediated by the PPAR signaling pathway. Our study provides new insights into the toxicity of BP-QDs to human health.
Collapse
Affiliation(s)
- Chao Shen
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiaoyan Ding
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jinpeng Ruan
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Fengkai Ruan
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Weiping Hu
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jiyi Huang
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chengyong He
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yi Yu
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Zhenghong Zuo
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
5
|
Schenkl C, Heyne E, Doenst T, Schulze PC, Nguyen TD. Targeting Mitochondrial Metabolism to Save the Failing Heart. Life (Basel) 2023; 13:life13041027. [PMID: 37109556 PMCID: PMC10143865 DOI: 10.3390/life13041027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable progress in treating cardiac disorders, the prevalence of heart failure (HF) keeps growing, making it a global medical and economic burden. HF is characterized by profound metabolic remodeling, which mostly occurs in the mitochondria. Although it is well established that the failing heart is energy-deficient, the role of mitochondria in the pathophysiology of HF extends beyond the energetic aspects. Changes in substrate oxidation, tricarboxylic acid cycle and the respiratory chain have emerged as key players in regulating myocardial energy homeostasis, Ca2+ handling, oxidative stress and inflammation. This work aims to highlight metabolic alterations in the mitochondria and their far-reaching effects on the pathophysiology of HF. Based on this knowledge, we will also discuss potential metabolic approaches to improve cardiac function.
Collapse
Affiliation(s)
- Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Paul Christian Schulze
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tien Dung Nguyen
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
6
|
Pedriali G, Ramaccini D, Bouhamida E, Wieckowski MR, Giorgi C, Tremoli E, Pinton P. Perspectives on mitochondrial relevance in cardiac ischemia/reperfusion injury. Front Cell Dev Biol 2022; 10:1082095. [PMID: 36561366 PMCID: PMC9763599 DOI: 10.3389/fcell.2022.1082095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the most common cause of death worldwide and in particular, ischemic heart disease holds the most considerable position. Even if it has been deeply studied, myocardial ischemia-reperfusion injury (IRI) is still a side-effect of the clinical treatment for several heart diseases: ischemia process itself leads to temporary damage to heart tissue and obviously the recovery of blood flow is promptly required even if it worsens the ischemic injury. There is no doubt that mitochondria play a key role in pathogenesis of IRI: dysfunctions of these important organelles alter cell homeostasis and survival. It has been demonstrated that during IRI the system of mitochondrial quality control undergoes alterations with the disruption of the complex balance between the processes of mitochondrial fusion, fission, biogenesis and mitophagy. The fundamental role of mitochondria is carried out thanks to the finely regulated connection to other organelles such as plasma membrane, endoplasmic reticulum and nucleus, therefore impairments of these inter-organelle communications exacerbate IRI. This review pointed to enhance the importance of the mitochondrial network in the pathogenesis of IRI with the aim to focus on potential mitochondria-targeting therapies as new approach to control heart tissue damage after ischemia and reperfusion process.
Collapse
Affiliation(s)
- Gaia Pedriali
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | | | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| |
Collapse
|
7
|
Qi X, Zhu Z, Wang Y, Wen Z, Jiang Z, Zhang L, Pang Y, Lu J. Research progress on the relationship between mitochondrial function and heart failure: A bibliometric study from 2002 to 2021. Front Mol Biosci 2022; 9:1036364. [PMID: 36330217 PMCID: PMC9622797 DOI: 10.3389/fmolb.2022.1036364] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/07/2022] [Indexed: 11/14/2022] Open
Abstract
Heart failure is one of the major public health problems in the world. In recent years, more and more attention has been paid to the relationship between heart failure and mitochondrial function. In the past 2 decades, a growing number of research papers in this field have been published. This study conducted a bibliometric analysis of the published literature on the relationship between MF and HF in the past 20 years by utilizing Microsoft Excel 2019, Biblio metric analysis platform, WoSCC database, VosViewer and Citespace. The results show that the papers have increased year by year and China and the United States are the leading countries in this field, as well as the countries with the most cooperation and exchanges. University of california system is the research institution with the greatest impacts on research results, and Yip H.K. is the author with more papers. The American Journal of Physiology-heart and Circulatory Physiology is probably the most popular magazine. At present, most of the published articles on mitochondria and HF are cited from internationally influential journals. The research focus includes oxidative stress, metabolic dysfunction, mitochondrial Ca2+ homeostasis imbalance, mitochondrial quality control and mitochondrial dysfunction mediated by inflammation in the pathogenesis of HF. Targeted regulating of mitochondria will be the keynote of future research on prevention and treatment of HF.
