1
|
Vázquez-Carrada M, Vilchis-Landeros MM, Vázquez-Meza H, Uribe-Ramírez D, Matuz-Mares D. A New Perspective on the Role of Alterations in Mitochondrial Proteins Involved in ATP Synthesis and Mobilization in Cardiomyopathies. Int J Mol Sci 2025; 26:2768. [PMID: 40141413 PMCID: PMC11943459 DOI: 10.3390/ijms26062768] [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: 01/11/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The heart requires a continuous energy supply to sustain its unceasing contraction-relaxation cycle. Mitochondria, a double-membrane organelle, generate approximately 90% of cellular energy as adenosine triphosphate (ATP) through oxidative phosphorylation, utilizing the electrochemical gradient established by the respiratory chain. Mitochondrial function is compromised by damage to mitochondrial DNA, including point mutations, deletions, duplications, or inversions. Additionally, disruptions to proteins associated with mitochondrial membranes regulating metabolic homeostasis can impair the respiratory chain's efficiency. This results in diminished ATP production and increased generation of reactive oxygen species. This review provides an overview of mutations affecting mitochondrial transporters and proteins involved in mitochondrial energy synthesis, particularly those involved in ATP synthesis and mobilization, and it examines their role in the pathogenesis of specific cardiomyopathies.
Collapse
Affiliation(s)
- Melissa Vázquez-Carrada
- Institute of Microbiology, Cluster of Excellence on Plant Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - María Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Héctor Vázquez-Meza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Daniel Uribe-Ramírez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Av, Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, Ciudad de México C.P. 07738, Mexico;
| | - Deyamira Matuz-Mares
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| |
Collapse
|
2
|
Peng T, Xiang J, Tian Y, Tang X, Wang L, Gao L, Luo OJ, Huang L, Chen G. Lycium barbarum glycopeptide ameliorates aging phenotypes and enhances cardiac metabolism by activating the PINK1/Parkin-mediated mitophagy pathway in D-galactose-induced mice. Exp Gerontol 2025; 200:112686. [PMID: 39827719 DOI: 10.1016/j.exger.2025.112686] [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: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Aging is a complex biological process that disrupts tissue structure and impairs physiological function, which contributes to the development of age-related diseases such as cardiovascular disorders. However, effective treatment strategies are lacking. OBJECTIVE To investigate the geroprotective effects of Lycium barbarum glycopeptide (LbGp) and its potential mechanisms in a D-galactose-induced accelerated aging mouse model. METHODS Mice were subcutaneously injected with D-galactose (500 mg/kg/day) for 12 weeks to induce aging, while LbGp was orally administered (100 mg/kg/day) throughout the study. The geroprotective effects of LbGp were assessed by behavioral tests, cardiac echocardiography, pathohistological and transcriptomic analyses. Transmission electron microscopy was used to observe the ultrastructure of mitochondria. Mitochondrial stress assays and JC-1 fluorescent probe were conducted to evaluate mitochondrial function. Flow cytometer and western blot were performed to assess mitophagy flux. RESULTS LbGp treatment improved the aging phenotypes of D-galactose-induced mice, with a pronounced enhancement in cardiac function compared to neurocognitive and skeletal muscle functions. Transcriptome analysis indicated that LbGp ameliorated energy metabolism in the heart. Mitochondrial assays revealed LbGp improved mitochondrial function and preserved structural integrity of the mitochondrial inner membrane. LbGp attenuated mitochondrial fission and restored impaired PINK1/Parkin-mediated mitophagy pathway caused by D-galactose in cardiomyocytes. CONCLUSION LbGp can ameliorate aging phenotypes and enhance cardiac metabolism by activating the PINK1/Parkin-mediated mitophagy pathway in D-galactose-induced mice. These findings underscore its potential as a therapeutic agent for aging and aging-related cardiovascular diseases.
Collapse
Affiliation(s)
- Tianchan Peng
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jian Xiang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yun Tian
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaogen Tang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lina Wang
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China
| | - Oscar Junhong Luo
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China
| | - Li'an Huang
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China.
| |
Collapse
|
3
|
Sahu Y, Jamadade P, Ch Maharana K, Singh S. Role of mitochondrial homeostasis in D-galactose-induced cardiovascular ageing from bench to bedside. Mitochondrion 2024; 78:101923. [PMID: 38925493 DOI: 10.1016/j.mito.2024.101923] [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: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Ageing is an inevitable phenomenon which affects the cellular to the organism level in the progression of the time. Oxidative stress and inflammation are now widely regarded as the key processes involved in the aging process, which may then cause significant harm to mitochondrial DNA, leading to apoptosis. Normal circulatory function is a significant predictor of disease-free life expectancy. Indeed, disorders affecting the cardiovascular system, which are becoming more common, are the primary cause of worldwide morbidity, disability, and mortality. Cardiovascular aging may precede or possibly underpin overall, age-related health decline. Numerous studies have foundmitochondrial mechanistc approachplays a vital role in the in the onset and development of aging. The D-galactose (D-gal)-induced aging model is well recognized and commonly used in the aging study. In this review we redeposit the association of the previous and current studies on mitochondrial homeostasis and its underlying mechanisms in D-galactose cardiovascular ageing. Further we focus the novel and the treatment strategies to combat the major complication leading to the cardiovascular ageing.
Collapse
Affiliation(s)
- Yogita Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Pratiksha Jamadade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Krushna Ch Maharana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India.
| |
Collapse
|
4
|
Hinton A, Claypool SM, Neikirk K, Senoo N, Wanjalla CN, Kirabo A, Williams CR. Mitochondrial Structure and Function in Human Heart Failure. Circ Res 2024; 135:372-396. [PMID: 38963864 PMCID: PMC11225798 DOI: 10.1161/circresaha.124.323800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Steven M. Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Nanami Senoo
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH (C.R.W.)
| |
Collapse
|
5
|
Zhang Y, Du X, Zhao L, Sun Y. Construction of dose prediction model and identification of sensitive genes for space radiation based on single-sample networks under spaceflight conditions. Int J Radiat Biol 2024; 100:777-790. [PMID: 38471034 DOI: 10.1080/09553002.2024.2327393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE To identify sensitive genes for space radiation, we integrated the transcriptomic samples of spaceflight mice from GeneLab and predicted the radiation doses absorbed by individuals in space. METHODS AND MATERIALS A single-sample network (SSN) for each individual sample was constructed. Then, using machine learning and genetic algorithms, we built the regression models to predict the absorbed dose equivalent based on the topological structure of SSNs. Moreover, we analyzed the SSNs from each tissue and compared the similarities and differences among them. RESULTS Our model exhibited excellent performance with the following metrics: R 2 = 0.980 , MSE = 6.74 e - 04 , and the Pearson correlation coefficient of 0.990 (p value <.0001) between predicted and actual values. We identified 20 key genes, the majority of which had been proven to be associated with radiation. However, we uniquely established them as space radiation sensitive genes for the first time. Through further analysis of the SSNs, we discovered that the different tissues exhibited distinct mechanisms in response to space stressors. CONCLUSIONS The topology structures of SSNs effectively predicted radiation doses under spaceflight conditions, and the SSNs revealed the gene regulatory patterns within the organisms under space stressors.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Xiaohui Du
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Lei Zhao
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Yeqing Sun
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| |
Collapse
|
6
|
Fujii K, Fujiwara-Tani R, Nukaga S, Ohmori H, Luo Y, Nishida R, Sasaki T, Miyagawa Y, Nakashima C, Kawahara I, Ogata R, Ikemoto A, Sasaki R, Kuniyasu H. Involvement of Ferroptosis Induction and Oxidative Phosphorylation Inhibition in the Anticancer-Drug-Induced Myocardial Injury: Ameliorative Role of Pterostilbene. Int J Mol Sci 2024; 25:3015. [PMID: 38474261 DOI: 10.3390/ijms25053015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 02/25/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
Patients with cancer die from cardiac dysfunction second only to the disease itself. Cardiotoxicity caused by anticancer drugs has been emphasized as a possible cause; however, the details remain unclear. To investigate this mechanism, we treated rat cardiomyoblast H9c2 cells with sunitinib, lapatinib, 5-fluorouracil, and cisplatin to examine their effects. All anticancer drugs increased ROS, lipid peroxide, and iron (II) levels in the mitochondria and decreased glutathione peroxidase-4 levels and the GSH/GSSG ratio. Against this background, mitochondrial iron (II) accumulates through the unregulated expression of haem oxygenase-1 and ferrochelatase. Anticancer-drug-induced cell death was suppressed by N-acetylcysteine, deferoxamine, and ferrostatin, indicating ferroptosis. Anticancer drug treatment impairs mitochondrial DNA and inhibits oxidative phosphorylation in H9c2 cells. Similar results were observed in the hearts of cancer-free rats treated with anticancer drugs in vitro. In contrast, treatment with pterostilbene inhibited the induction of ferroptosis and rescued the energy restriction induced by anticancer drugs both in vitro and in vivo. These findings suggest that induction of ferroptosis and inhibition of oxidative phosphorylation are mechanisms by which anticancer drugs cause myocardial damage. As pterostilbene ameliorates these mechanisms, it is expected to have significant clinical applications.
