1
|
Shi J, Jin Y, Lin S, Li X, Zhang D, Wu J, Qi Y, Li Y. Mitochondrial non-energetic function and embryonic cardiac development. Front Cell Dev Biol 2024; 12:1475603. [PMID: 39435335 PMCID: PMC11491369 DOI: 10.3389/fcell.2024.1475603] [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: 08/04/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
The initial contraction of the heart during the embryonic stage necessitates a substantial energy supply, predominantly derived from mitochondrial function. However, during embryonic heart development, mitochondria influence beyond energy supplementation. Increasing evidence suggests that mitochondrial permeability transition pore opening and closing, mitochondrial fusion and fission, mitophagy, reactive oxygen species production, apoptosis regulation, Ca2+ homeostasis, and cellular redox state also play critical roles in early cardiac development. Therefore, this review aims to describe the essential roles of mitochondrial non-energetic function embryonic cardiac development.
Collapse
Affiliation(s)
- Jingxian Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuxi Jin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Zheng H, Xu Y, Liehn EA, Rusu M. Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction. Int J Mol Sci 2024; 25:3114. [PMID: 38542087 PMCID: PMC10970003 DOI: 10.3390/ijms25063114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/31/2024] [Accepted: 02/10/2024] [Indexed: 06/26/2024] Open
Abstract
Currently, coronary artery bypass and reperfusion therapies are considered the gold standard in long-term treatments to restore heart function after acute myocardial infarction. As a drawback of these restoring strategies, reperfusion after an ischemic insult and sudden oxygen exposure lead to the exacerbated synthesis of additional reactive oxidative species and the persistence of increased oxidation levels. Attempts based on antioxidant treatment have failed to achieve an effective therapy for cardiovascular disease patients. The controversial use of vitamin C as an antioxidant in clinical practice is comprehensively systematized and discussed in this review. The dose-dependent adsorption and release kinetics mechanism of vitamin C is complex; however, this review may provide a holistic perspective on its potential as a preventive supplement and/or for combined precise and targeted therapeutics in cardiovascular management therapy.
Collapse
Affiliation(s)
- Huabo Zheng
- Department of Cardiology, Angiology and Intensive Care, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany;
- Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark;
| | - Yichen Xu
- Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark;
- Department of Histology and Embryology, Medicine and Life Sciences, Hainan Medical University, Haikou 571199, China
| | - Elisa A. Liehn
- Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark;
- National Institute of Pathology “Victor Babes”, Splaiul Independentei Nr. 99-101, 050096 Bucharest, Romania
| | - Mihaela Rusu
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany
| |
Collapse
|
3
|
Guo Z, Zhu J, Qin G, Jia Y, Liu Z, Yang N, Guo R. Static Magnetic Fields Promote Generation of Muscle Lineage Cells from Pluripotent Stem Cells and Myoblasts. Stem Cell Rev Rep 2023; 19:1402-1414. [PMID: 37000377 DOI: 10.1007/s12015-023-10535-z] [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] [Accepted: 03/21/2023] [Indexed: 04/01/2023]
Abstract
Static magnetic fields (SMFs) exhibit numerous biological effects and regulate the proliferation and differentiation of several adult stem cells. However, the role of SMFs in the self-renewal maintenance and developmental potential of pluripotent embryonic stem cells (ESCs) remains largely uninvestigated. Here, we show that SMFs promote the expression of the core pluripotent markers Sox2 and SSEA-1. Furthermore, SMFs facilitate the differentiation of ESCs into cardiomyocytes and skeletal muscle cells. Consistently, transcriptome analysis reveals that muscle lineage differentiation and skeletal system specification of ESCs are remarkably strengthened by SMF stimuli. Additionally, when treated with SMFs, C2C12 myoblasts exhibit an increased proliferation rate, improved expression of skeletal muscle markers and elevated myogenic differentiation capacity compared with control cells. Together, our data show that SMFs effectively promote muscle cell generation from pluripotent stem cells and myoblasts. The noninvasive and convenient physical stimuli can be used to increase the production of muscle cells in regenerative medicine and the manufacture of cultured meat in cellular agriculture.
Collapse
Affiliation(s)
- Zhaoyuan Guo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jiahao Zhu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guanyu Qin
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yumei Jia
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zheng Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Na Yang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China
- INDUC Scientific Co., Ltd, No. 28-132 Jinshan North Photoelectric Science and Technology Park, Wuxi, 214000, China
| | - Renpeng Guo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
4
|
Di Sante M, Antonucci S, Pontarollo L, Cappellaro I, Segat F, Deshwal S, Greotti E, Grilo LF, Menabò R, Di Lisa F, Kaludercic N. Monoamine oxidase A-dependent ROS formation modulates human cardiomyocyte differentiation through AKT and WNT activation. Basic Res Cardiol 2023; 118:4. [PMID: 36670288 PMCID: PMC9859871 DOI: 10.1007/s00395-023-00977-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 12/21/2022] [Accepted: 01/07/2023] [Indexed: 01/21/2023]
Abstract
During embryonic development, cardiomyocytes undergo differentiation and maturation, processes that are tightly regulated by tissue-specific signaling cascades. Although redox signaling pathways involved in cardiomyogenesis are established, the exact sources responsible for reactive oxygen species (ROS) formation remain elusive. The present study investigates whether ROS produced by the mitochondrial flavoenzyme monoamine oxidase A (MAO-A) play a role in cardiomyocyte differentiation from human induced pluripotent stem cells (hiPSCs). Wild type (WT) and MAO-A knock out (KO) hiPSCs were generated by CRISPR/Cas9 genome editing and subjected to cardiomyocyte differentiation. Mitochondrial ROS levels were lower in MAO-A KO compared to the WT cells throughout the differentiation process. MAO-A KO hiPSC-derived cardiomyocytes (hiPSC-CMs) displayed sarcomere disarray, reduced α- to β-myosin heavy chain ratio, GATA4 upregulation and lower macroautophagy levels. Functionally, genetic ablation of MAO-A negatively affected intracellular Ca2+ homeostasis in hiPSC-CMs. Mechanistically, MAO-A generated ROS contributed to the activation of AKT signaling that was considerably attenuated in KO cells. In addition, MAO-A ablation caused a reduction in WNT pathway gene expression consistent with its reported stimulation by ROS. As a result of WNT downregulation, expression of MESP1 and NKX2.5 was significantly decreased in MAO-A KO cells. Finally, MAO-A re-expression during differentiation rescued expression levels of cardiac transcription factors, contractile structure, and intracellular Ca2+ homeostasis. Taken together, these results suggest that MAO-A mediated ROS generation is necessary for the activation of AKT and WNT signaling pathways during cardiac lineage commitment and for the differentiation of fully functional human cardiomyocytes.
Collapse
Affiliation(s)
- Moises Di Sante
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Salvatore Antonucci
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Laura Pontarollo
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Ilaria Cappellaro
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Francesca Segat
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Soni Deshwal
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Elisa Greotti
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Luis F Grilo
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Roberta Menabò
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy.
| | - Nina Kaludercic
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127, Padua, Italy.
| |
Collapse
|
5
|
Kumar VHS. Cardiovascular Morbidities in Adults Born Preterm: Getting to the Heart of the Matter! CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9121843. [PMID: 36553286 PMCID: PMC9777245 DOI: 10.3390/children9121843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Advances in perinatal and neonatal care have led to improved survival of preterm infants into adulthood. However, the shift in focus to long-term health in adults born preterm requires a clear understanding of the impact of prematurity on developing organ systems and the development of adult-oriented disease. A less well-recognized area of risk for surviving preterm infants is their cardiometabolic health. Epidemiologic evidence has linked preterm birth to the development of systemic hypertension, type 2 diabetes, metabolic syndrome, heart failure, and ischemic heart disease. Of more significant concern is that the risk of cardiometabolic disorders is higher in adults born preterm compared to full-term infants. The interconnected nature of the cardio-pulmonary system means worsening morbidity and mortality in adults born preterm. Addressing the problems of adults born preterm holistically would help promote cardiovascular health, wellness, and quality of life over their lifetime. Recognizing that adults born preterm are a unique subset of the population is a challenge in the current healthcare environment. Addressing issues relevant to adults born preterm in the clinically and research domain, using technology to characterize cardiopulmonary physiology and exercise tolerance, developing screening tools for early diagnosis and treatment, and robust follow-up of these infants with access to longitudinal data would improve both the quality and longevity of life in adults born preterm.
Collapse
Affiliation(s)
- Vasantha H S Kumar
- Division of Neonatology, Department of Pediatrics, John R Oishei Children's Hospital, University at Buffalo, 1001 5th Floor Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
6
|
Gera J, Budakoti P, Suhag M, Mandal L, Mandal S. Physiological ROS controls Upd3-dependent modeling of ECM to support cardiac function in Drosophila. SCIENCE ADVANCES 2022; 8:eabj4991. [PMID: 35179958 PMCID: PMC8856619 DOI: 10.1126/sciadv.abj4991] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Despite their highly reactive nature, reactive oxygen species (ROS) at the physiological level serve as signaling molecules regulating diverse biological processes. While ROS usually act autonomously, they also function as local paracrine signals by diffusing out of the cells producing them. Using in vivo molecular genetic analyses in Drosophila, we provide evidence for ROS-dependent paracrine signaling that does not entail ROS release. We show that elevated levels of physiological ROS within the pericardial cells activate a signaling cascade transduced by Ask1, c-Jun N-terminal kinase, and p38 to regulate the expression of the cytokine Unpaired 3 (Upd3). Upd3 released by the pericardial cells controls fat body-specific expression of the extracellular matrix (ECM) protein Pericardin, essential for cardiac function and healthy life span. Therefore, our work reveals an unexpected inter-organ communication circuitry wherein high physiological levels of ROS regulate cytokine-dependent modulation of cardiac ECM with implications in normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Jayati Gera
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Prerna Budakoti
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Meghna Suhag
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Lolitika Mandal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
- Corresponding author.
| |
Collapse
|
7
|
Chen PH, Tjong WY, Yang HC, Liu HY, Stern A, Chiu DTY. Glucose-6-Phosphate Dehydrogenase, Redox Homeostasis and Embryogenesis. Int J Mol Sci 2022; 23:ijms23042017. [PMID: 35216131 PMCID: PMC8878822 DOI: 10.3390/ijms23042017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Normal embryogenesis requires complex regulation and precision, which depends on multiple mechanistic details. Defective embryogenesis can occur by various mechanisms. Maintaining redox homeostasis is of importance during embryogenesis. NADPH, as produced from the action of glucose-6-phosphate dehydrogenase (G6PD), has an important role in redox homeostasis, serving as a cofactor for glutathione reductase in the recycling of glutathione from oxidized glutathione and for NADPH oxidases and nitric oxide synthases in the generation of reactive oxygen (ROS) and nitrogen species (RNS). Oxidative stress differentially influences cell fate and embryogenesis. While low levels of stress (eustress) by ROS and RNS promote cell growth and differentiation, supra-physiological concentrations of ROS and RNS can lead to cell demise and embryonic lethality. G6PD-deficient cells and organisms have been used as models in embryogenesis for determining the role of redox signaling in regulating cell proliferation, differentiation and migration. Embryogenesis is also modulated by anti-oxidant enzymes, transcription factors, microRNAs, growth factors and signaling pathways, which are dependent on redox regulation. Crosstalk among transcription factors, microRNAs and redox signaling is essential for embryogenesis.