Collapse
Affiliation(s)
- Xiang Qi
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhide Zhu
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yuhan Wang
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhihao Wen
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, Chinad
| | - Zhixiong Jiang
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Liren Zhang
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yan Pang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, Chinad
- *Correspondence: Yan Pang, ; Jianqi Lu,
| | - Jianqi Lu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, Chinad
- *Correspondence: Yan Pang, ; Jianqi Lu,
| |
Collapse
|
8
|
Danylovych YV, Danylovych HV, Kolomiets OV, Sviatnenko MD, Kosterin SO. Biochemical properties of H +-Ca 2+-exchanger in the myometrium mitochondria. Curr Res Physiol 2022; 5:369-380. [PMID: 36176920 PMCID: PMC9513619 DOI: 10.1016/j.crphys.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022] Open
Abstract
Some biochemical properties of the H+-Ca2+-exchanger in uterine smooth muscle mitochondria have been described. The experiments were performed on a suspension of isolated mitochondria from the myometrium of rats. Methods of confocal microscopy, spectrofluorimetry and photon correlation spectroscopy were used. Fluo-4 probe was used to record changes in ionized Ca2+ in the matrix and cytosol; pH changes in the matrix were evaluated with BCECF. It was experimentally proved that in the myometrium instead of Na+-Ca2+-exchanger the H+-Ca2+-exchanger functions. It was activated at a physiological pH value, was carried out in stoichiometry 1: 1 and was electrogenic. The transport system was modulated by magnesium ions and the diuretic amiloride, but was not sensitive to changes in the concentration of extra-mitochondrial potassium ions. H+-Ca2+-exchanger was suppressed by antibodies against the LETM1 protein. Calmodulin may act as a regulator of H+-Ca2+-exchanger by inhibiting it.
Collapse
Affiliation(s)
- Yurii V. Danylovych
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Hanna V. Danylovych
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Oksana V. Kolomiets
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Marina D. Sviatnenko
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Sergiy O. Kosterin
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
9
|
Behavioral innovation and genomic novelty are associated with the exploitation of a challenging dietary opportunity by an avivorous bat. iScience 2022; 25:104973. [PMID: 36093062 PMCID: PMC9459691 DOI: 10.1016/j.isci.2022.104973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/12/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022] Open
Abstract
Foraging on nocturnally migrating birds is one of the most challenging foraging tasks in the animal kingdom. Only three bat species (e.g., Ia io) known to date can prey on migratory birds. However, how these bats have exploited this challenging dietary niche remains unknown. Here, we demonstrate that I. io hunts at the altitude of migrating birds during the bird migration season. The foraging I. io exhibited high flight altitudes (up to 4945 m above sea level) and high flight speeds (up to 143.7 km h−1). I. io in flight can actively prey on birds in the night sky via echolocation cues. Genes associated with DNA damage repair, hypoxia adaptation, biting and mastication, and digestion and metabolism have evolved to adapt to this species’ avivorous habits. Our results suggest that the evolution of behavioral innovation and genomic novelty are associated with the exploitation of challenging dietary opportunities. Predation on nocturnally migrating birds is rare and challenging in nature Bats exhibit high flight altitude and speed associated with foraging on migrating birds Bats can actively prey on birds in the night sky via echolocation cues The adaptive evolution of genes enables bats to adapt to the avivorous habits
Collapse
|
10
|
Kit O, Frantsiyants E, Shikhlyarova A, Neskubina I, Kaplieva I, Cheryarina N, Vereskunova A, Trepitaki L, Pogorelova Y, Bandovkina V, Surikova E, Kachesova P, Sheiko E, Kotieva I, Gusareva M, Luganskaya R, Bosenko E. Biological effects of mitochondrial therapy: preventing development of myocardial infarction and blocking metastatic aggression of B16/F10 melanoma. CARDIOMETRY 2022. [DOI: 10.18137/cardiometry.2022.22.5055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The aim is to evaluate the physiological parameters of the efficacy of cardiac mitochondria transplantation in male mice with chronic neurogenic pain and B16/F10 melanoma growth. Materials and methods. Male mice (n=37) of the C57BL/6 line were used in the research work. The animals covered experimental groups as follows: mice with chronic neurogenic pain (CNP) + B16/F10 melanoma (n=27); mice with CNB + B16/ F10 melanoma + mitochondrial therapy (MC therapy) (n=10). Mitochondria were isolated from the heart of an intact rat with the use of differential centrifugation. An introduction of mitochondria to mice was carried out daily intraperitoneally at a dose of 3.3 mg of protein for 3 weeks. Statistical analysis of the results is carried out with the Statistica 10.0 software. Results. On day 21 (week 3) of the experiment, macroscopically in the melanoma tissue in the group of animals with MC therapy, there were 2.5 times more necrosis cases than in the group without MC therapy. During the examination of the internal organs, no metastases were detected in the animals treated with MC therapy, while in 100% of the animals without MC therapy metastases were found in the lungs and in 95% of them in the spleen. In the animals received MC therapy, there was no damage to the heart muscle in 75% of the cases, while in the group of the animals without MC therapy, the presence of lesions in the form of bruises on the surface of the heart was macroscopically detected in 100% of the animals. Conclusion. Thus, intraperitoneal transplantation of intact heart mitochondria contributed to the prevention of myocardial infarction and metastases to internal organs in the C57BL/6 male mice with B16/F10 melanoma growing against the background of chronic neurogenic pain.