Collapse
Grants
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 21K06926 Ministry of Education, Culture, Sports, Science and Technology
- 21K11223 Ministry of Education, Culture, Sports, Science and Technology
- 22K11423 Ministry of Education, Culture, Sports, Science and Technology
- 23K16547 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Shota Nukaga
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Yi Luo
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ryoichi Nishida
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Yoshihiro Miyagawa
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Chie Nakashima
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ayaka Ikemoto
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Rika Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| |
Collapse
|
7
|
Gallo G, Rubattu S, Volpe M. Mitochondrial Dysfunction in Heart Failure: From Pathophysiological Mechanisms to Therapeutic Opportunities. Int J Mol Sci 2024; 25:2667. [PMID: 38473911 DOI: 10.3390/ijms25052667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/17/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Mitochondrial dysfunction, a feature of heart failure, leads to a progressive decline in bioenergetic reserve capacity, consisting in a shift of energy production from mitochondrial fatty acid oxidation to glycolytic pathways. This adaptive process of cardiomyocytes does not represent an effective strategy to increase the energy supply and to restore the energy homeostasis in heart failure, thus contributing to a vicious circle and to disease progression. The increased oxidative stress causes cardiomyocyte apoptosis, dysregulation of calcium homeostasis, damage of proteins and lipids, leakage of mitochondrial DNA, and inflammatory responses, finally stimulating different signaling pathways which lead to cardiac remodeling and failure. Furthermore, the parallel neurohormonal dysregulation with angiotensin II, endothelin-1, and sympatho-adrenergic overactivation, which occurs in heart failure, stimulates ventricular cardiomyocyte hypertrophy and aggravates the cellular damage. In this review, we will discuss the pathophysiological mechanisms related to mitochondrial dysfunction, which are mainly dependent on increased oxidative stress and perturbation of the dynamics of membrane potential and are associated with heart failure development and progression. We will also provide an overview of the potential implication of mitochondria as an attractive therapeutic target in the management and recovery process in heart failure.
Collapse
Affiliation(s)
- Giovanna Gallo
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, 00189 Rome, RM, Italy
| | - Speranza Rubattu
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, 00189 Rome, RM, Italy
- IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | | |
Collapse
|
8
|
Wang X, Wang B, Yang F, Shang K, Chen S, Zhang Y. Associations between plasma metal elements and risk of cognitive impairment among Chinese older adults. Front Aging Neurosci 2024; 16:1353286. [PMID: 38384934 PMCID: PMC10879289 DOI: 10.3389/fnagi.2024.1353286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/29/2024] [Indexed: 02/23/2024] Open
Abstract
Background The relationship between plasma metal elements and cognitive function is unclear, especially in extremely older individuals. This present study aimed to explore the association between plasma metal concentrations and the risk of cognitive impairment (CI) in Chinese extremely older adults. Methods Individuals aged ≥90 years with plasm metal concentration data from the fifth wave of the 2008 Chinese Longitudinal Healthy Longevity Survey were included. Plasma selenium (Se), manganese (Mn), magnesium (Mg), calcium (Ca), iron (Fe), copper (Cu), and zinc (Zn) concentrations were measured using inductively coupled plasma optical emission spectroscopy. Cognitive function was assessed by the Chinese version of the mini-mental state examination. Results The study enrolled 408 participants. Participants with CI had significantly lower plasma Se, Mn, and Fe levels and higher Ca levels than those with normal cognitive function (p < 0.05). Plasma Se, Mn, Ca, and Fe concentrations were significantly associated with CI risk in both single- and multiple-element logistic regression models. Additionally, the multiple-element model results showed that the adjusted odds ratios for CI were 0.042 (95% confidence interval 0.016-0.109), 0.106 (0.044-0.255), 7.629 (3.211-18.124) and 0.092 (0.036-0.233) for the highest quartiles compared to the lowest quartiles of Se, Mn, Ca, and Fe, respectively. Moreover, subgroup analyses by age, sex, and body mass index suggested a consistent significant correlation (p < 0.05). Conclusion Therefore, decreased plasma Se, Mn, and Fe and increased plasma Ca levels were associated with CI risk in Chinese older adults. These findings are of great significance for the development of programs to delay cognitive decline in the elderly.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Binbin Wang
- School of Life Science, Shanxi Normal University, Taiyuan, China
| | - Fuwen Yang
- Department of Neurology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaijian Shang
- Department of Emergency Medicine, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shaowei Chen
- Department of Hematology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yue Zhang
- School of Public Health, Department of Epidemiology, Shanxi Medical University, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry Education, Taiyuan, China
| |
Collapse
|
9
|
Liu W, Mossel P, Schwach V, Slart RHJA, Luurtsema G. Cardiac PET Imaging of ATP Binding Cassette (ABC) Transporters: Opportunities and Challenges. Pharmaceuticals (Basel) 2023; 16:1715. [PMID: 38139840 PMCID: PMC10748140 DOI: 10.3390/ph16121715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Adenosine triphosphate binding cassette (ABC) transporters are a broad family of membrane protein complexes that use energy to transport molecules across cells and/or intracellular organelle lipid membranes. Many drugs used to treat cardiac diseases have an affinity for these transporters. Among others, P-glycoprotein (P-gp) plays an essential role in regulating drug concentrations that reach cardiac tissue and therefore contribute to cardiotoxicity. As a molecular imaging modality, positron emission tomography (PET) has emerged as a viable technique to investigate the function of P-gp in organs and tissues. Using PET imaging to evaluate cardiac P-gp function provides new insights for drug development and improves the precise use of medications. Nevertheless, information in this field is limited. In this review, we aim to examine the current applications of ABC transporter PET imaging and its tracers in the heart, with a specific emphasis on P-gp. Furthermore, the opportunities and challenges in this novel field will be discussed.
Collapse
Affiliation(s)
- Wanling Liu
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
| | - Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherlands;
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
- Department of Biomedical Photonic Imaging, University of Twente, 7500 AE Enschede, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
| |
Collapse
|
10
|
Gupta T, Najumuddin, Rajendran D, Gujral A, Jangra A. Metabolism configures immune response across multi-systems: Lessons from COVID-19. Adv Biol Regul 2023; 90:100977. [PMID: 37690286 DOI: 10.1016/j.jbior.2023.100977] [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: 03/10/2023] [Revised: 07/19/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023]
Abstract
Several studies over the last decade demonstrate the recruitment of immune cells, increased inflammatory cytokines, and chemokine in patients with metabolic diseases, including heart failure, parenchymal inflammation, obesity, tuberculosis, and diabetes mellitus. Metabolic rewiring of immune cells is associated with the severity and prevalence of these diseases. The risk of developing COVID-19/SARS-CoV-2 infection increases in patients with metabolic dysfunction (heart failure, diabetes mellitus, and obesity). Several etiologies, including fatigue, dyspnea, and dizziness, persist even months after COVID-19 infection, commonly known as Post-Acute Sequelae of CoV-2 (PASC) or long COVID. A chronic inflammatory state and metabolic dysfunction are the factors that contribute to long COVID. Here, this study explores the potential link between pathogenic metabolic and immune alterations across different organ systems that could underlie COVID-19 and PASC. These interactions could be utilized for targeted future therapeutic approaches.