Collapse
Affiliation(s)
- Po-Hsiang Chen
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Wen-Ye Tjong
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Correspondence: ; Tel.: +886-3-6108175; Fax: +886-3-6102327
| | - Hui-Ya Liu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Arnold Stern
- Grossman School of Medicine, New York University, New York, NY 10016, USA;
| | - Daniel Tsun-Yee Chiu
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
| |
Collapse
|
8
|
Redox Homeostasis and Regulation in Pluripotent Stem Cells: Uniqueness or Versatility? Int J Mol Sci 2021; 22:ijms222010946. [PMID: 34681606 PMCID: PMC8535588 DOI: 10.3390/ijms222010946] [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: 09/16/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022] Open
Abstract
Pluripotent stem cells (PSCs) hold great potential both in studies on developmental biology and clinical practice. Mitochondrial metabolism that encompasses pathways that generate ATP and produce ROS significantly differs between PSCs and somatic cells. Correspondingly, for quite a long time it was believed that the redox homeostasis in PSCs is also highly specific due to the hypoxic niche of their origin-within the pre-implantation blastocyst. However, recent research showed that redox parameters of cultivated PSCs have much in common with that of their differentiated progeny cells. Moreover, it has been proven that, similar to somatic cells, maintaining the physiological ROS level is critical for the regulation of PSC identity, proliferation, differentiation, and de-differentiation. In this review, we aimed to summarize the studies of redox metabolism and signaling in PSCs to compare the redox profiles of pluripotent and differentiated somatic cells. We collected evidence that PSCs possess metabolic plasticity and are able to adapt to both hypoxia and normoxia, that pluripotency is not strictly associated with anaerobic conditions, and that cellular redox homeostasis is similar in PSCs and many other somatic cells under in vitro conditions that may be explained by the high conservatism of the redox regulation system.
Collapse
|
9
|
Gene Therapy: Targeting Cardiomyocyte Proliferation to Repopulate the Ischemic Heart. J Cardiovasc Pharmacol 2021; 78:346-360. [PMID: 34516452 DOI: 10.1097/fjc.0000000000001072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/16/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Adult mammalian cardiomyocytes show scarce division ability, which makes the heart ineffective in replacing lost contractile cells after ischemic cardiomyopathy. In the past decades, there have been increasing efforts in the search for novel strategies to regenerate the injured myocardium. Among them, gene therapy is one of the most promising ones, based on recent and emerging studies that support the fact that functional cardiomyocyte regeneration can be accomplished by the stimulation and enhancement of the endogenous ability of these cells to achieve cell division. This capacity can be targeted by stimulating several molecules, such as cell cycle regulators, noncoding RNAs, transcription, and metabolic factors. Therefore, the proposed target, together with the selection of the vector used, administration route, and the experimental animal model used in the development of the therapy would determine the success in the clinical field.
Collapse
|
10
|
Dudek J, Kutschka I, Maack C. Metabolic and Redox Regulation of Cardiovascular Stem Cell Biology and Pathology. Antioxid Redox Signal 2021; 35:163-181. [PMID: 33121253 DOI: 10.1089/ars.2020.8201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Cardiovascular stem cells are important for regeneration and repair of damaged tissue. Recent Advances: Pluripotent stem cells have a unique metabolism, which is adopted for their energetic and biosynthetic demand as rapidly proliferating cells. Stem cell differentiation requires an exceptional metabolic flexibility allowing for metabolic remodeling between glycolysis and oxidative phosphorylation. Critical Issues: Respiration is associated with the generation of reactive oxygen species (ROS) by the mitochondrial respiratory chain. But also the membrane-bound protein nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase, NOX) contributes to ROS levels. ROS not only play a significant role in stem cell differentiation and tissue renewal but also cause senescence and contribute to tissue aging. Future Directions: For utilization of stem cells in therapeutic approaches, a deep understanding of the molecular mechanisms how metabolism and the cellular redox state regulate stem cell differentiation is required. Modulating the redox state of stem cells using antioxidative agents may be suitable to enhance activity of endothelial progenitor cells. Antioxid. Redox Signal. 35, 163-181.
Collapse
Affiliation(s)
- Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
11
|
Maraldi T, Angeloni C, Prata C, Hrelia S. NADPH Oxidases: Redox Regulators of Stem Cell Fate and Function. Antioxidants (Basel) 2021; 10:973. [PMID: 34204425 PMCID: PMC8234808 DOI: 10.3390/antiox10060973] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
One of the major sources of reactive oxygen species (ROS) generated within stem cells is the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes (NOXs), which are critical determinants of the redox state beside antioxidant defense mechanisms. This balance is involved in another one that regulates stem cell fate: indeed, self-renewal, proliferation, and differentiation are decisive steps for stem cells during embryo development, adult tissue renovation, and cell therapy application. Ex vivo culture-expanded stem cells are being investigated for tissue repair and immune modulation, but events such as aging, senescence, and oxidative stress reduce their ex vivo proliferation, which is crucial for their clinical applications. Here, we review the role of NOX-derived ROS in stem cell biology and functions, focusing on positive and negative effects triggered by the activity of different NOX isoforms. We report recent findings on downstream molecular targets of NOX-ROS signaling that can modulate stem cell homeostasis and lineage commitment and discuss the implications in ex vivo expansion and in vivo engraftment, function, and longevity. This review highlights the role of NOX as a pivotal regulator of several stem cell populations, and we conclude that these aspects have important implications in the clinical utility of stem cells, but further studies on the effects of pharmacological modulation of NOX in human stem cells are imperative.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| | - Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| |
Collapse
|
12
|
Effect of Diphenyleneiodonium Chloride on Intracellular Reactive Oxygen Species Metabolism with Emphasis on NADPH Oxidase and Mitochondria in Two Therapeutically Relevant Human Cell Types. Pharmaceutics 2020; 13:pharmaceutics13010010. [PMID: 33374729 PMCID: PMC7823933 DOI: 10.3390/pharmaceutics13010010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) have recently been recognized as important signal transducers, particularly regulating proliferation and differentiation of cells. Diphenyleneiodonium (DPI) is known as an inhibitor of the nicotinamide adenine dinucleotide phosphate oxidase (NOX) and is also affecting mitochondrial function. The aim of this study was to investigate the effect of DPI on ROS metabolism and mitochondrial function in human amniotic membrane mesenchymal stromal cells (hAMSCs), human bone marrow mesenchymal stromal cells (hBMSCs), hBMSCs induced into osteoblast-like cells, and osteosarcoma cell line MG-63. Our data suggested a combination of a membrane potential sensitive fluorescent dye, tetramethylrhodamine methyl ester (TMRM), and a ROS-sensitive dye, CM-H2DCFDA, combined with a pretreatment with mitochondria-targeted ROS scavenger MitoTEMPO as a good tool to examine effects of DPI. We observed critical differences in ROS metabolism between hAMSCs, hBMSCs, osteoblast-like cells, and MG-63 cells, which were linked to energy metabolism. In cell types using predominantly glycolysis as the energy source, such as hAMSCs, DPI predominantly interacted with NOX, and it was not toxic for the cells. In hBMSCs, the ROS turnover was influenced by NOX activity rather than by the mitochondria. In cells with aerobic metabolism, such as MG 63, the mitochondria became an additional target for DPI, and these cells were prone to the toxic effects of DPI. In summary, our data suggest that undifferentiated cells rather than differentiated parenchymal cells should be considered as potential targets for DPI.
Collapse
|
13
|
Stem Cell Metabolism: Powering Cell-Based Therapeutics. Cells 2020; 9:cells9112490. [PMID: 33207756 PMCID: PMC7696341 DOI: 10.3390/cells9112490] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Cell-based therapeutics for cardiac repair have been extensively used during the last decade. Preclinical studies have demonstrated the effectiveness of adoptively transferred stem cells for enhancement of cardiac function. Nevertheless, several cell-based clinical trials have provided largely underwhelming outcomes. A major limitation is the lack of survival in the harsh cardiac milieu as only less than 1% donated cells survive. Recent efforts have focused on enhancing cell-based therapeutics and understanding the biology of stem cells and their response to environmental changes. Stem cell metabolism has recently emerged as a critical determinant of cellular processes and is uniquely adapted to support proliferation, stemness, and commitment. Metabolic signaling pathways are remarkably sensitive to different environmental signals with a profound effect on cell survival after adoptive transfer. Stem cells mainly generate energy through glycolysis while maintaining low oxidative phosphorylation (OxPhos), providing metabolites for biosynthesis of macromolecules. During commitment, there is a shift in cellular metabolism, which alters cell function. Reprogramming stem cell metabolism may represent an attractive strategy to enhance stem cell therapy for cardiac repair. This review summarizes the current literature on how metabolism drives stem cell function and how this knowledge can be applied to improve cell-based therapeutics for cardiac repair.
Collapse
|
14
|
Roles of Reactive Oxygen Species in Cardiac Differentiation, Reprogramming, and Regenerative Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2102841. [PMID: 32908625 PMCID: PMC7475763 DOI: 10.1155/2020/2102841] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in mechanisms of heart development and regenerative therapies such as the use of pluripotent stem cells. The roles of ROS mediating cell fate are dependent on the intensity of stimuli, cellular context, and metabolic status. ROS mainly act through several targets (such as kinases and transcription factors) and have diverse roles in different stages of cardiac differentiation, proliferation, and maturation. Therefore, further detailed investigation and characterization of redox signaling will help the understanding of the molecular mechanisms of ROS during different cellular processes and enable the design of targeted strategies to foster cardiac regeneration and functional recovery. In this review, we focus on the roles of ROS in cardiac differentiation as well as transdifferentiation (direct reprogramming). The potential mechanisms are discussed in regard to ROS generation pathways and regulation of downstream targets. Further methodological optimization is required for translational research in order to robustly enhance the generation efficiency of cardiac myocytes through metabolic modulations. Additionally, we highlight the deleterious effect of the host's ROS on graft (donor) cells in a paracrine manner during stem cell-based implantation. This knowledge is important for the development of antioxidant strategies to enhance cell survival and engraftment of tissue engineering-based technologies. Thus, proper timing and level of ROS generation after a myocardial injury need to be tailored to ensure the maximal efficacy of regenerative therapies and avoid undesired damage.