Collapse
|
11
|
Mitochondrial Ca 2+ Homeostasis: Emerging Roles and Clinical Significance in Cardiac Remodeling. Int J Mol Sci 2022; 23:ijms23063025. [PMID: 35328444 PMCID: PMC8954803 DOI: 10.3390/ijms23063025] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 01/27/2023] Open
Abstract
Mitochondria are the sites of oxidative metabolism in eukaryotes where the metabolites of sugars, fats, and amino acids are oxidized to harvest energy. Notably, mitochondria store Ca2+ and work in synergy with organelles such as the endoplasmic reticulum and extracellular matrix to control the dynamic balance of Ca2+ concentration in cells. Mitochondria are the vital organelles in heart tissue. Mitochondrial Ca2+ homeostasis is particularly important for maintaining the physiological and pathological mechanisms of the heart. Mitochondrial Ca2+ homeostasis plays a key role in the regulation of cardiac energy metabolism, mechanisms of death, oxygen free radical production, and autophagy. The imbalance of mitochondrial Ca2+ balance is closely associated with cardiac remodeling. The mitochondrial Ca2+ uniporter (mtCU) protein complex is responsible for the uptake and release of mitochondrial Ca2+ and regulation of Ca2+ homeostasis in mitochondria and consequently, in cells. This review summarizes the mechanisms of mitochondrial Ca2+ homeostasis in physiological and pathological cardiac remodeling and the regulatory effects of the mitochondrial calcium regulatory complex on cardiac energy metabolism, cell death, and autophagy, and also provides the theoretical basis for mitochondrial Ca2+ as a novel target for the treatment of cardiovascular diseases.
Collapse
|
12
|
Zhang X, Tomar N, Kandel SM, Audi SH, Cowley AW, Dash RK. Substrate- and Calcium-Dependent Differential Regulation of Mitochondrial Oxidative Phosphorylation and Energy Production in the Heart and Kidney. Cells 2021; 11:131. [PMID: 35011693 PMCID: PMC8750792 DOI: 10.3390/cells11010131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dehydrogenases are differentially stimulated by Ca2+. Ca2+ has also diverse regulatory effects on mitochondrial transporters and other enzymes. However, the consequences of these regulatory effects on mitochondrial oxidative phosphorylation (OxPhos) and ATP production, and the dependencies of these consequences on respiratory substrates, have not been investigated between the kidney and heart despite the fact that kidney energy requirements are second only to those of the heart. Our objective was, therefore, to elucidate these relationships in isolated mitochondria from the kidney outer medulla (OM) and heart. ADP-induced mitochondrial respiration was measured at different CaCl2 concentrations in the presence of various respiratory substrates, including pyruvate + malate (PM), glutamate + malate (GM), alpha-ketoglutarate + malate (AM), palmitoyl-carnitine + malate (PCM), and succinate + rotenone (SUC + ROT). The results showed that, in both heart and OM mitochondria, and for most complex I substrates, Ca2+ effects are biphasic: small increases in Ca2+ concentration stimulated, while large increases inhibited mitochondrial respiration. Furthermore, significant differences in substrate- and Ca2+-dependent O2 utilization towards ATP production between heart and OM mitochondria were observed. With PM and PCM substrates, Ca2+ showed more prominent stimulatory effects in OM than in heart mitochondria, while with GM and AM substrates, Ca2+ had similar biphasic regulatory effects in both OM and heart mitochondria. In contrast, with complex II substrate SUC + ROT, only inhibitory effects on mitochondrial respiration was observed in both the heart and the OM. We conclude that the regulatory effects of Ca2+ on mitochondrial OxPhos and ATP synthesis are biphasic, substrate-dependent, and tissue-specific.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Sunil M. Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Said H. Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, WI 53223, USA;