Collapse
Affiliation(s)
- Tinku Gupta
- Department of Pharmacognosy & Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed University), M. B. Road, New Delhi 110062, India
| | - Najumuddin
- Program of Biotechnology, Department of Applied Sciences, Faculty of Engineering, Science and Technology, Hamdard University, Karachi, Pakistan
| | - Dhanya Rajendran
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Akash Gujral
- Department of Medicine, Nyu Grossman School of Medicine, NY, USA
| | - Ashok Jangra
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India.
| |
Collapse
|
11
|
Du J, Yu D, Li J, Si L, Zhu D, Li B, Gao Y, Sun L, Wang X, Wang X. Asiatic acid protects against pressure overload-induced heart failure in mice by inhibiting mitochondria-dependent apoptosis. Free Radic Biol Med 2023; 208:545-554. [PMID: 37717794 DOI: 10.1016/j.freeradbiomed.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Mitochondrial dysfunction and subsequent cardiomyocyte apoptosis significantly contribute to pressure overload-induced heart failure (HF). A highly oxidative environment leads to mitochondrial damage, further exacerbating this condition. Asiatic acid (AA), a proven antioxidant and anti-hypertrophic agent, might provide a solution, but its role and mechanisms in chronic pressure overload-induced HF remain largely unexplored. METHODS We induced pressure overload in mice using transverse aortic constriction (TAC) and treated them with AA (100 mg/kg/day) or vehicle daily by oral gavage for 8 weeks. The effects of AA on mitochondrial dysfunction, oxidative stress-associated signaling pathways, and overall survival were evaluated. Additionally, an in vitro model using hydrogen peroxide-exposed neonatal rat cardiomyocytes was established to further investigate the role of AA in oxidative stress-induced mitochondrial apoptosis. RESULTS AA treatment significantly improved survival and alleviated cardiac dysfunction in TAC-induced HF mice. It preserved mitochondrial structure, reduced the LVW/BW ratio by 20.24%, mitigated TAC-induced mitochondrial-dependent apoptosis by significantly lowering the Bax/Bcl-2 ratio and cleaved caspase-9/3 levels, and attenuated oxidative stress. AA treatment protected cardiomyocytes from hydrogen peroxide-induced apoptosis, with concurrent modulation of mitochondrial-dependent apoptosis pathway-related proteins and the JNK pathway. CONCLUSIONS Our findings suggest that AA effectively combats chronic TAC-induced and hydrogen peroxide-induced cardiomyocyte apoptosis through a mitochondria-dependent mechanism. AA reduces cellular levels of oxidative stress and inhibits the activation of the JNK pathway, highlighting its potential therapeutic value in the treatment of HF.
Collapse
Affiliation(s)
- Junjie Du
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Dongmin Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jinghang Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Linjie Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Dawei Zhu
- Department of Cardiothoracic Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211000, China
| | - Ben Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yizhou Gao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lifu Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xufeng Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
12
|
Gorący A, Rosik J, Szostak J, Szostak B, Retfiński S, Machaj F, Pawlik A. Improving mitochondrial function in preclinical models of heart failure: therapeutic targets for future clinical therapies? Expert Opin Ther Targets 2023; 27:593-608. [PMID: 37477241 DOI: 10.1080/14728222.2023.2240021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/19/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Heart failure is a complex clinical syndrome resulting from the unsuccessful compensation of symptoms of myocardial damage. Mitochondrial dysfunction is a process that occurs because of an attempt to adapt to the disruption of metabolic and energetic pathways occurring in the myocardium. This, in turn, leads to further dysfunction in cardiomyocyte processes. Currently, many therapeutic strategies have been implemented to improve mitochondrial function, but their effectiveness varies widely. AREAS COVERED This review focuses on new models of therapeutic strategies targeting mitochondrial function in the treatment of heart failure. EXPERT OPINION Therapeutic strategies targeting mitochondria appear to be a valuable option for treating heart failure. Currently, the greatest challenge is to develop new research models that could restore the disrupted metabolic processes in mitochondria as comprehensively as possible. Only the development of therapies that focus on improving as many dysregulated mitochondrial processes as possible in patients with heart failure will be able to bring the expected clinical improvement, along with inhibition of disease progression. Combined strategies involving the reduction of the effects of oxidative stress and mitochondrial dysfunction, appear to be a promising possibility for developing new therapies for a complex and multifactorial disease such as heart failure.
Collapse
Affiliation(s)
- Anna Gorący
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Joanna Szostak
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Szymon Retfiński
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Filip Machaj
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
- Department of Medical Biology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
13
|
Seydi E, Hassani MK, Naderpour S, Arjmand A, Pourahmad J. Cardiotoxicity of chloroquine and hydroxychloroquine through mitochondrial pathway. BMC Pharmacol Toxicol 2023; 24:26. [PMID: 37085872 PMCID: PMC10119838 DOI: 10.1186/s40360-023-00666-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/27/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Medical therapies can cause cardiotoxicity. Chloroquine (QC) and hydroxychloroquine (HQC) are drugs used in the treatment of malaria and skin and rheumatic disorders. These drugs were considered to help treatment of coronavirus disease (COVID-19) in 2019. Despite the low cost and availability of QC and HQC, reports indicate that this class of drugs can cause cardiotoxicity. The mechanism of this event is not well known, but evidence shows that QC and HQC can cause cardiotoxicity by affecting mitochondria and lysosomes. METHODS Therefore, our study was designed to investigate the effects of QC and HQC on heart mitochondria. In order to achieve this aim, mitochondrial function, reactive oxygen species (ROS) level, mitochondrial membrane disruption, and cytochrome c release in heart mitochondria were evaluated. Statistical significance was determined using the one-way and two-way analysis of variance (ANOVA) followed by post hoc Tukey to evaluate mitochondrial succinate dehydrogenase (SDH) activity and cytochrome c release, and Bonferroni test to evaluate the ROS level, mitochondrial membrane potential (MMP) collapse, and mitochondrial swelling. RESULTS Based on ANOVA analysis (one-way), the results of mitochondrial SDH activity showed that the IC50 concentration for CQ is 20 µM and for HCQ is 50 µM. Based on two-way ANOVA analysis, the highest effect of CQ and HCQ on the generation of ROS, collapse in the MMP, and mitochondrial swelling were observed at 40 µM and 100 µM concentrations, respectively (p < 0.05). Also, the highest effect of these two drugs has been observed in 60 min (p < 0.05). The statistical results showed that compared to CQ, HCQ is able to cause the release of cytochrome c from mitochondria in all applied concentrations (p < 0.05). CONCLUSIONS The results suggest that QC and HQC can cause cardiotoxicity which can lead to heart disorders through oxidative stress and disfunction of heart mitochondria.
Collapse
Affiliation(s)
- Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran
| | - Mozhgan Karbalaei Hassani
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saghi Naderpour
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus, Cyprus
| | - Abdollah Arjmand
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Jalal Pourahmad
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Extracellular vesicles DJ-1 derived from hypoxia-conditioned hMSCs alleviate cardiac hypertrophy by suppressing mitochondria dysfunction and preventing ATRAP degradation. Pharmacol Res 2023; 187:106607. [PMID: 36509316 DOI: 10.1016/j.phrs.2022.106607] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND As a pathological myocardial remodeling process in a variety of cardiovascular diseases, cardiac hypertrophy still has no effective treatment. Human mesenchymal stem cells (hMSCs) derived extracellular vesicles (EVs) has been recognized as a promising treatment strategy for cardiac disease. METHODS In this study, the inhibitory effects on cardiac hypertrophy are compared between normoxia-conditioned hMSC-derived EVs (Nor-EVs) and hypoxia-conditioned hMSC-derived EVs (Hypo-EVs) in neonatal rat cardiomyocytes (NRCMs) after angiotensin II (Ang II) stimulation and in a mouse model of transverse aortic constriction (TAC). RESULTS We demonstrate that Hypo-EVs exert an increased inhibitory effect on cardiac hypertrophy compared with Nor-EVs. Parkinson disease protein 7 (PARK7/DJ-1) is identify as a differential protein between Nor-EVs and Hypo-EVs by quantitative proteomics analysis. Results show that DJ-1, which is rich in Hypo-EVs, alleviates mitochondrial dysfunction and excessive mitochondrial reactive oxygen species (mtROS) production as an antioxidant. Mechanistic studies demonstrate for the first time that DJ-1 may suppress cardiac hypertrophy by inhibiting the activity of proteasome subunit beta type 10 (PSMB10) through a direct physical interaction. This interaction can inhibit angiotensin II type 1 receptor (AT1R)-mediated signaling pathways resulting in cardiac hypertrophy through alleviating ubiquitination degradation of AT1R-associated protein (ATRAP). CONCLUSIONS When taken together, our study suggests that Hypo-EVs have significant potential as a novel therapeutic agent for the treatment of cardiac hypertrophy.