Collapse
|
15
|
Goss KN, Haraldsdottir K, Beshish AG, Barton GP, Watson AM, Palta M, Chesler NC, Francois CJ, Wieben O, Eldridge MW. Association Between Preterm Birth and Arrested Cardiac Growth in Adolescents and Young Adults. JAMA Cardiol 2020; 5:910-919. [PMID: 32432648 DOI: 10.1001/jamacardio.2020.1511] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Importance Premature birth is associated with substantially higher lifetime risk for cardiovascular disease, including arrhythmia, ischemic disease, and heart failure, although the underlying mechanisms are poorly understood. Objective To characterize cardiac structure and function in adolescents and young adults born preterm using cardiac magnetic resonance imaging (MRI). Design, Setting, and Participants This cross-sectional cohort study at an academic medical center included adolescents and young adults born moderately to extremely premature (20 in the adolescent cohort born from 2003 to 2004 and 38 in the young adult cohort born in the 1980s and 1990s) and 52 age-matched participants who were born at term and underwent cardiac MRI. The dates of analysis were February 2016 to October 2019. Exposures Premature birth (gestational age ≤32 weeks) or birth weight less than 1500 g. Main Outcomes and Measures Main study outcomes included MRI measures of biventricular volume, mass, and strain. Results Of 40 adolescents (24 [60%] girls), the mean (SD) age of participants in the term and preterm groups was 13.3 (0.7) years and 13.0 (0.7) years, respectively. Of 70 adults (43 [61%] women), the mean (SD) age of participants in the term and preterm groups was 25.4 (2.9) years and 26.5 (3.5) years, respectively. Participants from both age cohorts who were born prematurely had statistically significantly smaller biventricular cardiac chamber size compared with participants in the term group: the mean (SD) left ventricular end-diastolic volume index was 72 (7) vs 80 (9) and 80 (10) vs 92 (15) mL/m2 for adolescents and adults in the preterm group compared with age-matched participants in the term group, respectively (P < .001), and the mean (SD) left ventricular end-systolic volume index was 30 (4) vs 34 (6) and 32 (7) vs 38 (8) mL/m2, respectively (P < .001). Stroke volume index was also reduced in adolescent vs adult participants in the preterm group vs age-matched participants in the term group, with a mean (SD) of 42 (7) vs 46 (7) and 48 (7) vs 54 (9) mL/m2, respectively (P < .001), although biventricular ejection fractions were preserved. Biventricular mass was statistically significantly lower in adolescents and adults born preterm: the mean (SD) left ventricular mass index was 39.6 (5.9) vs 44.4 (7.5) and 40.7 (7.3) vs 49.8 (14.0), respectively (P < .001). Cardiac strain analyses demonstrated a hypercontractile heart, primarily in the right ventricle, in adults born prematurely. Conclusions and Relevance In this cross-sectional study, adolescents and young adults born prematurely had statistically significantly smaller biventricular cardiac chamber size and decreased cardiac mass. Although function was preserved in both age groups, these morphologic differences may be associated with elevated lifetime cardiovascular disease risk after premature birth.
Collapse
Affiliation(s)
- Kara N Goss
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Kristin Haraldsdottir
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Kinesiology, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Arij G Beshish
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Gregory P Barton
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Andrew M Watson
- Department of Orthopedic and Rehabilitation Medicine, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Mari Palta
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Naomi C Chesler
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Biomedical Engineering, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Chris J Francois
- Department of Biomedical Engineering, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Oliver Wieben
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Biomedical Engineering, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Marlowe W Eldridge
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Kinesiology, School of Medicine and Public Health, University of Wisconsin-Madison.,Department of Biomedical Engineering, School of Medicine and Public Health, University of Wisconsin-Madison
| |
Collapse
|
16
|
NOX2 Is Critical to Endocardial to Mesenchymal Transition and Heart Development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1679045. [PMID: 32655758 PMCID: PMC7320281 DOI: 10.1155/2020/1679045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/19/2020] [Accepted: 05/18/2020] [Indexed: 12/05/2022]
Abstract
NADPH oxidases (NOX) are a major source of reactive oxygen species (ROS) production in the heart. ROS signaling regulates gene expression, cell proliferation, apoptosis, and migration. However, the role of NOX2 in embryonic heart development remains elusive. We hypothesized that deficiency of Nox2 disrupts endocardial to mesenchymal transition (EndMT) and results in congenital septal and valvular defects. Our data show that 34% of Nox2−/− neonatal mice had various congenital heart defects (CHDs) including atrial septal defects (ASD), ventricular septal defects (VSD), atrioventricular canal defects (AVCD), and malformation of atrioventricular and aortic valves. Notably, Nox2−/− embryonic hearts show abnormal development of the endocardial cushion as evidenced by decreased cell proliferation and an increased rate of apoptosis. Additionally, Nox2 deficiency disrupted EndMT of atrioventricular cushion explants ex vivo. Furthermore, treatment with N-acetylcysteine (NAC) to reduce ROS levels in the wild-type endocardial cushion explants decreased the number of cells undergoing EndMT. Importantly, deficiency of Nox2 was associated with reduced expression of Gata4, Tgfβ2, Bmp2, Bmp4, and Snail1, which are critical to endocardial cushion and valvoseptal development. We conclude that NOX2 is critical to EndMT, endocardial cushion cell proliferation, and normal embryonic heart development.
Collapse
|
17
|
The Role of Oxidative Stress in Cardiac Disease: From Physiological Response to Injury Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5732956. [PMID: 32509147 PMCID: PMC7244977 DOI: 10.1155/2020/5732956] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/11/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive chemical species containing oxygen, controlled by both enzymatic and nonenzymatic antioxidant defense systems. In the heart, ROS play an important role in cell homeostasis, by modulating cell proliferation, differentiation, and excitation-contraction coupling. Oxidative stress occurs when ROS production exceeds the buffering capacity of the antioxidant defense systems, leading to cellular and molecular abnormalities, ultimately resulting in cardiac dysfunction. In this review, we will discuss the physiological sources of ROS in the heart, the mechanisms of oxidative stress-related myocardial injury, and the implications of experimental studies and clinical trials with antioxidant therapies in cardiovascular diseases.
Collapse
|
18
|
Cardiac regeneration as an environmental adaptation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118623. [DOI: 10.1016/j.bbamcr.2019.118623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
|
19
|
Rasmussen ML, Taneja N, Neininger AC, Wang L, Robertson GL, Riffle SN, Shi L, Knollmann BC, Burnette DT, Gama V. MCL-1 Inhibition by Selective BH3 Mimetics Disrupts Mitochondrial Dynamics Causing Loss of Viability and Functionality of Human Cardiomyocytes. iScience 2020; 23:101015. [PMID: 32283523 PMCID: PMC7155208 DOI: 10.1016/j.isci.2020.101015] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/25/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022] Open
Abstract
MCL-1 is a well-characterized inhibitor of cell death that has also been shown to be a regulator of mitochondrial dynamics in human pluripotent stem cells. We used cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) to uncover whether MCL-1 is crucial for cardiac function and survival. Inhibition of MCL-1 by BH3 mimetics resulted in the disruption of mitochondrial morphology and dynamics as well as disorganization of the actin cytoskeleton. Interfering with MCL-1 function affects the homeostatic proximity of DRP-1 and MCL-1 at the outer mitochondrial membrane, resulting in decreased functionality of hiPSC-CMs. Cardiomyocytes display abnormal cardiac performance even after caspase inhibition, supporting a nonapoptotic activity of MCL-1 in hiPSC-CMs. BH3 mimetics targeting MCL-1 are promising anti-tumor therapeutics. Progression toward using BCL-2 family inhibitors, especially targeting MCL-1, depends on understanding its canonical function not only in preventing apoptosis but also in the maintenance of mitochondrial dynamics and function. BH3 mimetics targeting MCL-1 disrupt the mitochondrial network of human iPSC-CMs The BH3-mimetic-mediated effects on mitochondrial dynamics are DRP-1-dependent Targeting MCL-1 affects the survival and function of human cardiomyocytes Human iPSC-derived cardiomyocytes can be used to reveal toxicity of MCL-1 inhibitors
Collapse
Affiliation(s)
- Megan L Rasmussen
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Nilay Taneja
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Abigail C Neininger
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Lili Wang
- Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Nashville, TN 37232, USA
| | - Gabriella L Robertson
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Stellan N Riffle
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Linzheng Shi
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Bjorn C Knollmann
- Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Nashville, TN 37232, USA
| | - Dylan T Burnette
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Vivian Gama
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
20
|
Choe MS, Yeo HC, Bae CM, Han HJ, Baek KM, Kim JS, Lim KS, Shin IS, Chang W, Yun SP, Lee HJ, Lee MY. Trolox-induced cardiac differentiation is mediated by the inhibition of Wnt/β-catenin signaling in human embryonic stem cells. Cell Biol Int 2019; 43:1505-1515. [PMID: 31293030 DOI: 10.1002/cbin.11200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 07/07/2019] [Indexed: 01/24/2023]
Abstract
Cardiac differentiation of human pluripotent stem cells may be induced under chemically defined conditions, wherein the regulation of Wnt/β-catenin pathway is often desirable. Here, we examined the effect of trolox, a vitamin E analog, on the cardiac differentiation of human embryonic stem cells (hESCs). 6-Hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid (Trolox) significantly enhanced cardiac differentiation in a time- and dose-dependent manner after the mesodermal differentiation of hESCs. Trolox promoted hESC cardiac differentiation through its inhibitory activity against the Wnt/β-catenin pathway. This study demonstrates an efficient cardiac differentiation method and reveals a novel Wnt/β-catenin regulator.