| | - Allen W. Cowley
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
13
|
Li P, Zhao H, Zhang J, Ning Y, Tu Y, Xu D, Zeng Q. Similarities and Differences Between HFmrEF and HFpEF. Front Cardiovasc Med 2021; 8:678614. [PMID: 34616777 PMCID: PMC8488158 DOI: 10.3389/fcvm.2021.678614] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
The new guidelines classify heart failure (HF) into three subgroups based on the ejection fraction (EF): HF with reduced EF (HFrEF), HF with mid-range EF (HFmrEF), and HF with preserved EF (HFpEF). The new guidelines regarding the declaration of HFmrEF as a unique phenotype have achieved the goal of stimulating research on the basic characteristics, pathophysiology, and treatment of HF patients with a left ventricular EF of 40-49%. Patients with HFmrEF have more often been described as an intermediate population between HFrEF and HFpEF patients; however, with regard to etiology and clinical indicators, they are more similar to the HFrEF population. Concerning clinical prognosis, they are closer to HFpEF because both populations have a good prognosis and quality of life. Meanwhile, growing evidence indicates that HFmrEF and HFpEF show heterogeneity in presentation and pathophysiology, and the emergence of this heterogeneity often plays a crucial role in the prognosis and treatment of the disease. To date, the exact mechanisms and effective treatment strategies of HFmrEF and HFpEF are still poorly understood, but some of the current evidence, from observational studies and post-hoc analyses of randomized controlled trials, have shown that patients with HFmrEF may benefit more from HFrEF treatment strategies, such as beta-blockers, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, mineralocorticoid receptor antagonists, and sacubitril/valsartan. This review summarizes available data from current clinical practice and mechanistic studies in terms of epidemiology, etiology, clinical indicators, mechanisms, and treatments to discuss the potential association between HFmrEF and HFpEF patients.
Collapse
Affiliation(s)
- Peixin Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hengli Zhao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jianyu Zhang
- Department of Cardiology, Foshan First People's Hospital, Foshan, Guangdong, China
| | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yan Tu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
14
|
Modesti L, Danese A, Angela Maria Vitto V, Ramaccini D, Aguiari G, Gafà R, Lanza G, Giorgi C, Pinton P. Mitochondrial Ca 2+ Signaling in Health, Disease and Therapy. Cells 2021; 10:cells10061317. [PMID: 34070562 PMCID: PMC8230075 DOI: 10.3390/cells10061317] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
The divalent cation calcium (Ca2+) is considered one of the main second messengers inside cells and acts as the most prominent signal in a plethora of biological processes. Its homeostasis is guaranteed by an intricate and complex system of channels, pumps, and exchangers. In this context, by regulating cellular Ca2+ levels, mitochondria control both the uptake and release of Ca2+. Therefore, at the mitochondrial level, Ca2+ plays a dual role, participating in both vital physiological processes (ATP production and regulation of mitochondrial metabolism) and pathophysiological processes (cell death, cancer progression and metastasis). Hence, it is not surprising that alterations in mitochondrial Ca2+ (mCa2+) pathways or mutations in Ca2+ transporters affect the activities and functions of the entire cell. Indeed, it is widely recognized that dysregulation of mCa2+ signaling leads to various pathological scenarios, including cancer, neurological defects and cardiovascular diseases (CVDs). This review summarizes the current knowledge on the regulation of mCa2+ homeostasis, the related mechanisms and the significance of this regulation in physiology and human diseases. We also highlight strategies aimed at remedying mCa2+ dysregulation as promising therapeutical approaches.