Collapse
|
15
|
Huang X, Zeng Z, Li S, Xie Y, Tong X. The Therapeutic Strategies Targeting Mitochondrial Metabolism in Cardiovascular Disease. Pharmaceutics 2022; 14:pharmaceutics14122760. [PMID: 36559254 PMCID: PMC9788260 DOI: 10.3390/pharmaceutics14122760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is a group of systemic disorders threatening human health with complex pathogenesis, among which mitochondrial energy metabolism reprogramming has a critical role. Mitochondria are cell organelles that fuel the energy essential for biochemical reactions and maintain normal physiological functions of the body. Mitochondrial metabolic disorders are extensively involved in the progression of CVD, especially for energy-demanding organs such as the heart. Therefore, elucidating the role of mitochondrial metabolism in the progression of CVD is of great significance to further understand the pathogenesis of CVD and explore preventive and therapeutic methods. In this review, we discuss the major factors of mitochondrial metabolism and their potential roles in the prevention and treatment of CVD. The current application of mitochondria-targeted therapeutic agents in the treatment of CVD and advances in mitochondria-targeted gene therapy technologies are also overviewed.
Collapse
Affiliation(s)
- Xiaoyang Huang
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zhenhua Zeng
- Biomedical Research Center, Hunan University of Medicine, Huaihua 418000, China
| | - Siqi Li
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Yufei Xie
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyong Tong
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
- Correspondence:
| |
Collapse
|
16
|
Fedotcheva T, Shimanovsky N, Fedotcheva N. Involvement of Multidrug Resistance Modulators in the Regulation of the Mitochondrial Permeability Transition Pore. MEMBRANES 2022; 12:membranes12090890. [PMID: 36135908 PMCID: PMC9502193 DOI: 10.3390/membranes12090890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 05/12/2023]
Abstract
The permeability transition pore in mitochondria (MPTP) and the ATP-binding cassette transporters (АВС transporters) in cell membranes provide the efflux of low-molecular compounds across mitochondrial and cell membranes, respectively. The inhibition of ABC transporters, especially of those related to multi drug resistance (MDR) proteins, is an actively explored approach to enhance intracellular drug accumulation and increase thereby the efficiency of anticancer therapy. Although there is evidence showing the simultaneous effect of some inhibitors on both MDR-related proteins and mitochondrial functions, their influence on MPTP has not been previously studied. We examined the participation of verapamil and quinidine, classified now as the first generation of MDR modulators, and avermectin, which has recently been actively studied as an MDR inhibitor, in the regulation of the MPTP opening. In experiments on rat liver mitochondria, we found that quinidine lowered and verapamil increased the threshold concentrations of calcium ions required for MPTP opening, and that they both decreased the rate of calcium-induced swelling of mitochondria. These effects may be associated with the positive charge of the drugs and their aliphatic properties. Avermectin not only decreased the threshold concentration of calcium ions, but also by itself induced the opening of MPTP and the mitochondrial swelling inhibited by ADP and activated by carboxyatractyloside, the substrate and inhibitor of adenine nucleotide translocase (ANT), which suggests the involvement of ANT in the process. Thus, these data indicate an additional opportunity to evaluate the effectiveness of MDR modulators in the context of their influence on the mitochondrial-dependent apoptosis.
Collapse
Affiliation(s)
- Tatiana Fedotcheva
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, Moscow 117997, Russia
| | - Nikolai Shimanovsky
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, Moscow 117997, Russia
| | - Nadezhda Fedotcheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya St. 3, Pushchino 142290, Russia
- Correspondence:
| |
Collapse
|
17
|
Protective Effect of Natural Medicinal Plants on Cardiomyocyte Injury in Heart Failure: Targeting the Dysregulation of Mitochondrial Homeostasis and Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3617086. [PMID: 36132224 PMCID: PMC9484955 DOI: 10.1155/2022/3617086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/16/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Heart failure occurs because of various cardiovascular pathologies, such as coronary artery disease or cardiorenal syndrome, eventually reaching end-stage disease. Various factors contribute to cardiac structural or functional changes that result in systolic or diastolic dysfunction. Several studies have confirmed that the key factor in heart failure progression is myocardial cell death, and mitophagy is the major mechanism regulating myocardial cell death in heart failure. The clinical mechanisms of heart failure are well understood in practice. However, the essential role of mitophagic regulation in heart failure has only recently received widespread attention. Receptor-mediated mitophagy is involved in various mitochondrial processes like oxidative stress injury, energy metabolism disorders, and calcium homeostasis, which are also the main causes of heart failure. Understanding of the diverse regulatory mechanisms in mitophagy and the complexity of its pathophysiology in heart failure remains incomplete. Related studies have found that various natural medicinal plants and active ingredients, such as flavonoids and saponins, can regulate mitophagy to a certain extent, improve myocardial function, and protect myocardial cells. This review comprehensively covers the relevant mechanisms of different types of mitophagy in regulating heart failure pathology and controlling mitochondrial adaptability to stress injury. Further, it explores the relationship between mitophagy and cardiac ejection dysfunction. Natural medicinal plant-targeted regulation strategies and scientific evidence on mitophagy were provided to elucidate current and potential strategies to apply mitophagy-targeted therapy for heart failure.
Collapse
|
18
|
Zou L, Li B, Xiong L, Wang Y, Xie W, Huang X, Liang Y, Wei T, Liu N, Chang X, Bai C, Wu T, Xue Y, Zhang T, Tang M. Urban fine particulate matter causes cardiac hypertrophy through calcium-mediated mitochondrial bioenergetics dysfunction in mice hearts and human cardiomyocytes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119236. [PMID: 35367502 DOI: 10.1016/j.envpol.2022.119236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
In recent years, the cardiovascular toxicity of urban fine particulate matter (PM2.5) has sparked significant alarm. Mitochondria produce 90% of ATP and make up 30% of the volume of cardiomyocytes. Thus knowledge of myocardial mitochondrial dysfunction due to PM2.5 exposure is essential for further cardiotoxic effects. Here, the mechanism of PM2.5-induced cardiac hypertrophy through calcium overload and mitochondrial dysfunction was investigated in vivo and in vitro. Male and female BALB/c mice were given 1.28, 5.5, and 11 mg PM2.5/kg bodyweight weekly through oropharyngeal inhalation for four weeks and were assigned to low, medium, and high dose groups, respectively. PM2.5-induced myocardial edema and cardiac hypertrophy were detected in the high-dose group. Mitochondria were scattered and ruptured with abnormal ultrastructural morphology. In vitro experiments on human cardiomyocyte AC16 showed that exposure to PM2.5 for 24 h caused opened mitochondrial permeability transition pore --leading to excessive calcium production, decreased mitochondrial membrane potential, weakened mitochondrial respiratory metabolism capacity, and decreased ATP production. Nevertheless, the administration of calcium chelator ameliorated the mitochondrial damage in the PM2.5-treated group. Our in vivo and in vitro results confirmed that calcium overload under PM2.5 exposure triggered mTOR/AKT/GSK-3β activation, leading to mitochondrial bioenergetics dysfunction and cardiac hypertrophy.
Collapse
Affiliation(s)
- Lingyue Zou
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Binjing Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Lilin Xiong
- Department of Environmental Health, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, 210003, China
| | - Yan Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Wenjing Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xiaoquan Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ying Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Tingting Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Na Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xiaoru Chang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Changcun Bai
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Tianshu Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
19
|
Mozihim AK, Chung I, Said NABM, Jamil AHA. Reprogramming of Fatty Acid Metabolism in Gynaecological Cancers: Is There a Role for Oestradiol? Metabolites 2022; 12:metabo12040350. [PMID: 35448537 PMCID: PMC9031151 DOI: 10.3390/metabo12040350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Gynaecological cancers are among the leading causes of cancer-related death among women worldwide. Cancer cells undergo metabolic reprogramming to sustain the production of energy and macromolecules required for cell growth, division and survival. Emerging evidence has provided significant insights into the integral role of fatty acids on tumourigenesis, but the metabolic role of high endogenous oestrogen levels and increased gynaecological cancer risks, notably in obesity, is less understood. This is becoming a renewed research interest, given the recently established association between obesity and incidence of many gynaecological cancers, including breast, ovarian, cervical and endometrial cancers. This review article, hence, comprehensively discusses how FA metabolism is altered in these gynaecological cancers, highlighting the emerging role of oestradiol on the actions of key regulatory enzymes of lipid metabolism, either directly through its classical ER pathways, or indirectly via the IGIFR pathway. Given the dramatic rise in obesity and parallel increase in the prevalence of gynaecological cancers among premenopausal women, further clarifications of the complex mechanisms underpinning gynaecological cancers are needed to inform future prevention efforts. Hence, in our review, we also highlight opportunities where metabolic dependencies can be exploited as viable therapeutic targets for these hormone-responsive cancers.