Collapse
Affiliation(s)
- Mu Seog Choe
- Department of Molecular Pharmacology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Korea
| | - Han Cheol Yeo
- Department of Molecular Pharmacology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Korea
| | - Chang Min Bae
- Department of Molecular Pharmacology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Seoul, 08826, Korea
| | - Kyung Min Baek
- Department of Cardiovascular and Neurologic Disease, College of Oriental Medicine, Daegu Haany University, 136 Sincheondong-ro, Daegu, 42158, Korea
| | - Joong Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju, Jeollanam-do 58245, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Cheongju, Chungcheongbuk-do 28116, Korea
| | - In-Sik Shin
- Department of Veterinary Pharmacology, College of Veterinary Medicine (BK21 Project Team), Chonnam National University, 77 Yongbong-ro, Gwangju, 61186, Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, 2 Busandaehak-ro, Busan, 46241, Korea
| | - Seung Pil Yun
- Department of Pharmacology, School of Medicine, Gyeongsang National University, 15 Jinjudae-ro 816, Jinju, 52727, Korea
| | - Ho Jin Lee
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar street, New Haven, Connecticut, 06520, USA
| | - Min Young Lee
- Department of Molecular Pharmacology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Korea
| |
Collapse
|
21
|
Peters MMC, Meijs TA, Gathier W, Doevendans PAM, Sluijter JPG, Chamuleau SAJ, Neef K. Follistatin-like 1 in Cardiovascular Disease and Inflammation. Mini Rev Med Chem 2019; 19:1379-1389. [PMID: 30864520 DOI: 10.2174/1389557519666190312161551] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/16/2022]
Abstract
Follistatin-like 1 (FSTL1), a secreted glycoprotein, has been shown to participate in regulating developmental processes and to be involved in states of disease and injury. Spatiotemporal regulation and posttranslational modifications contribute to its specific functions and make it an intriguing candidate to study disease mechanisms and potentially develop new therapies. With cardiovascular diseases as the primary cause of death worldwide, clarification of mechanisms underlying cardiac regeneration and revascularization remains essential. Recent findings on FSTL1 in both acute coronary syndrome and heart failure emphasize its potential as a target for cardiac regenerative therapy. With this review, we aim to shed light on the role of FSTL1 specifically in cardiovascular disease and inflammation.
Collapse
Affiliation(s)
- Marijn M C Peters
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| | - Timion A Meijs
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| | - Wouter Gathier
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| | - Pieter A M Doevendans
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| | - Steven A J Chamuleau
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| | - Klaus Neef
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, Netherlands
| |
Collapse
|
22
|
Bottje WG. BOARD INVITED REVIEW: Oxidative stress and efficiency: the tightrope act of mitochondria in health and disease1,2. J Anim Sci 2019; 97:3169-3179. [PMID: 31247079 DOI: 10.1093/jas/skz219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is an unavoidable consequence of aerobic metabolism. Whereas high amounts of mitochondrial reactive oxygen species (ROS) can cause oxidation, low levels play important roles in signal transduction. In a Pedigree male (PedM) broiler model of feed efficiency (FE), the low FE phenotype was characterized by increased ROS in isolated mitochondria (muscle, liver, and duodenum) with a pervasive protein oxidation in mitochondria and tissues. Subsequent proteogenomic studies in muscle revealed evidence of enhanced mitoproteome abundance, enhanced mitochondrial phosphocreatine shuttling expression, and enhanced ribosome assembly in the high FE phenotype. Surprisingly, an enhanced infrastructure would foster greater repair of damaged proteins or organelles through the autophagy and proteosome pathways in the high FE phenotype. Although protein and organelle degradation, recycling, and reconstruction would be energetically expensive, it is possible that energy invested into maintaining optimal function of proteins and organelles contributes to cellular efficiency in the high FE phenotype. New findings in mitochondrial physiology have been reported in the last several years. Reverse electron transport (RET), once considered an artifact of in vitro conditions, now is recognized to play significant roles in inflammation, ischemia-reperfusion, muscle differentiation, and energy utilization. A topology of ROS production indicates that ROS derived from Complex I of the respiratory chain primarily causes oxidation, whereas ROS generated from Complex III are primarily involved in cell signaling. It is also apparent that there is a constant fission and fusion process that mitochondria undergo that help maintain optimal mitochondrial function and enables mitochondria to adjust to periods of nutrient limitation and nutrient excess. Understanding the balancing act that mitochondria play in health and disease will continue to be a vital biological component in health-production efficiency and disease in commercial animal agriculture.
Collapse
Affiliation(s)
- Walter G Bottje
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR
| |
Collapse
|
23
|
Early IgG Response to Foot and Mouth Disease Vaccine Formulated with a Vegetable Oil Adjuvant. Vaccines (Basel) 2019; 7:vaccines7040143. [PMID: 31600943 PMCID: PMC6963984 DOI: 10.3390/vaccines7040143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
The present study evaluated soybean oil (SO) containing vitamin E (VE) and ginseng saponins (GS) (SO-VE-GS) for their adjuvant effect on foot-and-mouth disease (FMD) vaccine. Since mineral oil ISA 206 is a common adjuvant used in the FMD vaccine, it was used as a control adjuvant in this study. VE and GS were found to have a synergistic adjuvant effect. When mice were immunized with the FMD vaccine emulsified in SO with VE and GS, significantly higher serum IgG, IgG1, and IgG2a were found than VE and GS used alone. SO-VE-GS and ISA 206 behaved differently in adjuvant activities. When mice were immunized with the FMD vaccine adjuvanted with SO-VE-GS, significantly higher and earlier production of serum IgG was found than that adjuvanted with ISA 206. Although both adjuvants significantly increased the number of bone marrow plasma cells, a stimulation index of lymphocytes (SI) as well as the production of IL-4 and IL-6, SO-VE-GS promoted significantly higher SI and the ratio of CD4+/CD8+ T cells with production of increased IFN-γ and decreased TGF-β1 as compared with the ISA 206 group. The data suggested that SO-VE-GS activated Th1/Th2 immune responses. Transcriptome analysis of splenocytes showed that differentially expressed genes (DEGs), immune-related gene ontology (GO) terms, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were significantly enriched in the SO-VE-GS group. Therefore, the potent adjuvant effect of SO-VE-GS on the FMD vaccine may be attributed to the immune-related gene profile expressed in lymphocytes. Due to its plant origin and due to being much cheaper than imported mineral oil ISA 206, SO-VE-GS deserves further study in relation to vaccines used in food animals.
Collapse
|
24
|
Abstract
In mammals, most cardiomyocytes (CMs) become polyploid (they have more than two complete sets of chromosomes). The purpose of this review is to evaluate assumptions about CM ploidy that are commonly discussed, even if not experimentally demonstrated, and to highlight key issues that are still to be resolved. Topics discussed here include (a) technical and conceptual difficulties in defining a polyploid CM, (b) the candidate role of reactive oxygen as a proximal trigger for the onset of polyploidy, (c) the relationship between polyploidization and other aspects of CM maturation, (d) recent insights related to the regenerative role of the subpopulation of CMs that are not polyploid, and (e) speculations as to why CMs become polyploid at all. New approaches to experimentally manipulate CM ploidy may resolve some of these long-standing and fundamental questions.
Collapse
Affiliation(s)
- Peiheng Gan
- Department of Regenerative Medicine and Cell Biology and Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina 29425, USA; .,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, California 90033, USA
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology and Anatomy, and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology and Department of Medicine Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina 29425, USA;
| |
Collapse
|
25
|
Bottje W. Oxidative metabolism and efficiency: the delicate balancing act of mitochondria. Poult Sci 2019; 98:4223-4230. [DOI: 10.3382/ps/pey405] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
26
|
Zhao Q, Sun Q, Zhou L, Liu K, Jiao K. Complex Regulation of Mitochondrial Function During Cardiac Development. J Am Heart Assoc 2019; 8:e012731. [PMID: 31215339 PMCID: PMC6662350 DOI: 10.1161/jaha.119.012731] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Qiancong Zhao
- Department of Cardiovascular SurgeryThe Second Hospital of Jilin UniversityChangchunChina
- Department of GeneticsThe University of Alabama at BirminghamAL
| | - Qianchuang Sun
- Department of AnesthesiologyThe Second Hospital of Jilin UniversityChangchunChina
- Department of GeneticsThe University of Alabama at BirminghamAL
| | - Lufang Zhou
- Department of MedicineThe University of Alabama at BirminghamAL
| | - Kexiang Liu
- Department of Cardiovascular SurgeryThe Second Hospital of Jilin UniversityChangchunChina
| | - Kai Jiao
- Department of GeneticsThe University of Alabama at BirminghamAL
| |
Collapse
|
27
|
Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration. Trends Cell Biol 2019; 29:514-530. [DOI: 10.1016/j.tcb.2019.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 01/08/2023]
|
28
|
The Role of Reactive Oxygen Species in In Vitro Cardiac Maturation. Trends Mol Med 2019; 25:482-493. [PMID: 31080142 DOI: 10.1016/j.molmed.2019.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 12/27/2022]
Abstract
Recent advances in developmental biology and biomedical engineering have significantly improved the efficiency and purity of cardiomyocytes (CMs) generated from pluripotent stem cells (PSCs). Regardless of the protocol used to derive CMs, these cells exhibit hallmarks of functional immaturity. In this Opinion, we focus on reactive oxygen species (ROS), signaling molecules that can potentially modulate cardiac maturation. We outline how ROS impacts nearly every aspect associated with cardiac maturation, including contractility, calcium handling, metabolism, and hypertrophy. Though the precise role of ROS in cardiac maturation has yet to be elucidated, ROS may provide a valuable perspective for understanding the molecular mechanisms for cardiac maturation under various conditions.
Collapse
|
29
|
Adult Cardiac Stem Cell Aging: A Reversible Stochastic Phenomenon? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5813147. [PMID: 30881594 PMCID: PMC6383393 DOI: 10.1155/2019/5813147] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 11/08/2018] [Indexed: 12/17/2022]
Abstract
Aging is by far the dominant risk factor for the development of cardiovascular diseases, whose prevalence dramatically increases with increasing age reaching epidemic proportions. In the elderly, pathologic cellular and molecular changes in cardiac tissue homeostasis and response to injury result in progressive deteriorations in the structure and function of the heart. Although the phenotypes of cardiac aging have been the subject of intense study, the recent discovery that cardiac homeostasis during mammalian lifespan is maintained and regulated by regenerative events associated with endogenous cardiac stem cell (CSC) activation has produced a crucial reconsideration of the biology of the adult and aged mammalian myocardium. The classical notion of the adult heart as a static organ, in terms of cell turnover and renewal, has now been replaced by a dynamic model in which cardiac cells continuously die and are then replaced by CSC progeny differentiation. However, CSCs are not immortal. They undergo cellular senescence characterized by increased ROS production and oxidative stress and loss of telomere/telomerase integrity in response to a variety of physiological and pathological demands with aging. Nevertheless, the old myocardium preserves an endogenous functionally competent CSC cohort which appears to be resistant to the senescent phenotype occurring with aging. The latter envisions the phenomenon of CSC ageing as a result of a stochastic and therefore reversible cell autonomous process. However, CSC aging could be a programmed cell cycle-dependent process, which affects all or most of the endogenous CSC population. The latter would infer that the loss of CSC regenerative capacity with aging is an inevitable phenomenon that cannot be rescued by stimulating their growth, which would only speed their progressive exhaustion. The resolution of these two biological views will be crucial to design and develop effective CSC-based interventions to counteract cardiac aging not only improving health span of the elderly but also extending lifespan by delaying cardiovascular disease-related deaths.