Collapse
Affiliation(s)
- Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Alberto Danese
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Daniela Ramaccini
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Giovanni Lanza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
- Correspondence: ; Tel.: +39-0532-455802
| |
Collapse
|
15
|
Morciano G, Vitto VAM, Bouhamida E, Giorgi C, Pinton P. Mitochondrial Bioenergetics and Dynamism in the Failing Heart. Life (Basel) 2021; 11:436. [PMID: 34066065 PMCID: PMC8151847 DOI: 10.3390/life11050436] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
The heart is responsible for pumping blood, nutrients, and oxygen from its cavities to the whole body through rhythmic and vigorous contractions. Heart function relies on a delicate balance between continuous energy consumption and generation that changes from birth to adulthood and depends on a very efficient oxidative metabolism and the ability to adapt to different conditions. In recent years, mitochondrial dysfunctions were recognized as the hallmark of the onset and development of manifold heart diseases (HDs), including heart failure (HF). HF is a severe condition for which there is currently no cure. In this condition, the failing heart is characterized by a disequilibrium in mitochondrial bioenergetics, which compromises the basal functions and includes the loss of oxygen and substrate availability, an altered metabolism, and inefficient energy production and utilization. This review concisely summarizes the bioenergetics and some other mitochondrial features in the heart with a focus on the features that become impaired in the failing heart.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care&Research, 48033 Cotignola, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Esmaa Bouhamida
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care&Research, 48033 Cotignola, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| |
Collapse
|
16
|
Cao X, Ma Q, Wang B, Qian Q, Liu N, Liu T, Dong X. Silencing long non-coding RNA MIAT ameliorates myocardial dysfunction induced by myocardial infarction via MIAT/miR-10a-5p/EGR2 axis. Aging (Albany NY) 2021; 13:11188-11206. [PMID: 33819189 PMCID: PMC8109106 DOI: 10.18632/aging.202785] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Long non-coding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) has been widely-demonstrated to function as diagnostic markers for acute myocardial infarction (MI). This study was designed to explore the modulatory role of MIAT and its underlying molecular mechanism in MI. Firstly, MI mouse model was developed via ligation of the descending branch of the left coronary artery, and cell model was established through exposure to hypoxic conditions. Online prediction indicated that MIAT could bind to microRNA-10a-5p (miR-10a-5p), while miR-10a-5p was highlighted to bind to early growth response gene-2 (EGR2). MIAT and EGR2 were subsequently determined to be highly-expressed, whereas miR-10a-5p was found to be poorly-expressed in cardiomyocytes exposed to hypoxia as well as in MI mice using RT-qPCR and Western blot assay. The binding relationships between MIAT and miR-10a-5p, and between miR-10a-5p and EGR2 were further confirmed by dual-luciferase reporter and RNA immunoprecipitation assays. The results of in vitro and in vivo experimentation also suggested that overexpression of miR-10a-5p or silencing of MIAT and EGR2 reduced cardiomyocyte apoptosis and increased ATP content, thus alleviating the impairment of cardiac function following MI. In a word, inhibition of MIAT protects against cardiac dysfunction induced by MI through the crosstalk with miR-10a-5p/EGR2.
Collapse
Affiliation(s)
- Xiangke Cao
- School of Life Sciences, North China University of Science and Technology, Tangshan 063210, P.R. China
| | - Qinghua Ma
- Department of Preventive Health, The Third People's Hospital Of Xiangcheng District In Suzhou, Suzhou 215134, P.R. China
| | - Bin Wang
- Department of Pediatrics, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, P.R. China
| | - Qingqiang Qian
- Department of Neurology, Tangshan Gongren Hospital, Tangshan 063000, P.R. China
| | - Ning Liu
- Department of Cardiovascular Diseases, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, P.R. China
| | - Tiejun Liu
- Department of Anesthesiology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, P.R. China
| | - Xiaoliu Dong
- Department of Neurology, Tangshan People's Hospital, Tangshan 063001, P.R. China
| |
Collapse
|
17
|
Qi Y, Li JJ, Di XH, Zhang Y, Chen JL, Wu ZX, Man ZY, Bai RY, Lu F, Tong J, Liu XL, Deng XL, Zhang J, Zhang X, Zhang Y, Xie W. Excess sarcoplasmic reticulum-mitochondria calcium transport induced by Sphingosine-1-phosphate contributes to cardiomyocyte hypertrophy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118970. [PMID: 33529640 DOI: 10.1016/j.bbamcr.2021.118970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 10/22/2022]
Abstract
Sphingosine-1-phosphate (S1P) has been shown to possess pro-hypertrophic properties in the heart, but the detailed molecular mechanism that underlies the pathological process is rarely explored. In the present study, we aim to explore the role of S1P-mediated intracellular Ca2+ signaling, with a focus on sarcoplasmic reticulum (SR)-mitochondria communication, in cardiomyocyte hypertrophy. Cultured neonatal rat ventricular myocytes (NRVMs) displayed significantly hypertrophic growth after treatment with 1 μmol/L S1P for 48 h, as indicated by the cell surface area or mRNA expressions of hypertrophic marker genes (ANP, BNP and β-MHC). Importantly, mitochondrial Ca2+ and reactive oxygen species (ROS) levels were dramatically elevated upon S1P stimulation, and pharmacological blockage of which abolished NRVM hypertrophy. 0.5 Hz electrical pacing induced similar cytosolic Ca2+ kinetics to S1P stimulation, but unaffected the peak of mitochondrial [Ca2+]. With interference of the expression of type 2 inositol 1,4,5-trisphosphate receptors (IP3R2), which are unemployed in electrical paced Ca2+ activity but may be activated by S1P, alteration in mitochondrial Ca2+ as well as the hypertrophic effect in NRVMs under S1P stimulation were attenuated. The hypertrophic effect of S1P can also be abolished by pharmacological block of S1PR1 or Gi signaling. Collectively, our study highlights the mechanistic role of IP3R2-mediated excess SR-mitochondria Ca2+ transport in S1P-induced cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Ying Qi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Jing-Jing Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xiao-Hui Di
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Yu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Jie-Long Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Zi-Xuan Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Zi-Yue Man
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Ru-Yue Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Fujian Lu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jie Tong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xue-Liang Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Jianbao Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xing Zhang
- Department of Aerospace Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.