Collapse
Affiliation(s)
- Azilleo Kristo Mozihim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Nur Akmarina B. M. Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
| | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
- Correspondence: ; Tel.: +60-3-7967-4909
| |
Collapse
|
20
|
Karmi O, Rowland L, King SD, Manrique-Acevedo C, Cabantchik IZ, Nechushtai R, Mittler R. The [2Fe-2S] protein CISD2 plays a key role in preventing iron accumulation in cardiomyocytes. FEBS Lett 2022; 596:747-761. [PMID: 34997963 DOI: 10.1002/1873-3468.14277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/11/2021] [Accepted: 12/30/2021] [Indexed: 11/08/2022]
Abstract
Considered a key aging gene, CISD2, encoding CDGSH iron-sulfur domain-containing protein 2, plays a central role in regulating calcium homeostasis, preventing mitochondrial dysfunction, and the activation of autophagy and apoptosis in different cells. Here, we show that cardiomyocytes from CISD2-null mice accumulate high levels of iron and contain high levels of transferrin receptor and ferritin. Using proteomics and transmission electron microscopy, we further show that the lack of CISD2 induces several features of the aging process in young mice, but other features are not induced. Taken together, our findings suggest that CISD2 protects cardiomyocytes from overaccumulation of iron, which is common in aging hearts and can contribute to the pathogenesis of heart failure.
Collapse
Affiliation(s)
- Ola Karmi
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Israel
| | - Linda Rowland
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Skylar D King
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Ioav Z Cabantchik
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Israel
| | - Rachel Nechushtai
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Israel
| | - Ron Mittler
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- The Division of Plant Sciences and Interdisciplinary Plant Group, College of Agriculture, Food and Natural Resources, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
21
|
Wang HT, Ju J, Wang SC, Zhang YH, Liu CY, Wang T, Yu X, Wang F, Cheng XR, Wang K, Chen ZY. Insights Into Ferroptosis, a Novel Target for the Therapy of Cancer. Front Oncol 2022; 12:812534. [PMID: 35280796 PMCID: PMC8914339 DOI: 10.3389/fonc.2022.812534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 01/17/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death (PCD) characterized by an excess iron accumulation and subsequent unbalanced redox states. Ferroptosis is different from the already reported PCD and has unique morphological features and biochemical processes. Ferroptosis was first elaborated by Brent R. Stockwell’s lab in 2012, in which small molecules erastin and RSL-3 induce PCD in Ras mutant cell lines. Ferroptosis involves various physiological processes and occurrence of disease and especially shows strong potential in cancer treatment. Development of small molecule compounds based on Stockwell’s research was found to kill cancer cells, and some FDA-approved drugs were discovered to result in ferroptosis of cancer cells. Radiotherapy and checkpoint therapy have been widely used as a treatment for many types of cancer. Recently, some papers have reported that chemotherapy, radiotherapy, and checkpoint therapy induce ferroptosis of cancer cells, which provides new strategies for cancer treatment. Nevertheless, the limitless proliferation of tumor cells and the lack of cell death mechanisms are important reasons for drug resistance for tumor therapy. Therefore, we reviewed the molecular mechanism of ferroptosis and sensitivity to ferroptosis of different cancer cells and tumor treatment strategy.
Collapse
Affiliation(s)
- Hong-Tao Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- Science and Technology Department, Qingdao University, Qingdao, China
| | - Jie Ju
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Shao-Cong Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yu-Hui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cui-Yun Liu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Tao Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue Yu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Fei Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue-Ru Cheng
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- *Correspondence: Kun Wang, ; Zhao-Yang Chen,
| | - Zhao-Yang Chen
- Cardiology Department, Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, China
- *Correspondence: Kun Wang, ; Zhao-Yang Chen,
| |
Collapse
|
22
|
Jiang J, Wang L, Zhang C, Zhao X. Health risks of sulfentrazone exposure during zebrafish embryo-larvae development at environmental concentration. CHEMOSPHERE 2022; 288:132632. [PMID: 34687687 DOI: 10.1016/j.chemosphere.2021.132632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/05/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
Knowledge about the negative effects and mechanism of sulfentrazone (SUL) on aquatic early life stages is still limited. Here we investigated the lethal and sub-lethal effects of SUL during zebrafish embryo-larvae development. Results demonstrated that the 96 h and 120 h-LC50 of SUL to embryonic zebrafish was 2.02 mg/L, and the 30 d-LC50 was 0.899 mg/L after embryos exposed to SUL for 30 d. High concentrations of SUL delayed yolk sac absorption, disordered the hatching and heart rate during zebrafish embryonic stage, while 0.0100-0.100 mg/L SUL had no phenotypic changes on embryonic development, but decreased the body weight of larvae after 30 d exposure. RNA-seq identified 321, 394 and 727 differentially expressed genes in larvae after embryos exposed to 0.0100 mg/L, 0.0400 mg/L and 0.400 mg/L SUL for 30 d, found that the transcriptional profiles involved in heart development and endocrine disruption were simultaneously influenced by different concentrations of SUL, such as adrenergic signaling in cardiomyocytes, cardiac muscle contraction, cell adhesion molecules and steroid biosynthesis. Biochemical analysis showed that SUL increased the levels of E2, T3 and TSH, induced the activities of mitochondrial complex IV, cytochrome c oxidase, Ca2+-ATPase, total Na+K+-ATPase and Ca2+Mg2+-ATPase, and decreased ATP formation after embryos exposed to SUL for 5 d and 30 d. Further comprehensive analysis demonstrated that SUL caused more significantly alteration on the transcript, level or activity of the key elements involved in heart development and endocrine disruption after 30 d exposure, indicated long-term SUL exposure might cause more negative effects on zebrafish at doses below the presumed no-observed-adverse-effect level during early life development. The results inferred the environmental concentration of SUL might cause potential cardiac and endocrine health risk in zebrafish later life stages, also facilitated a better understanding of the sub-lethal effects and molecular mechanism of SUL on aquatic organism.
Collapse
Affiliation(s)
- Jinhua Jiang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Luyan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Changpeng Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Xueping Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China.
| |
Collapse
|
23
|
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: 5.5] [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
|
24
|
Li G, Shao Y, Guo HC, Zhi Y, Qiao B, Ma K, Lai YQ, Du J, Li Y. MicroRNA-27b-3p downregulates FGF1 and aggravates pathological cardiac remodelling. Cardiovasc Res 2021; 118:2139-2151. [PMID: 34358309 PMCID: PMC9302889 DOI: 10.1093/cvr/cvab248] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Indexed: 12/13/2022] Open
Abstract
AIMS The heart undergoes pathological remodelling under increased stress and neuronal imbalance. MicroRNAs (miRNAs) are involved in post-transcriptional regulation of genes in cardiac physiology and pathology. However, the mechanisms underlying miRNA-mediated regulation of pathological cardiac remodelling remain to be studied. This study aimed to explore the function of endogenous microRNA-27b-3p (miR-27b-3p) in pathological cardiac remodelling. METHODS AND RESULTS miR-27b-3p expression was elevated in the heart of a transverse aortic constriction (TAC)-induced cardiac hypertrophy mouse model. MiR-27b-knockout mice showed significantly attenuated cardiac hypertrophy, fibrosis, and inflammation induced by two independent pathological cardiac hypertrophy models, TAC and Angiotensin II (Ang II) perfusion. Transcriptome sequencing analysis revealed that miR-27b deletion significantly downregulated TAC-induced cardiac hypertrophy, fibrosis, and inflammatory genes. We identified fibroblast growth factor 1 (FGF1) as a miR-27b-3p target gene in the heart and was upregulated in miR-27b-null mice. We found that both recombinant FGF1 (rFGF1) and inhibition of miR-27b-3p enhanced mitochondrial oxidative phosphorylation (OXPHOS) and inhibited cardiomyocyte hypertrophy. Importantly, rFGF1 administration inhibited cardiac hypertrophy and fibrosis in TAC or Ang II-induced models, and enhanced OXPHOS by activating PGC1α/β. CONCLUSIONS Our study demonstrated that miR-27b-3p induces pathological cardiac remodelling and suggests that inhibition of endogenous miR-27b-3p or administration of FGF1 might have the potential to suppress cardiac remodelling in a clinical setting. TRANSLATIONAL PERSPECTIVE MicroRNAs (miRNAs) are involved in post-transcriptional regulation of genes in cardiac physiology and pathology. However, the mechanisms underlying miRNA-mediated regulation of pathological cardiac remodelling remain to be studied. We show for the first time that miR-27b deletion attenuates cardiac hypertrophy, fibrosis, and inflammation and that rFGF1 administration inhibits cardiac hypertrophy and fibrosis in TAC- or Ang II-induced models, and enhances OXPHOS by activating PGC1α/β. Our findings suggest that miR-27b-3p and FGF1 may be potential therapeutic targets to treat conditions characterised by pathological cardiac remodelling.