Collapse
|
30
|
Role of NADPH oxidase in cooperative reactive oxygen species generation in dopaminergic neurons induced by combined treatment with dieldrin and lindane. Toxicol Lett 2018; 299:47-55. [DOI: 10.1016/j.toxlet.2018.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/31/2018] [Accepted: 09/16/2018] [Indexed: 11/19/2022]
|
31
|
Choi SA, Kim YH, Park YH, Yang HJ, Jeong PS, Cha JJ, Yoon SB, Kim JS, Song BS, Lee JH, Sim BW, Huh JW, Song IS, Lee SR, Kim MK, Kim JM, Bae YS, Imakawa K, Kim SU, Chang KT. Novel crosstalk between Vps26a and Nox4 signaling during neurogenesis. Cell Death Differ 2018; 26:1582-1599. [PMID: 30464227 PMCID: PMC6748115 DOI: 10.1038/s41418-018-0226-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/21/2018] [Accepted: 09/28/2018] [Indexed: 12/25/2022] Open
Abstract
Despite numerous studies on the molecular switches governing the conversion of stemness to differentiation in embryonic stem cells (ESCs), little is known about the involvement of the retromer complex. Under neural differentiation conditions, Vps26a deficiency (Vps26a-/-) or knockdown suppressed the loss of stemness and subsequent neurogenesis from ESCs or embryonic carcinoma cells, respectively, as evidenced by the long-lasting expression of stemness markers and the slow appearance of neuronal differentiation markers. Interestingly, relatively low reactive oxygen species (ROS) levels were generated during differentiation of Vps26a-/- ESCs, and treatment with an antioxidant or inhibitor of NADPH oxidase (Nox), a family of ROS-generating enzymes, led to restoration of stemness in wild-type cells to the level of Vps26a-/- cells during neurogenesis. Importantly, a novel interaction between Vps26a and Nox4 linked to the activation of ERK1/2 depended highly on ROS levels during neurogenesis, which were strongly suppressed in differentiating Vps26a-/- ESCs. Moreover, inhibition of phosphorylated ERK1/2 (pERK1/2) resulted in decreased ROS and Nox4 levels, indicating the mutual dependency between pERK1/2 and Nox4-derived ROS during neurogenesis. These results suggest that Vps26a regulates stemness by actively cooperating with the Nox4/ROS/ERK1/2 cascade during neurogenesis. Our findings have important implications for understanding the regulation of stemness via crosstalk between the retromer molecule and redox signaling, and may contribute to the development of ESC-based therapeutic strategies for the mass production of target cells.
Collapse
Affiliation(s)
- Seon-A Choi
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Laboratory of Animal Reproduction and Physiology, Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hae-Jun Yang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea
| | - Pil-Soo Jeong
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea
| | - Jae-Jin Cha
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea
| | - Seung-Bin Yoon
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea
| | - Ji-Su Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jong-Hee Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea
| | - Jae-Won Huh
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - In-Sung Song
- Department of Biomedical Sciences, College of Medicine, Ulsan University, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Sang-Rae Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Min-Kyu Kim
- Laboratory of Animal Reproduction and Physiology, Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jin-Man Kim
- College of Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Yun Soo Bae
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Kazuhiko Imakawa
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, 319-0206, Japan.,Institute of Agricultural Sciences, Tokai University, Kumamoto, 862-8652, Japan
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea. .,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Kyu-Tae Chang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, 28116, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
32
|
Rampon C, Volovitch M, Joliot A, Vriz S. Hydrogen Peroxide and Redox Regulation of Developments. Antioxidants (Basel) 2018; 7:E159. [PMID: 30404180 PMCID: PMC6262372 DOI: 10.3390/antiox7110159] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 01/16/2023] Open
Abstract
Reactive oxygen species (ROS), which were originally classified as exclusively deleterious compounds, have gained increasing interest in the recent years given their action as bona fide signalling molecules. The main target of ROS action is the reversible oxidation of cysteines, leading to the formation of disulfide bonds, which modulate protein conformation and activity. ROS, endowed with signalling properties, are mainly produced by NADPH oxidases (NOXs) at the plasma membrane, but their action also involves a complex machinery of multiple redox-sensitive protein families that differ in their subcellular localization and their activity. Given that the levels and distribution of ROS are highly dynamic, in part due to their limited stability, the development of various fluorescent ROS sensors, some of which are quantitative (ratiometric), represents a clear breakthrough in the field and have been adapted to both ex vivo and in vivo applications. The physiological implication of ROS signalling will be presented mainly in the frame of morphogenetic processes, embryogenesis, regeneration, and stem cell differentiation. Gain and loss of function, as well as pharmacological strategies, have demonstrated the wide but specific requirement of ROS signalling at multiple stages of these processes and its intricate relationship with other well-known signalling pathways.
Collapse
Affiliation(s)
- Christine Rampon
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- Sorbonne Paris Cité, Univ Paris Diderot, Biology Department, 75205 Paris CEDEX 13, France.
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- École Normale Supérieure, Department of Biology, PSL Research University, 75005 Paris, France.
| | - Alain Joliot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
| | - Sophie Vriz
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- Sorbonne Paris Cité, Univ Paris Diderot, Biology Department, 75205 Paris CEDEX 13, France.
| |
Collapse
|
33
|
Nugud A, Sandeep D, El-Serafi AT. Two faces of the coin: Minireview for dissecting the role of reactive oxygen species in stem cell potency and lineage commitment. J Adv Res 2018; 14:73-79. [PMID: 30023134 PMCID: PMC6047483 DOI: 10.1016/j.jare.2018.05.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/29/2022] Open
Abstract
Reactive oxygen species (ROS) are produced as by-products of several intracellular metabolic pathways and are reduced to more stable molecules by several protective pathways. The presence of high levels of ROS can be associated with disturbance of cell function and could lead to apoptosis. The presence of ROS within the physiological range has many effects on several signalling pathways. In stem cells, this role can range between keeping the potency of the naive stem cells to differentiation towards a certain lineage. In addition, the level of certain ROS would change according to the differentiation stage. For example, the presence of ROS can be associated with increasing the proliferation of mesenchymal stem cells, decreasing the potency of embryonic stem cells and adding to the genomic stability of induced pluripotent stem cells. ROS can enhance the differentiation of stem cells into cardiomyocytes, adipocytes, endothelial cells, keratinocytes and neurons. In the meantime, ROS inhibits osteogenesis and enhances the differentiation of cartilage to the hypertrophic stage, which is associated with chondrocyte death. Thus, ROS may form a link between naïve stem cells in the body and the environment. In addition, monitoring of ROS levels in vitro may help in tissue regeneration studies.
Collapse
Affiliation(s)
- Ahmed Nugud
- Sharjah Institute for Medical and Health Research, University of Sharjah, United Arab Emirates
| | - Divyasree Sandeep
- Sharjah Institute for Medical and Health Research, University of Sharjah, United Arab Emirates
| | - Ahmed T. El-Serafi
- Sharjah Institute for Medical and Health Research, University of Sharjah, United Arab Emirates
- Faculty of Medicine, Suez Canal University, Egypt
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| |
Collapse
|
34
|
Sthijns MMJPE, van Blitterswijk CA, LaPointe VLS. Redox regulation in regenerative medicine and tissue engineering: The paradox of oxygen. J Tissue Eng Regen Med 2018; 12:2013-2020. [PMID: 30044552 PMCID: PMC6221092 DOI: 10.1002/term.2730] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/07/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022]
Abstract
One of the biggest challenges in tissue engineering and regenerative medicine is to incorporate a functioning vasculature to overcome the consequences of a lack of oxygen and nutrients in the tissue construct. Otherwise, decreased oxygen tension leads to incomplete metabolism and the formation of the so‐called reactive oxygen species (ROS). Cells have many endogenous antioxidant systems to ensure a balance between ROS and antioxidants, but if this balance is disrupted by factors such as high levels of ROS due to long‐term hypoxia, there will be tissue damage and dysfunction. Current attempts to solve the oxygen problem in the field rarely take into account the importance of the redox balance and are instead centred on releasing or generating oxygen. The first problem with this approach is that although oxygen is necessary for life, it is paradoxically also a highly toxic molecule. Furthermore, although some oxygen‐generating biomaterials produce oxygen, they also generate hydrogen peroxide, a ROS, as an intermediate product. In this review, we discuss why it would be a superior strategy to supplement oxygen delivery with molecules to safeguard the important redox balance. Redox sensor proteins that can stimulate the anaerobic metabolism, angiogenesis, and enhancement of endogenous antioxidant systems are discussed as promising targets. We propose that redox regulating biomaterials have the potential to tackle some of the challenges related to angiogenesis and that the knowledge in this review will help scientists in tissue engineering and regenerative medicine realize this aim.