| | - Wenjun Xie
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
18
|
Miranda-Silva D, Lima T, Rodrigues P, Leite-Moreira A, Falcão-Pires I. Mechanisms underlying the pathophysiology of heart failure with preserved ejection fraction: the tip of the iceberg. Heart Fail Rev 2021; 26:453-478. [PMID: 33411091 DOI: 10.1007/s10741-020-10042-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifaceted syndrome with a complex aetiology often associated with several comorbidities, such as left ventricle pressure overload, diabetes mellitus, obesity, and kidney disease. Its pathophysiology remains obscure mainly due to the complex phenotype induced by all these associated comorbidities and to the scarcity of animal models that adequately mimic HFpEF. Increased oxidative stress, inflammation, and endothelial dysfunction are currently accepted as key players in HFpEF pathophysiology. However, we have just started to unveil HFpEF complexity and the role of calcium handling, energetic metabolism, and mitochondrial function remain to clarify. Indeed, the enlightenment of such cellular and molecular mechanisms represents an opportunity to develop novel therapeutic approaches and thus to improve HFpEF treatment options. In the last decades, the number of research groups dedicated to studying HFpEF has increased, denoting the importance and the magnitude achieved by this syndrome. In the current technological and web world, the amount of information is overwhelming, driving us not only to compile the most relevant information about the theme but also to explore beyond the tip of the iceberg. Thus, this review aims to encompass the most recent knowledge related to HFpEF or HFpEF-associated comorbidities, focusing mainly on myocardial metabolism, oxidative stress, and energetic pathways.
Collapse
Affiliation(s)
- Daniela Miranda-Silva
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Tânia Lima
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Patrícia Rodrigues
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
19
|
The Physiological and Pathological Roles of Mitochondrial Calcium Uptake in Heart. Int J Mol Sci 2020; 21:ijms21207689. [PMID: 33080805 PMCID: PMC7589179 DOI: 10.3390/ijms21207689] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Calcium ion (Ca2+) plays a critical role in the cardiac mitochondria function. Ca2+ entering the mitochondria is necessary for ATP production and the contractile activity of cardiomyocytes. However, excessive Ca2+ in the mitochondria results in mitochondrial dysfunction and cell death. Mitochondria maintain Ca2+ homeostasis in normal cardiomyocytes through a comprehensive regulatory mechanism by controlling the uptake and release of Ca2+ in response to the cellular demand. Understanding the mechanism of modulating mitochondrial Ca2+ homeostasis in the cardiomyocyte could bring new insights into the pathogenesis of cardiac disease and help developing the strategy to prevent the heart from damage at an early stage. In this review, we summarized the latest findings in the studies on the cardiac mitochondrial Ca2+ homeostasis, focusing on the regulation of mitochondrial calcium uptake, which acts as a double-edged sword in the cardiac function. Specifically, we discussed the dual roles of mitochondrial Ca2+ in mitochondrial activity and the impact on cardiac function, the molecular basis and regulatory mechanisms, and the potential future research interest.
Collapse
|
20
|
Mitochondrial Ca 2+ regulation in the etiology of heart failure: physiological and pathophysiological implications. Acta Pharmacol Sin 2020; 41:1301-1309. [PMID: 32694759 PMCID: PMC7608470 DOI: 10.1038/s41401-020-0476-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) represents one of the leading causes of cardiovascular diseases with high rates of hospitalization, morbidity and mortality worldwide. Ample evidence has consolidated a crucial role for mitochondrial injury in the progression of HF. It is well established that mitochondrial Ca2+ participates in the regulation of a wide variety of biological processes, including oxidative phosphorylation, ATP synthesis, reactive oxygen species (ROS) generation, mitochondrial dynamics and mitophagy. Nonetheless, mitochondrial Ca2+ overload stimulates mitochondrial permeability transition pore (mPTP) opening and mitochondrial swelling, resulting in mitochondrial injury, apoptosis, cardiac remodeling, and ultimately development of HF. Moreover, mitochondria possess a series of Ca2+ transport influx and efflux channels, to buffer Ca2+ in the cytoplasm. Interaction at mitochondria-associated endoplasmic reticulum membranes (MAMs) may also participate in the regulation of mitochondrial Ca2+ homeostasis and plays an essential role in the progression of HF. Here, we provide an overview of regulation of mitochondrial Ca2+ homeostasis in maintenance of cardiac function, in an effort to identify novel therapeutic strategies for the management of HF.