Collapse
Affiliation(s)
- Guoqi Li
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yihui Shao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Hong-Chang Guo
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Ying Zhi
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Bokang Qiao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Ke Ma
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yong-Qiang Lai
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| |
Collapse
|
25
|
Wasyluk W, Nowicka-Stążka P, Zwolak A. Heart Metabolism in Sepsis-Induced Cardiomyopathy-Unusual Metabolic Dysfunction of the Heart. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147598. [PMID: 34300048 PMCID: PMC8303349 DOI: 10.3390/ijerph18147598] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022]
Abstract
Due to the need for continuous work, the heart uses up to 8% of the total energy expenditure. Due to the relatively low adenosine triphosphate (ATP) storage capacity, the heart's work is dependent on its production. This is possible due to the metabolic flexibility of the heart, which allows it to use numerous substrates as a source of energy. Under normal conditions, a healthy heart obtains approximately 95% of its ATP by oxidative phosphorylation in the mitochondria. The primary source of energy is fatty acid oxidation, the rest of the energy comes from the oxidation of pyruvate. A failed heart is characterised by a disturbance in these proportions, with the contribution of individual components as a source of energy depending on the aetiology and stage of heart failure. A unique form of cardiac dysfunction is sepsis-induced cardiomyopathy, characterised by a significant reduction in energy production and impairment of cardiac oxidation of both fatty acids and glucose. Metabolic disorders appear to contribute to the pathogenesis of cardiac dysfunction and therefore are a promising target for future therapies. However, as many aspects of the metabolism of the failing heart remain unexplained, this issue requires further research.
Collapse
Affiliation(s)
- Weronika Wasyluk
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Patrycja Nowicka-Stążka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
| | - Agnieszka Zwolak
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
| |
Collapse
|
26
|
Feng W, Liu J, Wang S, Hu Y, Pan H, Hu T, Guan H, Zhang D, Mao Y. Alginate oligosaccharide alleviates D-galactose-induced cardiac ageing via regulating myocardial mitochondria function and integrity in mice. J Cell Mol Med 2021; 25:7157-7168. [PMID: 34227740 PMCID: PMC8335675 DOI: 10.1111/jcmm.16746] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022] Open
Abstract
Ageing is a crucial risk factor for the development of age‐related cardiovascular diseases. Therefore, the molecular mechanisms of ageing and novel anti‐ageing interventions need to be deeply studied. Alginate oligosaccharide (AOS) possesses high pharmacological activities and beneficial effects. Our study was undertaken to investigate whether AOS could be used as an anti‐ageing drug to alleviate cardiac ageing. D‐galactose (D‐gal)‐induced C57BL/6J ageing mice were established by subcutaneous injection of D‐gal (200 mg·kg‐1·d‐1) for 8 weeks. AOS (50, 100 and 150 mg·kg‐1·d‐1) were administrated intragastrically for the last 4 weeks. As a result, AOS prevented cardiac dysfunction in D‐gal‐induced ageing mice, including partially preserved ejection fraction (EF%) and fractional shortening (FS%). AOS inhibited D‐gal‐induced up‐regulation of natriuretic peptides A (ANP), brain natriuretic peptide (BNP) and ageing markers p53 and p21 in a dose‐dependent manner. To further explore the potential mechanisms contributing to the anti‐ageing protective effect of AOS, the age‐related mitochondrial compromise was analysed. Our data indicated that AOS alleviated D‐gal‐induced cardiac ageing by improving mitochondrial biogenesis, maintaining the mitochondrial integrity and enhancing the efficient removal of impaired mitochondria. AOS also decreased the ROS production and oxidative stress status, which, in turn, further inhibiting cardiac mitochondria from being destroyed. Together, these results demonstrate that AOS may be an effective therapeutic agent to alleviate cardiac ageing.
Collapse
Affiliation(s)
- Wenjing Feng
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, China
| | - Jianya Liu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shan Wang
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi Hu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Pan
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting Hu
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Marine Biomedical Research Institute of Qingdao, Qingdao, China
| | - Huashi Guan
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, China
| | - Yongjun Mao
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Zhang X, Zhang Z, Wang P, Han Y, Liu L, Li J, Chen Y, Liu D, Wang J, Tian X, Zhao Q, Yan F. Bawei Chenxiang Wan Ameliorates Cardiac Hypertrophy by Activating AMPK/PPAR-α Signaling Pathway Improving Energy Metabolism. Front Pharmacol 2021; 12:653901. [PMID: 34149410 PMCID: PMC8209424 DOI: 10.3389/fphar.2021.653901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/13/2021] [Indexed: 12/31/2022] Open
Abstract
Bawei Chenxiang Wan (BCW), a well-known traditional Chinese Tibetan medicine formula, is effective for the treatment of acute and chronic cardiovascular diseases. In the present study, we investigated the effect of BCW in cardiac hypertrophy and underlying mechanisms. The dose of 0.2, 0.4, and 0.8 g/kg BCW treated cardiac hypertrophy in SD rat model induced by isoprenaline (ISO). Our results showed that BCW (0.4 g/kg) could repress cardiac hypertrophy, indicated by macro morphology, heart weight to body weight ratio (HW/BW), left ventricle heart weight to body weight ratio (LVW/BW), hypertrophy markers, heart function, pathological structure, cross-sectional area (CSA) of myocardial cells, and the myocardial enzymes. Furthermore, we declared the mechanism of BCW anti-hypertrophy effect was associated with activating adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-α (PPAR-α) signals, which regulate carnitine palmitoyltransferase1β (CPT-1β) and glucose transport-4 (GLUT-4) to ameliorate glycolipid metabolism. Moreover, BCW also elevated mitochondrial DNA-encoded genes of NADH dehydrogenase subunit 1(ND1), cytochrome b (Cytb), and mitochondrially encoded cytochrome coxidase I (mt-co1) expression, which was associated with mitochondria function and oxidative phosphorylation. Subsequently, knocking down AMPK by siRNA significantly can reverse the anti-hypertrophy effect of BCW indicated by hypertrophy markers and cell surface of cardiomyocytes. In conclusion, BCW prevents ISO-induced cardiomyocyte hypertrophy by activating AMPK/PPAR-α to alleviate the disturbance in energy metabolism. Therefore, BCW can be used as an alternative drug for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Zhiying Zhang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Pengxiang Wang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Yiwei Han
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Lijun Liu
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Jie Li
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Yichun Chen
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Duxia Liu
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Jinying Wang
- School of Medical Science, Jinan University, Guangzhou, China
| | - Xiaoying Tian
- School of Medical Science, Jinan University, Guangzhou, China
| | - Qin Zhao
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Fengxia Yan
- School of Medical Science, Jinan University, Guangzhou, China
| |
Collapse
|
28
|
Bakogiannis C, Briasoulis A, Mouselimis D, Tsarouchas A, Papageorgiou N, Papadopoulos C, Fragakis N, Vassilikos V. Iron deficiency as therapeutic target in heart failure: a translational approach. Heart Fail Rev 2021; 25:173-182. [PMID: 31230175 DOI: 10.1007/s10741-019-09815-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Heart failure (HF) is a potentially debilitating condition, with a prognosis comparable to many forms of cancer. It is often complicated by anemia and iron deficiency (ID), which have been shown to even further harm patients' functional status and hospitalization risk. Iron is a cellular micronutrient that is essential for oxygen uptake and transportation, as well as mitochondrial energy production. Iron is crucially involved in electrochemical stability, maintenance of structure, and contractility of cardiomyocytes. There is mounting evidence that ID indeed hampers the homeostasis of these properties. Animal model and stem cell research has verified these findings on the cellular level, while clinical trials that treat ID in HF patients have shown promising results in improving real patient outcomes, as electromechanically compromised cardiomyocytes translate to HF exacerbations and arrhythmias in patients. In this article, we review our current knowledge on the role of iron in cardiac muscle cells, the contribution of ID to anemia and HF pathophysiology and the capacity of IV iron therapy to ameliorate the patients' arrhythmogenic profile, quality of life, and prognosis.