Collapse
Affiliation(s)
- Mireille M J P E Sthijns
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Clemens A van Blitterswijk
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Vanessa L S LaPointe
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
35
|
Tu C, Allen A, Deng W, Conroy O, Nambiar M, Zoldan J. Commonly used thiol-containing antioxidants reduce cardiac differentiation and alter gene expression ratios of sarcomeric isoforms. Exp Cell Res 2018; 370:150-159. [PMID: 29920245 DOI: 10.1016/j.yexcr.2018.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/07/2018] [Accepted: 06/15/2018] [Indexed: 01/31/2023]
Abstract
Reactive oxygen species (ROS) scavengers such as beta-mercaptoethanol (BME) and monothiol glycerol (MTG) are extensively used in stem cell research to prevent cellular oxidative stress. However, how these antioxidant supplements impact stem cell cardiac differentiation, a process regulated by redox-signaling remains unknown. In this study, we found that removal of BME from the conventional high-glucose, serum-based differentiation medium improved cardiac differentiation efficiency by 2-3 fold. BME and MTG treatments during differentiation significantly reduced mRNA expression of cardiac progenitor markers (NKX2.5 and ISL1) as well as sarcomeric markers (MLC2A, MLC2V, TNNI3, MYH6 and MYH7), suggesting reduced cardiomyogenesis by BME or MTG. Moreover, BME and MTG altered the expression ratios between the sarcomeric isoforms. In particular, TNNI3 to TNNI1 ratio and MLC2V to MLC2A ratio were significantly lower in BME or MTG treated cells than untreated cells, implying altered cardiomyocyte phenotype and maturity. Lastly, BME and MTG treatments resulted in less frequent beating, slower contraction and relaxation velocities than untreated cells. Interestingly, none of the above-mentioned effects was observed with Trolox, a non-thiol based antioxidant, despite its strong antioxidant activity. This work demonstrates that commonly used antioxidant supplements may cause considerable changes to cellular redox state and the outcome of differentiation.
Collapse
Affiliation(s)
- Chengyi Tu
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Alicia Allen
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Wei Deng
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Olivia Conroy
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Madhavi Nambiar
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Janet Zoldan
- Department of Biomedical Engineering, University of Texas at Austin, United States.
| |
Collapse
|
36
|
Nazem S, Rabiee F, Ghaedi K, Babashah S, Sadeghizadeh M, Nasr-Esfahani MH. Fndc5 knockdown induced suppression of mitochondrial integrity and significantly decreased cardiac differentiation of mouse embryonic stem cells. J Cell Biochem 2018; 119:4528-4539. [PMID: 29239022 DOI: 10.1002/jcb.26590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022]
Abstract
Fibronectin type III domain-containing 5 protein (Fndc5) is a glycosylated protein with elevated expression in high energy demanded tissues as heart, brain, and muscle. It has been shown that upregulation of Fndc5 is regulated by peroxisome proliferator-activated receptor-γ coactivator-1 alpha (PGC-1α), which is known as a master regulator of mitochondrial function and biogenesis. Also, our group indicated that Fndc5 expression increases gradually during cardiac differentiation of mouse embryonic stem cells (mESCs). In this paper, to clarify the importance of Fndc5 in cardiac differentiation, we south to knock down Fndc5 expression by generation a stably transduced mESC line that derives the expression of a short hairpin RNA (shRNA) against Fndc5 gene following doxycycline (Dox) induction. Knock-down of Fndc5 demonstrated a considerable decrease in expression of cardiac progenitor and cardiomyocyte markers. Considering the fact that mitochondria play a crucial role in cardiac differentiation of ESCs, we investigated the role of Fndc5, as a downstream target of PGC1-α, on mitochondrial indices. Results showed that expression of nuclear encoded mitochondrial genes including PGC1-α, Atp5b, Ndufb5, and SOD2 significantly decreased. Moreover, mitochondrial membrane potential (ΔΨm) and relative ATP content of cardiomyocytes decreased markedly with relative ROS level increase. Together, our results suggest that Fndc5 attenuates process of cardiac differentiation of mESCs which is associated with modulation of mitochondrial function and gene expression.
Collapse
Affiliation(s)
- Shima Nazem
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farzaneh Rabiee
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
37
|
Dural effects of oxidative stress on cardiomyogenesis via Gata4 transcription and protein ubiquitination. Cell Death Dis 2018; 9:246. [PMID: 29445146 PMCID: PMC5833852 DOI: 10.1038/s41419-018-0281-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/25/2017] [Accepted: 12/28/2017] [Indexed: 01/22/2023]
Abstract
Oxidative stress generates reactive oxygen species (ROS) that can promote or inhibit cardiac differentiation of stem cells dependent on the intensity of stimuli as well as cellular context in redox and differentiation status. In the current study, we confirmed that suitable intensity of hydrogen peroxide at the formation stage of embryoid bodies (EBs) effectively favored the formation of spontaneously beating cardiomyocytes from P19 embryonal carcinoma cells. Mechanistic studies implicated that extrinsic ROS enhanced the Caspase-mediated degradation of Oct4 and Nanog, two factors that governing pluripotent property. Further experiments suggested that a cohort of Nanog together with histone deacetylase 4 (Hdac4) played a critical role in establishing and maintaining the silent transcriptional status of Gata4 and Nkx2.5 in undifferentiated cells. Thus, an impulse of hydrogen peroxide depleted Nanog and Hdac4 via a caspase-dependent manner to ameliorate the repression on Gata4 and Nkx2.5 promoters, thereby generating a persistent activation on cardiac differentiation program. Meanwhile, we found that excessive ROS-activated JNK cascade to facilitate the ubiquitination and subsequent degradation of Gata4 protein. Overall, our results indicate that suitable ROS promotes the activation of Gata4 in transcription, while excessive ROS targets Gata4 protein for proteasome-dependent degradation. Gata4 is an important modulator balancing the promoting and inhibitory effects of oxidative stress on differentiation program of cardiomyogenesis.
Collapse
|
38
|
Wei H, Cong X. The effect of reactive oxygen species on cardiomyocyte differentiation of pluripotent stem cells. Free Radic Res 2018; 52:150-158. [PMID: 29258365 DOI: 10.1080/10715762.2017.1420184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The coordination of metabolic shift with genetic circuits is critical to cell specification, but the metabolic mechanisms that drive cardiac development are largely unknown. Reactive oxygen species (ROS) are not only the by-product of mitochondrial metabolism, but play a critical role in signalling cascade of cardiac development as a second messenger. Various levels of ROS appear differential and even oppose effect on selfrenewal and cardiac differentiation of pluripotent stem cells (PSCs) at each stage of differentiation. The intracellular ROS and redox balance are meticulous regulated by several systems of ROS generation and scavenging, among which mitochondria and the NADPH oxidase (NOX) are major sources of intracellular ROS involved in cardiomyocyte differentiation. Some critical signalling modulators are activated or inactivated by oxidation, suggesting ROS can be involved in regulation of cell fate through these downstream targets. In this review, the literatures about major sources of ROS, the effect of ROS level on cardiac differentiation of PSCs, as well as the underlying mechanism of ROS in the control of cardiac fate of PSC are summarised and discussed.
Collapse
Affiliation(s)
- Hua Wei
- a Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University , Charleston , SC , USA
| | - Xiangfeng Cong
- b Centre of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease , Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| |
Collapse
|
39
|
Herrero D, Tomé M, Cañón S, Cruz FM, Carmona RM, Fuster E, Roche E, Bernad A. Redox-dependent BMI1 activity drives in vivo adult cardiac progenitor cell differentiation. Cell Death Differ 2018; 25:809-822. [PMID: 29323265 DOI: 10.1038/s41418-017-0022-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 01/04/2023] Open
Abstract
Accumulation of reactive oxygen species (ROS) is associated with several cardiovascular pathologies and with cell cycle exit by neonanatal cardiomyocytes, a key limiting factor in the regenerative capacity of the adult mammalian heart. The polycomb complex component BMI1 is linked to adult progenitors and is an important partner in DNA repair and redox regulation. We found that high BMI1 expression is associated with an adult Sca1+ cardiac progenitor sub-population with low ROS levels. In homeostasis, BMI1 repressed cell fate genes, including a cardiogenic differentiation program. Oxidative damage nonetheless modified BMI1 activity in vivo by derepressing canonical target genes in favor of their antioxidant and anticlastogenic functions. This redox-mediated mechanism is not restricted to damage situations, however, and we report ROS-associated differentiation of cardiac progenitors in steady state. These findings demonstrate how redox status influences the cardiac progenitor response, and identify redox-mediated BMI1 regulation with implications in maintenance of cellular identity in vivo.
Collapse
Affiliation(s)
- Diego Herrero
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - María Tomé
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Susana Cañón
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Francisco M Cruz
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Rosa María Carmona
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Encarna Fuster
- Department of Applied Biology-Nutrition and Institute of Bioengineering, University Miguel Hernández, Institute for Health and Biomedical Research (ISABIAL-FISABIO Fundation), Alicante, Spain
| | - Enrique Roche
- CIBERobn (Physiopathology of Obesity and Nutrition CB12/03/30038), Carlos III Health Research Institute (ISCIII), Madrid, Spain.,Department of Applied Biology-Nutrition and Institute of Bioengineering, University Miguel Hernández, Institute for Health and Biomedical Research (ISABIAL-FISABIO Fundation), Alicante, Spain
| | - Antonio Bernad
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain. .,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
| |
Collapse
|
40
|
Razaghi B, Steele SL, Prykhozhij SV, Stoyek MR, Hill JA, Cooper MD, McDonald L, Lin W, Daugaard M, Crapoulet N, Chacko S, Lewis SM, Scott IC, Sorensen PHB, Berman JN. hace1 Influences zebrafish cardiac development via ROS-dependent mechanisms. Dev Dyn 2017; 247:289-303. [PMID: 29024245 DOI: 10.1002/dvdy.24600] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/23/2017] [Accepted: 09/15/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In this study, we reveal a previously undescribed role of the HACE1 (HECT domain and Ankyrin repeat Containing E3 ubiquitin-protein ligase 1) tumor suppressor protein in normal vertebrate heart development using the zebrafish (Danio rerio) model. We examined the link between the cardiac phenotypes associated with hace1 loss of function to the expression of the Rho small family GTPase, rac1, which is a known target of HACE1 and promotes ROS production via its interaction with NADPH oxidase holoenzymes. RESULTS We demonstrate that loss of hace1 in zebrafish via morpholino knockdown results in cardiac deformities, specifically a looping defect, where the heart is either tubular or "inverted". Whole-mount in situ hybridization of cardiac markers shows distinct abnormalities in ventricular morphology and atrioventricular valve formation in the hearts of these morphants, as well as increased expression of rac1. Importantly, this phenotype appears to be directly related to Nox enzyme-dependent ROS production, as both genetic inhibition by nox1 and nox2 morpholinos or pharmacologic rescue using ROS scavenging agents restores normal cardiac structure. CONCLUSIONS Our study demonstrates that HACE1 is critical in the normal development and proper function of the vertebrate heart via a ROS-dependent mechanism. Developmental Dynamics 247:289-303, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Babak Razaghi
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Shelby L Steele
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sergey V Prykhozhij
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Matthew R Stoyek
- Department of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jessica A Hill
- Department of Marine Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Matthew D Cooper
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lindsay McDonald
- Department of Emergency Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - William Lin
- Undergraduate Program, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mads Daugaard
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada.,Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Simi Chacko
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Stephen M Lewis
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Ian C Scott
- Department of Molecular Genetics, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Poul H B Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason N Berman
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada.,IWK Health Centre, Halifax, Nova Scotia, Canada
| |
Collapse
|
41
|
Cardiac Aging – Benefits of Exercise, Nrf2 Activation and Antioxidant Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 999:231-255. [DOI: 10.1007/978-981-10-4307-9_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
Adam AAA, van Wenum M, van der Mark VA, Jongejan A, Moerland PD, Houtkooper RH, Wanders RJA, Oude Elferink RP, Chamuleau RAFM, Hoekstra R. AMC-Bio-Artificial Liver culturing enhances mitochondrial biogenesis in human liver cell lines: The role of oxygen, medium perfusion and 3D configuration. Mitochondrion 2017; 39:30-42. [PMID: 28844938 DOI: 10.1016/j.mito.2017.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/15/2017] [Accepted: 08/23/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Human liver cell lines, like HepaRG and C3A, acquire higher functionality when cultured in the AMC-Bio-Artificial Liver (AMC-BAL). The three main differences between BAL and monolayer culture are the oxygenation (40% vs 20%O2), dynamic vs absent medium perfusion and 3D vs 2D configuration. Here, we investigated the background of the differences between BAL-cultures and monolayers. METHODS We performed whole-genome microarray analysis on HepaRG monolayer and BAL-cultures. Next, mitochondrial biogenesis was studied in monolayer and BAL-cultures of HepaRG and C3A. The driving forces for mitochondrial biogenesis by BAL-culturing were investigated in representative culture models differing in oxygenation level, medium flow or 2D vs 3D configuration. RESULTS Gene-sets related to mitochondrial energy metabolism were most prominently up-regulated in HepaRG-BAL vs monolayer cultures. This was confirmed by a 2.4-fold higher mitochondrial abundance with increased expression of mitochondrial OxPhos complexes. Moreover, the transcript levels of mitochondria-encoded genes were up to 3.6-fold induced and mitochondrial membrane potential activity was 8.3-fold increased in BAL vs monolayers. Culturing with 40% O2, dynamic medium flow and/or in 3D increased the mitochondrial abundance and expression of mitochondrial complexes vs standard monolayer culturing. The stimulatory effect of the BAL culture on mitochondrial biogenesis was confirmed in C3A cells in which mitochondrial abundance increased 2.2-fold with induction of mitochondria-encoded genes. CONCLUSIONS AND GENERAL SIGNIFICANCE The increased functionality of liver cell lines upon AMC-BAL culturing is associated with increased mitochondrial biogenesis. High oxygenation, medium perfusion and 3D configuration contribute to the up-regulation of the mitochondrial biogenesis.