Collapse
|
21
|
Oropeza-Almazán Y, Blatter LA. Mitochondrial calcium uniporter complex activation protects against calcium alternans in atrial myocytes. Am J Physiol Heart Circ Physiol 2020; 319:H873-H881. [PMID: 32857593 DOI: 10.1152/ajpheart.00375.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cardiac alternans, defined as beat-to-beat alternations in action potential duration, cytosolic Ca transient (CaT) amplitude, and cardiac contraction is associated with atrial fibrillation (AF) and sudden cardiac death. At the cellular level, cardiac alternans is linked to abnormal intracellular calcium handling during excitation-contraction coupling. We investigated how pharmacological activation or inhibition of cytosolic Ca sequestration via mitochondrial Ca uptake and mitochondrial Ca retention affects the occurrence of pacing-induced CaT alternans in isolated rabbit atrial myocytes. Cytosolic CaTs were recorded using Fluo-4 fluorescence microscopy. Alternans was quantified as the alternans ratio (AR = 1 - CaTsmall/CaTlarge, where CaTsmall and CaTlarge are the amplitudes of the small and large CaTs of a pair of alternating CaTs). Inhibition of mitochondrial Ca sequestration via mitochondrial Ca uniporter complex (MCUC) with Ru360 enhanced the severity of CaT alternans (AR increase) and lowered the pacing frequency threshold for alternans. In contrast, stimulation of MCUC mediated mitochondrial Ca uptake with spermine-rescued alternans (AR decrease) and increased the alternans pacing threshold. Direct measurement of mitochondrial [Ca] in membrane permeabilized myocytes with Fluo-4 loaded mitochondria revealed that spermine enhanced and accelerated mitochondrial Ca uptake. Stimulation of mitochondrial Ca retention by preventing mitochondrial Ca efflux through the mitochondrial permeability transition pore with cyclosporin A also protected from alternans and increased the alternans pacing threshold. Pharmacological manipulation of MCUC activity did not affect sarcoplasmic reticulum Ca load. Our results suggest that activation of Ca sequestration by mitochondria protects from CaT alternans and could be a potential therapeutic target for cardiac alternans and AF prevention.NEW & NOTEWORTHY This study provides conclusive evidence that mitochondrial Ca uptake and retention protects from Ca alternans, whereas uptake inhibition enhances Ca alternans. The data suggest pharmacological mitochondrial Ca cycling modulation as a potential therapeutic strategy for alternans-related cardiac arrhythmia prevention.
Collapse
Affiliation(s)
| | - Lothar A Blatter
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
22
|
Marin W. A-kinase anchoring protein 1 (AKAP1) and its role in some cardiovascular diseases. J Mol Cell Cardiol 2019; 138:99-109. [PMID: 31783032 DOI: 10.1016/j.yjmcc.2019.11.154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 01/09/2023]
Abstract
A-kinase anchoring proteins (AKAPs) play crucial roles in regulating compartmentalized multi-protein signaling networks related to PKA-mediated phosphorylation. The mitochondrial AKAP - AKAP1 proteins are enriched in heart and play cardiac protective roles. This review aims to thoroughly summarize AKAP1 variants from their sequence features to the structure-function relationships between AKAP1 and its binding partners, as well as the molecular mechanisms of AKAP1 in cardiac hypertrophy, hypoxia-induced myocardial infarction and endothelial cells dysfunction, suggesting AKAP1 as a candidate for cardiovascular therapy.
Collapse
Affiliation(s)
- Wenwen Marin
- Institute for Translational Medicine, Medical Faculty of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
23
|
Tarasova NV, Vishnyakova PA, Logashina YA, Elchaninov AV. Mitochondrial Calcium Uniporter Structure and Function in Different Types of Muscle Tissues in Health and Disease. Int J Mol Sci 2019; 20:4823. [PMID: 31569359 PMCID: PMC6801532 DOI: 10.3390/ijms20194823] [Citation(s) in RCA: 12] [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: 08/14/2019] [Revised: 09/12/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023] Open
Abstract
Calcium ions (Ca2+) influx to mitochondrial matrix is crucial for the life of a cell. Mitochondrial calcium uniporter (mtCU) is a protein complex which consists of the pore-forming subunit (MCU) and several regulatory subunits. MtCU is the main contributor to inward Ca2+ currents through the inner mitochondrial membrane. Extensive investigations of mtCU involvement into normal and pathological molecular pathways started from the moment of discovery of its molecular components. A crucial role of mtCU in the control of these pathways is now recognized in both health and disease. In particular, impairments of mtCU function have been demonstrated for cardiovascular and skeletal muscle-associated pathologies. This review summarizes the current state of knowledge on mtCU structure, regulation, and function in different types of muscle tissues in health and disease.