Collapse
Affiliation(s)
- Constantinos Bakogiannis
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54 352, Thessaloniki, Greece.
| | - Alexandros Briasoulis
- Division of Cardiovascular Medicine, Section of Heart Failure and Transplantation, University of Iowa, Iowa City, IA, USA
| | - Dimitrios Mouselimis
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54 352, Thessaloniki, Greece
| | - Anastasios Tsarouchas
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54 352, Thessaloniki, Greece
| | - Nikolaos Papageorgiou
- Electrophysiology Department, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Christodoulos Papadopoulos
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54 352, Thessaloniki, Greece
| | - Nikolaos Fragakis
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54 352, Thessaloniki, Greece
| | - Vassilios Vassilikos
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54 352, Thessaloniki, Greece
| |
Collapse
|
29
|
Akyuz E, Doganyigit Z, Eroglu E, Moscovicz F, Merelli A, Lazarowski A, Auzmendi J. Myocardial Iron Overload in an Experimental Model of Sudden Unexpected Death in Epilepsy. Front Neurol 2021; 12:609236. [PMID: 33643194 PMCID: PMC7905080 DOI: 10.3389/fneur.2021.609236] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
Uncontrolled repetitive generalized tonic-clonic seizures (GTCS) are the main risk factor for sudden unexpected death in epilepsy (SUDEP). GTCS can be observed in models such as Pentylenetetrazole kindling (PTZ-K) or pilocarpine-induced Status Epilepticus (SE-P), which share similar alterations in cardiac function, with a high risk of SUDEP. Terminal cardiac arrhythmia in SUDEP can develop as a result of a high rate of hypoxic stress-induced by convulsions with excessive sympathetic overstimulation that triggers a neurocardiogenic injury, recently defined as "Epileptic Heart" and characterized by heart rhythm disturbances, such as bradycardia and lengthening of the QT interval. Recently, an iron overload-dependent form of non-apoptotic cell death called ferroptosis was described at the brain level in both the PTZ-K and SE-P experimental models. However, seizure-related cardiac ferroptosis has not yet been reported. Iron overload cardiomyopathy (IOC) results from the accumulation of iron in the myocardium, with high production of reactive oxygen species (ROS), lipid peroxidation, and accumulation of hemosiderin as the final biomarker related to cardiomyocyte ferroptosis. Iron overload cardiomyopathy is the leading cause of death in patients with iron overload secondary to chronic blood transfusion therapy; it is also described in hereditary hemochromatosis. GTCS, through repeated hypoxic stress, can increase ROS production in the heart and cause cardiomyocyte ferroptosis. We hypothesized that iron accumulation in the "Epileptic Heart" could be associated with a terminal cardiac arrhythmia described in the IOC and the development of state-potentially in the development of SUDEP. Using the aforementioned PTZ-K and SE-P experimental models, after SUDEP-related repetitive GTCS, we observed an increase in the cardiac expression of hypoxic inducible factor 1α, indicating hypoxic-ischemic damage, and both necrotic cells and hemorrhagic areas were related to the possible hemosiderin production in the PTZ-K model. Furthermore, we demonstrated for the first time an accumulation of hemosiderin in the heart in the SE-P model. These results suggest that uncontrolled recurrent seizures, as described in refractory epilepsy, can give rise to high hypoxic stress in the heart, thus inducing hemosiderin accumulation as in IOC, and can act as an underlying hidden mechanism contributing to the development of a terminal cardiac arrhythmia in SUDEP. Because iron accumulation in tissues can be detected by non-invasive imaging methods, cardiac iron overload in refractory epilepsy patients could be treated with chelation therapy to reduce the risk of SUDEP.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, Medical School, Yozgat Bozok University, Yozgat, Turkey
| | - Zuleyha Doganyigit
- Department of Histology and Embryology, Medical School, Yozgat Bozok University, Yozgat, Turkey
| | - Ece Eroglu
- Medical School, Yozgat Bozok University, Yozgat, Turkey
| | - Franco Moscovicz
- Department of Clinical Biochemistry, School of Pharmacy and Biochemistry, Pathophysiology and Clinical Biochemistry Institute (INFIBIOC), University of Buenos Aires, Buenos Aires, Argentina
| | - Amalia Merelli
- Department of Clinical Biochemistry, School of Pharmacy and Biochemistry, Pathophysiology and Clinical Biochemistry Institute (INFIBIOC), University of Buenos Aires, Buenos Aires, Argentina
| | - Alberto Lazarowski
- Department of Clinical Biochemistry, School of Pharmacy and Biochemistry, Pathophysiology and Clinical Biochemistry Institute (INFIBIOC), University of Buenos Aires, Buenos Aires, Argentina
| | - Jerónimo Auzmendi
- Department of Clinical Biochemistry, School of Pharmacy and Biochemistry, Pathophysiology and Clinical Biochemistry Institute (INFIBIOC), University of Buenos Aires, Buenos Aires, Argentina.,National Council of Science and Technology (CONICET), Buenos Aires, Argentina
| |
Collapse
|
30
|
Weng Y, Zhang S, Huang W, Xie X, Ma Z, Fan Q. Efficacy of L-Carnitine for Dilated Cardiomyopathy: A Meta-Analysis of Randomized Controlled Trials. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9491615. [PMID: 33521132 PMCID: PMC7817303 DOI: 10.1155/2021/9491615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/24/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND L-carnitine mediates the utilization of fatty acids and glucose in the myocardium. The potential of L-carnitine in managing dilated cardiomyopathy (DCM) in patients has been extensively reported, with additional benefits. OBJECTIVE This meta-analysis purposed to explore the clinical efficacy of L-carnitine therapy on DCM patients. METHODS We searched publications up to May 2020 from several databases including PubMed, Embase, Cochrane Library, Chinese Biomedical (CBM) database, Chinese Science and Technology Periodicals database (VIP), Chinese National Knowledge Infrastructure (CNKI) database, and Wanfang database. Subsequently, publications that met the inclusion criteria were systematically evaluated by two independent reviewers. RESULTS A total of 23 RCTs conducted in China with 1455 DCM patients were included in this study. In the meta-analysis, L-carnitine therapy was associated with a considerable improvement in the overall efficacy (RR = 1.28, 95% CI (1.21-1.36), P < 0.0001), left ventricular ejection fraction (LVEF) (MD = 6.16%, 95% CI (4.50, 7.83), P < 0.0001), and cardiac output (CO) (MD = 0.88 L/min, 95% CI (0.51, 1.25), P < 0.0001) as compared to the control group. Moreover, L-carnitine therapy significantly decreased left ventricular end-diastolic dimension (LVEDD) (MD = -2.53, 95% CI (-3.95, -1.12), P = 0.0005), brain natriuretic peptide (BNP) (SMD = -1.71 ng/L, 95% CI (-3.02, -0.40), P = 0.01), and the transforming growth factor-beta (TGF-β1) (MD = -56.78 ng/L, 95% CI (-66.02, -47.53), P < 0.0001). CONCLUSIONS L-carnitine potentially enhanced the therapeutic efficiency in DCM patients. Following weaknesses in the evidence due to low methodological quality and high clinical heterogeneity in the included studies, well-designed trials are recommended.
Collapse
Affiliation(s)
- Yayun Weng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Shuo Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Wei Huang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Xianze Xie
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Zhiyuan Ma
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Qiaomei Fan
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| |
Collapse
|
31
|
Pierce JD, Shen Q, Vacek J, Rahman FK, Krueger KJ, Gupta B, Hiebert JB. Potential use of ubiquinol and d-ribose in patients with heart failure with preserved ejection fraction. Ann Med Surg (Lond) 2020; 55:77-80. [PMID: 32477499 PMCID: PMC7251495 DOI: 10.1016/j.amsu.2020.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 11/27/2022] Open
Abstract
•Manuscript Highlights.•HFpEF is associated with reduced ATP production in the myocardium.•Ubiquinol and d-ribose both contribute to the generation of myocardial ATP.•Both ubiquinol and d-ribose are being studied as supplemental treatments for patients with HFpEF.