Collapse
Affiliation(s)
- Aziza A A Adam
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Martien van Wenum
- Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Vincent A van der Mark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands; Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health Research Institute, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Perry D Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health Research Institute, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic and Metabolic Diseases, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic and Metabolic Diseases, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Ronald P Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Robert A F M Chamuleau
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands; Surgical Laboratory, Academic Medical Center (AMC), University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Gibson GE, Thakkar A. Mitochondria/metabolic reprogramming in the formation of neurons from peripheral cells: Cause or consequence and the implications to their utility. Neurochem Int 2017. [PMID: 28627365 DOI: 10.1016/j.neuint.2017.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The induction of pluripotent stem cells (iPSC) from differentiated cells such as fibroblasts and their subsequent conversion to neural progenitor cells (NPC) and finally to neurons is intriguing scientifically, and its potential to medicine is nearly infinite, but unrealized. A better understanding of the changes at each step of the transformation will enable investigators to better model neurological disease. Each step of conversion from a differentiated cell to an iPSC to a NPC to neurons requires large changes in glycolysis including aerobic glycolysis, the pentose shunt, the tricarboxylic acid cycle, the electron transport chain and in the production of reactive oxygen species (ROS). These mitochondrial/metabolic changes are required and their manipulation modifies conversions. These same mitochondrial/metabolic processes are altered in common neurological diseases so that factors related to the disease may alter the cellular transformation at each step including the final phenotype. A lack of understanding of these interactions could compromise the validity of the disease comparisons in iPSC derived neurons. Both the complexity and potential of iPSC derived cells for understanding and treating disease remain great.
Collapse
Affiliation(s)
- Gary E Gibson
- Weil Cornell Medicine, Brain and Mind Research Institute, Burke Medical Research, White Plains, NY 10605, United States.
| | - Ankita Thakkar
- Weil Cornell Medicine, Brain and Mind Research Institute, Burke Medical Research, White Plains, NY 10605, United States
| |
Collapse
|
44
|
Lingan JV, Alanzalon RE, Porter GA. Preventing permeability transition pore opening increases mitochondrial maturation, myocyte differentiation and cardiac function in the neonatal mouse heart. Pediatr Res 2017; 81:932-941. [PMID: 28141792 DOI: 10.1038/pr.2017.19] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/15/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND In embryonic myocytes, closure of the mitochondrial permeability transition pore (PTP) drives mitochondrial maturation and cardiac myocyte differentiation. Since neonatal cardiac myocytes remain relatively immature, we hypothesized that inducing PTP closure at this age, by inhibiting the PTP regulator, cyclophilin D (CyPD), genetically or with Cyclosporin A (CsA) and NIM811, would increase cardiac function by increasing mitochondrial maturation and myocyte differentiation. METHODS Cultured neonatal myocytes or neonatal mice were treated for 5 d with vehicle, CsA or NIM811. Mitochondrial function and structure were measured in vitro. Myocyte differentiation was assessed by immunolabeling for contractile proteins. Cardiac function was determined using echocardiography. RESULTS The probability of PTP opening was high in WT neonatal myocytes. Treatment with CsA or NIM811 in vitro increased mitochondrial structural complexity and membrane potential, decreased reactive oxygen species levels, and increased myocyte differentiation. WT mice treated with either CsA or NIM811 in vivo for the first 5 d of life had higher ejection fractions. Deleting CyPD had similar effects as CsA and NIM811 on all parameters. CONCLUSIONS It may be feasible to inhibit the PTP using available drugs to increase mitochondrial maturation, myocyte differentiation, and cardiac function in neonates.
Collapse
Affiliation(s)
- Jayson V Lingan
- Current affiliation: Department of Pediatrics, Benefits Health System, Great Falls, Montana.,Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Ryan E Alanzalon
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - George A Porter
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York.,Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
45
|
Nayernia Z, Colaianna M, Robledinos-Antón N, Gutzwiller E, Sloan-Béna F, Stathaki E, Hibaoui Y, Cuadrado A, Hescheler J, Stasia MJ, Saric T, Jaquet V, Krause KH. Decreased neural precursor cell pool in NADPH oxidase 2-deficiency: From mouse brain to neural differentiation of patient derived iPSC. Redox Biol 2017; 13:82-93. [PMID: 28575744 PMCID: PMC5454143 DOI: 10.1016/j.redox.2017.04.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 10/28/2022] Open
Abstract
There is emerging evidence for the involvement of reactive oxygen species (ROS) in the regulation of stem cells and cellular differentiation. Absence of the ROS-generating NADPH oxidase NOX2 in chronic granulomatous disease (CGD) patients, predominantly manifests as immune deficiency, but has also been associated with decreased cognition. Here, we investigate the role of NOX enzymes in neuronal homeostasis in adult mouse brain and in neural cells derived from human induced pluripotent stem cells (iPSC). High levels of NOX2 were found in mouse adult neurogenic regions. In NOX2-deficient mice, neurogenic regions showed diminished redox modifications, as well as decrease in neuroprecursor numbers and in expression of genes involved in neural differentiation including NES, BDNF and OTX2. iPSC from healthy subjects and patients with CGD were used to study the role of NOX2 in human in vitro neuronal development. Expression of NOX2 was low in undifferentiated iPSC, upregulated upon neural induction, and disappeared during neuronal differentiation. In human neurospheres, NOX2 protein and ROS generation were polarized within the inner cell layer of rosette structures. NOX2 deficiency in CGD-iPSCs resulted in an abnormal neural induction in vitro, as revealed by a reduced expression of neuroprogenitor markers (NES, BDNF, OTX2, NRSF/REST), and a decreased generation of mature neurons. Vector-mediated NOX2 expression in NOX2-deficient iPSCs rescued neurogenesis. Taken together, our study provides novel evidence for a regulatory role of NOX2 during early stages of neurogenesis in mouse and human.
Collapse
Affiliation(s)
- Zeynab Nayernia
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Marilena Colaianna
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Natalia Robledinos-Antón
- Instituto de Investigaciones Biomédicas "Alberto Sols", Faculty of Medicine, Autonomous University of Madrid (UAM), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eveline Gutzwiller
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Frédérique Sloan-Béna
- Hôpitaux Universitaires de Genève HUG, Laboratoires de Cytogénétique Constitutionnelle, Service de Médecine Génétique, Geneva, Switzerland
| | - Elisavet Stathaki
- Hôpitaux Universitaires de Genève HUG, Laboratoires de Cytogénétique Constitutionnelle, Service de Médecine Génétique, Geneva, Switzerland
| | - Yousef Hibaoui
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas "Alberto Sols", Faculty of Medicine, Autonomous University of Madrid (UAM), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Marie-José Stasia
- Université Grenoble Alpes, Techniques de l'Ingénierie Médicale et de la Complexité- Grenoble, F38000 Grenoble, France
| | - Tomo Saric
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Vincent Jaquet
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland.
| |
Collapse
|
46
|
Sun L, Moritake T, Ito K, Matsumoto Y, Yasui H, Nakagawa H, Hirayama A, Inanami O, Tsuboi K. Metabolic analysis of radioresistant medulloblastoma stem-like clones and potential therapeutic targets. PLoS One 2017; 12:e0176162. [PMID: 28426747 PMCID: PMC5398704 DOI: 10.1371/journal.pone.0176162] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022] Open
Abstract
Medulloblastoma is a fatal brain tumor in children, primarily due to the presence of treatment-resistant medulloblastoma stem cells. The energy metabolic pathway is a potential target of cancer therapy because it is often different between cancer cells and normal cells. However, the metabolic properties of medulloblastoma stem cells, and whether specific metabolic pathways are essential for sustaining their stem cell-like phenotype and radioresistance, remain unclear. We have established radioresistant medulloblastoma stem-like clones (rMSLCs) by irradiation of the human medulloblastoma cell line ONS-76. Here, we assessed reactive oxygen species (ROS) production, mitochondria function, oxygen consumption rate (OCR), energy state, and metabolites of glycolysis and tricarboxylic acid cycle in rMSLCs and parental cells. rMSLCs showed higher lactate production and lower oxygen consumption rate than parental cells. Additionally, rMSLCs had low mitochondria mass, low endogenous ROS production, and existed in a low-energy state. Treatment with the metabolic modifier dichloroacetate (DCA) resulted in mitochondria dysfunction, glycolysis inhibition, elongated mitochondria morphology, and increased ROS production. DCA also increased radiosensitivity by suppression of the DNA repair capacity through nuclear oxidization and accelerated the generation of acetyl CoA to compensate for the lack of ATP. Moreover, treatment with DCA decreased cancer stem cell-like characters (e.g., CD133 positivity and sphere-forming ability) in rMSLCs. Together, our findings provide insights into the specific metabolism of rMSLCs and illuminate potential metabolic targets that might be exploited for therapeutic benefit in medulloblastoma.