Collapse
Affiliation(s)
- Nadezhda V Tarasova
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya str. 8, bld. 2, Moscow 119991, Russia.
| | - Polina A Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, Moscow 117997, Russia.
| | - Yulia A Logashina
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya str. 8, bld. 2, Moscow 119991, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street 16/10, Moscow 117997, Russia.
| | - Andrey V Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, Moscow 117997, Russia.
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia.
- Peoples' Friendship University of Russia, 6 Miklukho-Maklaya Street, Moscow 117198, Russia.
| |
Collapse
|
24
|
Protective Effects of Euthyroidism Restoration on Mitochondria Function and Quality Control in Cardiac Pathophysiology. Int J Mol Sci 2019; 20:ijms20143377. [PMID: 31295805 PMCID: PMC6678270 DOI: 10.3390/ijms20143377] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunctions are major contributors to heart disease onset and progression. Under ischemic injuries or cardiac overload, mitochondrial-derived oxidative stress, Ca2+ dis-homeostasis, and inflammation initiate cross-talking vicious cycles leading to defects of mitochondrial DNA, lipids, and proteins, concurrently resulting in fatal energy crisis and cell loss. Blunting such noxious stimuli and preserving mitochondrial homeostasis are essential to cell survival. In this context, mitochondrial quality control (MQC) represents an expanding research topic and therapeutic target in the field of cardiac physiology. MQC is a multi-tier surveillance system operating at the protein, organelle, and cell level to repair or eliminate damaged mitochondrial components and replace them by biogenesis. Novel evidence highlights the critical role of thyroid hormones (TH) in regulating multiple aspects of MQC, resulting in increased organelle turnover, improved mitochondrial bioenergetics, and the retention of cell function. In the present review, these emerging protective effects are discussed in the context of cardiac ischemia-reperfusion (IR) and heart failure, focusing on MQC as a strategy to blunt the propagation of connected dangerous signaling cascades and limit adverse remodeling. A better understanding of such TH-dependent signaling could provide insights into the development of mitochondria-targeted treatments in patients with cardiac disease.
Collapse
|
25
|
Adaniya SM, O-Uchi J, Cypress MW, Kusakari Y, Jhun BS. Posttranslational modifications of mitochondrial fission and fusion proteins in cardiac physiology and pathophysiology. Am J Physiol Cell Physiol 2019; 316:C583-C604. [PMID: 30758993 DOI: 10.1152/ajpcell.00523.2018] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial fragmentation frequently occurs in chronic pathological conditions as seen in various human diseases. In fact, abnormal mitochondrial morphology and mitochondrial dysfunction are hallmarks of heart failure (HF) in both human patients and HF animal models. A link between mitochondrial fragmentation and cardiac pathologies has been widely proposed, but the physiological relevance of mitochondrial fission and fusion in the heart is still unclear. Recent studies have increasingly shown that posttranslational modifications (PTMs) of fission and fusion proteins are capable of directly modulating the stability, localization, and/or activity of these proteins. These PTMs include phosphorylation, acetylation, ubiquitination, conjugation of small ubiquitin-like modifier proteins, O-linked-N-acetyl-glucosamine glycosylation, and proteolysis. Thus, understanding the PTMs of fission and fusion proteins may allow us to understand the complexities that determine the balance of mitochondrial fission and fusion as well as mitochondrial function in various cell types and organs including cardiomyocytes and the heart. In this review, we summarize present knowledge regarding the function and regulation of mitochondrial fission and fusion in cardiomyocytes, specifically focusing on the PTMs of each mitochondrial fission/fusion protein. We also discuss the molecular mechanisms underlying abnormal mitochondrial morphology in HF and their contributions to the development of cardiac diseases, highlighting the crucial roles of PTMs of mitochondrial fission and fusion proteins. Finally, we discuss the future potential of manipulating PTMs of fission and fusion proteins as a therapeutic strategy for preventing and/or treating HF.
Collapse
Affiliation(s)
- Stephanie M Adaniya
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota.,Cardiovascular Research Center, Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University , Providence, Rhode Island
| | - Jin O-Uchi
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Michael W Cypress
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine , Tokyo , Japan
| | - Bong Sook Jhun
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|