Collapse
Affiliation(s)
- Janet D. Pierce
- School of Nursing, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Qiuhua Shen
- School of Nursing, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - James Vacek
- The University of Kansas Health System, 4000 Cambridge St, Kansas City, KS, 66160, USA
| | - Faith K. Rahman
- School of Nursing, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Kathryn J. Krueger
- School of Nursing, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Bhanu Gupta
- The University of Kansas Health System, 4000 Cambridge St, Kansas City, KS, 66160, USA
| | - John B. Hiebert
- School of Nursing, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| |
Collapse
|
32
|
Garcia AM, McPhaul JC, Sparagna GC, Jeffrey DA, Jonscher R, Patel SS, Sucharov CC, Stauffer BL, Miyamoto SD, Chatfield KC. Alteration of cardiolipin biosynthesis and remodeling in single right ventricle congenital heart disease. Am J Physiol Heart Circ Physiol 2020; 318:H787-H800. [PMID: 32056460 DOI: 10.1152/ajpheart.00494.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite advances in both medical and surgical therapies, individuals with single ventricle heart disease (SV) remain at high risk for the development of heart failure (HF). However, the molecular mechanisms underlying remodeling and eventual HF in patients with SV are poorly characterized. Cardiolipin (CL), an inner mitochondrial membrane phospholipid, is critical for proper mitochondrial function, and abnormalities in CL content and composition are known in various cardiovascular disease etiologies. The purpose of this study was to investigate myocardial CL content and composition in failing and nonfailing single right ventricle (RV) samples compared with normal control RV samples, to assess mRNA expression of CL biosynthetic and remodeling enzymes, and to quantitate relative mitochondrial copy number. A cross-sectional analysis of RV myocardial tissue from 22 failing SV (SVHF), 9 nonfailing SV (SVNF), and 10 biventricular control samples (BVNF) was performed. Expression of enzymes involved in CL biosynthesis and remodeling were analyzed using RT-qPCR and relative mitochondrial DNA copy number determined by qPCR. Normal phase high-pressure liquid chromatography coupled to electrospray ionization mass spectrometry was used to quantitate total and specific CL species. While mitochondrial copy number was not significantly different between groups, total CL content was significantly lower in SVHF myocardium compared with BVNF controls. Despite having lower total CL content however, the relative percentage of the major tetralinoleoyl CL species is preserved in SVHF samples relative to BVNF controls. Correspondingly, expression of enzymes involved in CL biosynthesis and remodeling were upregulated in SVHF samples when compared with both SVNF samples and BVNF controls.NEW & NOTEWORTHY The mechanisms underlying heart failure in the single ventricle (SV) congenital heart disease population are largely unknown. In this study we identify alterations in cardiac cardiolipin metabolism, composition, and content in children with SV heart disease. These findings suggest that cardiolipin could be a novel therapeutic target in this unique population of patients.
Collapse
Affiliation(s)
- Anastacia M Garcia
- Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Jessica C McPhaul
- Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Genevieve C Sparagna
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Danielle A Jeffrey
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Raleigh Jonscher
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Sonali S Patel
- Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Brian L Stauffer
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,Division of Cardiology, Denver Health Medical Center, Denver, Colorado
| | - Shelley D Miyamoto
- Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Kathryn C Chatfield
- Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
33
|
Polytarchou K, Dimitroglou Y, Varvarousis D, Christodoulis N, Psachoulia C, Pantziou C, Mourouzis I, Pantos C, Manolis AS. Methylmalonic acid and vitamin B12 in patients with heart failure. Hellenic J Cardiol 2019; 61:330-337. [PMID: 31740360 DOI: 10.1016/j.hjc.2019.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/01/2019] [Accepted: 10/16/2019] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Vitamin B12 deficiency among patients with heart failure (HF) may have been underestimated. High serum levels of methylmalonic acid (MMA) have been identified in several studies as an early indicator of vitamin B12 deficiency. Furthermore, MMA seems to constitute a biomarker of oxidative stress and mitochondrial dysfunction. There are scarce data regarding vitamin B12 and MMA in patients with HF. The aim of this study was to investigate vitamin B12 and MMA serum levels in patients with HF. METHODS One hundred five consecutive patients admitted to our hospital with symptoms and signs of acute decompensated HF were included in the study. Demographic and clinical characteristics as well as comorbidities and medical treatment before hospital admission were recorded. Transthoracic echocardiography was performed in all patients. Blood samples were collected during the first 24 hours of hospitalization and measured for complete blood count, biochemical profile, vitamin B12, N-terminal prohormone of brain natriuretic peptide, and MMA levels. Finally, 51 healthy individuals constituted the control group. RESULTS A total of 43.8% of patients with HF had elevated MMA levels, but only 10.5% had overt vitamin B12 deficiency, defined as serum cobalamin levels below 189 pg/ml. Mean MMA level was higher in patients with HF than in controls (33.0 ± 9.6 vs. 19.3 ± 6.3 ng/ml; p < 0.001). This difference remained significant when adjusted for age, sex, vitamin B12, and folate serum levels and kidney function (B = 14.7 (9.6-19.7); p < 0.001). MMA levels were higher in patients with acutely decompensated chronic HF than in those with newly diagnosed acute HF (34.7 ± 10.5 vs. 30.7 ± 7.8 ng/ml; p = 0.036). Correlation analysis revealed significantly negative correlation between MMA and vitamin B12 levels only in patients without comorbidities. CONCLUSION Patients with HF have elevated MMA levels, independent of age, gender, HF category, or comorbidities, possibly indicating subclinical vitamin B12 deficiency. Further research is needed to investigate subclinical vitamin B12 deficiency in patients with HF and/or to clarify whether MMA constitutes a biomarker of oxidative stress.
Collapse
Affiliation(s)
- Kali Polytarchou
- Third Department of Cardiology, Athens University School of Medicine, Athens, Greece; First Department of Cardiology, Evagelismos Hospital, Athens, Greece
| | - Yannis Dimitroglou
- First Department of Cardiology, Athens University School of Medicine, Athens, Greece
| | | | | | | | | | - Iordanis Mourouzis
- Third Department of Cardiology, Athens University School of Medicine, Athens, Greece
| | - Costas Pantos
- Third Department of Cardiology, Athens University School of Medicine, Athens, Greece
| | - Antonis S Manolis
- Third Department of Cardiology, Athens University School of Medicine, Athens, Greece; First Department of Cardiology, Athens University School of Medicine, Athens, Greece.
| |
Collapse
|
34
|
Scalise M, Console L, Galluccio M, Pochini L, Tonazzi A, Giangregorio N, Indiveri C. Exploiting Cysteine Residues of SLC Membrane Transporters as Targets for Drugs. SLAS DISCOVERY 2019; 24:867-881. [PMID: 31251685 DOI: 10.1177/2472555219856601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The observation that cysteine is the top gainer amino acid during evolution attracted the attention of scientists dealing with protein chemistry. The thiol group of cysteine, indeed, is a potential site for several types of reactions with variable specificity and strength. This feature proved to be promising also in the field of membrane transporters that represent boundary proteins fundamental for cell homeostasis. These proteins are classified, according to the driving force for transport, in primary or secondary active transporters. Another frequently used classification is nowadays based on phylogenesis. Two major groups are identified that take into account both criteria: the ABC and the SLC transporters, the second being much more numerous. The cellular localization of the transporters makes them very attractive for drug design. Moreover, the presence of at least one cysteine residue in all the annotated SLC transporters, so far, highlights the possibility of using the thiol (SH) residue for covalent drug targeting. Even if a delay exists in this research field due to the scarce knowledge of structure/function relationships, the setup of novel experimental tools for studying SLC proteins of plasma and organelle membranes opens an important perspective in pharmacology.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia e Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia e Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia e Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia e Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Annamaria Tonazzi
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), Bari, Italy
| | - Nicola Giangregorio
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), Bari, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia e Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
35
|
Paterek A, Mackiewicz U, Mączewski M. Iron and the heart: A paradigm shift from systemic to cardiomyocyte abnormalities. J Cell Physiol 2019; 234:21613-21629. [DOI: 10.1002/jcp.28820] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| | - Michał Mączewski
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| |
Collapse
|