Collapse
Affiliation(s)
- Lue Sun
- Department of Radiological Health Science, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Takashi Moritake
- Department of Radiological Health Science, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- * E-mail:
| | - Kazuya Ito
- Department of Radiobiology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshitaka Matsumoto
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hironobu Yasui
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hidehiko Nakagawa
- Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Aki Hirayama
- Center for Integrative Medicine, Tsukuba University of Technology, Tsukuba, Ibaraki, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Koji Tsuboi
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
47
|
Huang Y, Hong H, Li M, Liu J, Jiang C, Zhang H, Ye L, Zheng J. Age-Dependent Oxidative DNA Damage Does Not Correlate with Reduced Proliferation of Cardiomyocytes in Humans. PLoS One 2017; 12:e0170351. [PMID: 28099512 PMCID: PMC5242470 DOI: 10.1371/journal.pone.0170351] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 01/03/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Postnatal human cardiomyocyte proliferation declines rapidly with age, which has been suggested to be correlated with increases in oxidative DNA damage in mice and plays an important role in regulating cardiomyocyte proliferation. However, the relationship between oxidative DNA damage and age in humans is unclear. METHODS Sixty right ventricular outflow myocardial tissue specimens were obtained from ventricular septal defect infant patients during routine congenital cardiac surgery. These specimens were divided into three groups based on age: group A (age 0-6 months), group B (age, 7-12 months), and group C (>12 months). Each tissue specimen was subjected to DNA extraction, RNA extraction, and immunofluorescence. RESULTS Immunofluorescence and qRT-PCR analysis revealed that DNA damage markers-mitochondrial DNA copy number, oxoguanine 8, and phosphorylated ataxia telangiectasia mutated-were highest in Group B. However immunofluorescence and qRT-PCR demonstrated that two cell proliferation markers, Ki67 and cyclin D2, were decreased with age. In addition, wheat germ agglutinin-staining indicated that the average size of cardiomyocytes increased with age. CONCLUSIONS Oxidative DNA damage of cardiomyocytes was not correlated positively with age in human beings. Oxidative DNA damage is unable to fully explain the reduced proliferation of human cardiomyocytes.
Collapse
Affiliation(s)
- Yanhui Huang
- Department of anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haifa Hong
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Congenital Heart Diseases, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minghui Li
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Congenital Heart Diseases, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinfen Liu
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Congenital Heart Diseases, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chuan Jiang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Congenital Heart Diseases, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haibo Zhang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Congenital Heart Diseases, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (LY); (JZ)
| | - Jinghao Zheng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (LY); (JZ)
| |
Collapse
|
48
|
Foglia MJ, Poss KD. Building and re-building the heart by cardiomyocyte proliferation. Development 2016; 143:729-40. [PMID: 26932668 DOI: 10.1242/dev.132910] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adult human heart does not regenerate significant amounts of lost tissue after injury. Rather than making new, functional muscle, human hearts are prone to scarring and hypertrophy, which can often lead to fatal arrhythmias and heart failure. The most-cited basis of this ineffective cardiac regeneration in mammals is the low proliferative capacity of adult cardiomyocytes. However, mammalian cardiomyocytes can avidly proliferate during fetal and neonatal development, and both adult zebrafish and neonatal mice can regenerate cardiac muscle after injury, suggesting that latent regenerative potential exists. Dissecting the cellular and molecular mechanisms that promote cardiomyocyte proliferation throughout life, deciphering why proliferative capacity normally dissipates in adult mammals, and deriving means to boost this capacity are primary goals in cardiovascular research. Here, we review our current understanding of how cardiomyocyte proliferation is regulated during heart development and regeneration.
Collapse
Affiliation(s)
- Matthew J Foglia
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
49
|
Park J, Park JW, Oh H, Maria FS, Kang J, Tian X. Gene-Specific Assessment of Guanine Oxidation as an Epigenetic Modulator for Cardiac Specification of Mouse Embryonic Stem Cells. PLoS One 2016; 11:e0155792. [PMID: 27249188 PMCID: PMC4889044 DOI: 10.1371/journal.pone.0155792] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 05/03/2016] [Indexed: 12/11/2022] Open
Abstract
Epigenetics have essential roles in development and human diseases. Compared to the complex histone modifications, epigenetic changes on mammalian DNA are as simple as methylation on cytosine. Guanine, however, can be oxidized as an epigenetic change which can undergo base-pair transversion, causing a genetic difference. Accumulating evidence indicates that reactive oxygen species (ROS) are important signaling molecules for embryonic stem cell (ESC) differentiation, possibly through transient changes on genomic DNA such as 7,8-dihydro-8-oxoguanine (8-oxoG). Technical limitations on detecting such DNA modifications, however, restrict the investigation of the role of 8-oxoG in ESC differentiation. Here, we developed a Hoogsteen base pairing-mediated PCR-sequencing assay to detect 8-oxoG lesions that can subsequently cause G to T transversions during PCR. We then used this assay to assess the epigenetic and transient 8-oxoG formation in the Tbx5 gene of R1 mouse ESCs subjected to oxidative stress by removing 2-mercaptoethanol (2ME) from the culture media. To our surprise, significantly higher numbers of 8-oxoG-mediated G∙C to C∙G transversion, not G∙C to T∙A, were detected at 7th and 9th base position from the transcription start site of exon 1 of Tbx5 in ESCs in the (-)2ME than (+)2ME group (p < 0.05). This was consistent with the decrease in the amount of amplifiable of DNA harboring the 8-oxoG lesions at the Tbx5 promoter region in the oxidative stressed ESCs. The ESCs responded to oxidative stress, possibly through the epigenetic effects of guanine oxidation with decreased proliferation (p < 0.05) and increased formation of beating embryoid bodies (EBs; p < 0.001). Additionally, the epigenetic changes of guanine induced up-regulation of Ogg1 and PolB, two base excision repairing genes for 8-oxoG, in ESCs treated with (-)2ME (p < 0.01). Together, we developed a gene-specific and direct quantification assay for guanine oxidation. Using oxidative stressed mouse ESCs, we validated this assay and assessed the epigenetic effects of 8-oxoG by studying expression of DNA repair genes, ESC proliferation, and EB formation.
Collapse
Affiliation(s)
- Joonghoon Park
- Center for Regenerative Biology and Department of Animal Science, University of Connecticut, Storrs, Connecticut, 06269, United States of America
| | - Jong Woo Park
- Research Center for Epigenome Regulation, School of Pharmacy, Sungkyunkwan University, Suwon, 440746, Republic of Korea
| | - Hawmok Oh
- Research Center for Epigenome Regulation, School of Pharmacy, Sungkyunkwan University, Suwon, 440746, Republic of Korea
| | - Fernanda S Maria
- Center for Regenerative Biology and Department of Animal Science, University of Connecticut, Storrs, Connecticut, 06269, United States of America
- Department of Animal Reproduction, College of Veterinary Medicine, University of Sao Paulo, Sao Paulo, 05508, Brazil
| | - Jaeku Kang
- Department of Pharmacology, College of Medicine, Konyang University, Daejeon, 302718, Republic of Korea
| | - Xiuchun Tian
- Center for Regenerative Biology and Department of Animal Science, University of Connecticut, Storrs, Connecticut, 06269, United States of America
| |
Collapse
|
50
|
Liang J, Wu SY, Zhang D, Wang L, Leung KK, Leung PS. NADPH Oxidase-Dependent Reactive Oxygen Species Stimulate β-Cell Regeneration Through Differentiation of Endocrine Progenitors in Murine Pancreas. Antioxid Redox Signal 2016; 24:419-33. [PMID: 26464216 DOI: 10.1089/ars.2014.6135] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Reactive oxygen species (ROS) act as second messengers for redox modification of transcription factors essential for differentiation. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a major source of ROS, has been shown to regulate differentiation of various progenitor cells, while its role in pancreatic endocrine cell differentiation is unclear. This study was aimed at this knowledge gap. RESULTS Our results showed that ROS levels were dynamically changed during pancreas development concomitant with endocrine cell differentiation induced by modest exogenous ROS in rudiment cultures. NOX4, but not NOX2, the member of NADPH oxidase, was expressed persistently in endocrine lineage and showed high activity in critical pancreas development phase. Inhibition of NADPH oxidase activity impeded the differentiation of endocrine progenitors in vitro, and exogenous ROS reversed this effect. Studies performed in streptozotocin (STZ)-injected neonatal rats showed that diphenyleneiodonium (DPI) obstructed β-cell regeneration through the suppression of neurogenin 3 (NGN3) expression, but not Ki67-labeling β-cells, indicating that ROS stimulation promoted differentiation beyond proliferation of β-cells. Inhibition of NADPH oxidase also reduced expression of SRY (sex-determining region Y)-box 9 (SOX9), a transcriptional regulator of Ngn3, in endocrine precursor cells, both in vivo and in vitro. Overexpression of SOX9 attenuated the reduction of NGN3 induced by suppression of NADPH oxidase. INNOVATION AND CONCLUSION This is the first study to demonstrate NADPH oxidase, especially NOX4-dependent ROS that promotes pancreatic progenitor cell differentiation into endocrine cells both in vitro and in vivo, probably through the regulation of SOX9. We provide evidence that NADPH oxidase-dependent ROS-mediated signaling is necessary for endocrine cell differentiation, which provides a potential strategy for efficient generation of insulin-producing cells in clinical application.
Collapse
Affiliation(s)
- Juan Liang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Shang Ying Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Dan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Lin Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Kwan Keung Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| |
Collapse
|