51
|
Suliman HB, Zobi F, Piantadosi CA. Heme Oxygenase-1/Carbon Monoxide System and Embryonic Stem Cell Differentiation and Maturation into Cardiomyocytes. Antioxid Redox Signal 2016; 24:345-60. [PMID: 26725491 PMCID: PMC4779979 DOI: 10.1089/ars.2015.6342] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS The differentiation of embryonic stem (ES) cells into energetically efficient cardiomyocytes contributes to functional cardiac repair and is envisioned to ameliorate progressive degenerative cardiac diseases. Advanced cell maturation strategies are therefore needed to create abundant mature cardiomyocytes. In this study, we tested whether the redox-sensitive heme oxygenase-1/carbon monoxide (HO-1/CO) system, operating through mitochondrial biogenesis, acts as a mechanism for ES cell differentiation and cardiomyocyte maturation. RESULTS Manipulation of HO-1/CO to enhance mitochondrial biogenesis demonstrates a direct pathway to ES cell differentiation and maturation into beating cardiomyocytes that express adult structural markers. Targeted HO-1/CO interventions up- and downregulate specific cardiogenic transcription factors, transcription factor Gata4, homeobox protein Nkx-2.5, heart- and neural crest derivatives-expressed protein 1, and MEF2C. HO-1/CO overexpression increases cardiac gene expression for myosin regulatory light chain 2, atrial isoform, MLC2v, ANP, MHC-β, and sarcomere α-actinin and the major mitochondrial fusion regulators, mitofusin 2 and MICOS complex subunit Mic60. This promotes structural mitochondrial network expansion and maturation, thereby supporting energy provision for beating embryoid bodies. These effects are prevented by silencing HO-1 and by mitochondrial reactive oxygen species scavenging, while disruption of mitochondrial biogenesis and mitochondrial DNA depletion by loss of mitochondrial transcription factor A compromise infrastructure. This leads to failure of cardiomyocyte differentiation and maturation and contractile dysfunction. INNOVATION The capacity to augment cardiomyogenesis via a defined mitochondrial pathway has unique therapeutic potential for targeting ES cell maturation in cardiac disease. CONCLUSION Our findings establish the HO-1/CO system and redox regulation of mitochondrial biogenesis as essential factors in ES cell differentiation as well as in the subsequent maturation of these cells into functional cardiac cells.
Collapse
Affiliation(s)
- Hagir B Suliman
- 1 Department of Medicine, Duke University School of Medicine , Durham, North Carolina.,2 Department of Anesthesiology, Duke University School of Medicine , Durham, North Carolina.,3 Department of Pathology, Duke University School of Medicine , Durham, North Carolina
| | - Fabio Zobi
- 4 Department of Chemistry, University of Fribourg , Fribourg, Switzerland
| | - Claude A Piantadosi
- 1 Department of Medicine, Duke University School of Medicine , Durham, North Carolina.,2 Department of Anesthesiology, Duke University School of Medicine , Durham, North Carolina.,3 Department of Pathology, Duke University School of Medicine , Durham, North Carolina
| |
Collapse
|
52
|
Lewandowski J, Kolanowski TJ, Kurpisz M. Techniques for the induction of human pluripotent stem cell differentiation towards cardiomyocytes. J Tissue Eng Regen Med 2016; 11:1658-1674. [PMID: 26777594 DOI: 10.1002/term.2117] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 09/16/2015] [Accepted: 11/18/2015] [Indexed: 01/04/2023]
Abstract
The derivation of pluripotent stem cells from human embryos and the generation of induced pluripotent stem cells (iPSCs) from somatic cells opened a new chapter in studies on the regeneration of the post-infarction heart and regenerative medicine as a whole. Thus, protocols for obtaining iPSCs were enthusiastically adopted and widely used for further experiments on cardiac differentiation. iPSC-mediated cardiomyocytes (iPSC-CMs) under in vitro culture conditions are generated by simulating natural cardiomyogenesis and involve the wingless-type mouse mammary tumour virus integration site family (WNT), transforming growth factor beta (TGF-β) and fibroblast growth factor (FGF) signalling pathways. New strategies have been proposed to take advantage of small chemical molecules, organic compounds and even electric or mechanical stimulation. There are three main approaches to support cardiac commitment in vitro: embryoid bodis (EBs), monolayer in vitro cultures and inductive co-cultures with visceral endoderm-like (END2) cells. In EB technique initial uniform size of pluripotent stem cell (PSC) colonies has a pivotal significance. Hence, some methods were designed to support cells aggregation. Another well-suited procedure is based on culturing cells in monolayer conditions in order to improve accessibility of growth factors and nutrients. Other distinct tactics are using visceral endoderm-like cells to culture them with PSCs due to secretion of procardiac cytokines. Finally, the appropriate purification of the obtained cardiomyocytes is required prior to their administration to a patient under the prospective cellular therapy strategy. This goal can be achieved using non-genetic methods, such as the application of surface markers and fluorescent dyes. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jarosław Lewandowski
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz J Kolanowski
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Maciej Kurpisz
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
53
|
Thrivikraman G, Madras G, Basu B. Electrically driven intracellular and extracellular nanomanipulators evoke neurogenic/cardiomyogenic differentiation in human mesenchymal stem cells. Biomaterials 2016; 77:26-43. [DOI: 10.1016/j.biomaterials.2015.10.078] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 10/26/2015] [Accepted: 10/29/2015] [Indexed: 12/27/2022]
|
54
|
Apocynin and Diphenyleneiodonium Induce Oxidative Stress and Modulate PI3K/Akt and MAPK/Erk Activity in Mouse Embryonic Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:7409196. [PMID: 26788250 PMCID: PMC4691611 DOI: 10.1155/2016/7409196] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/13/2015] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species (ROS) are important regulators of cellular functions. In embryonic stem cells, ROS are suggested to influence differentiation status. Regulated ROS formation is catalyzed primarily by NADPH-dependent oxidases (NOXs). Apocynin and diphenyleneiodonium are frequently used inhibitors of NOXs; however, both exhibit uncharacterized effects not related to NOXs inhibition. Interestingly, in our model of mouse embryonic stem cells we demonstrate low expression of NOXs. Therefore we aimed to clarify potential side effects of these drugs. Both apocynin and diphenyleneiodonium impaired proliferation of cells. Surprisingly, we observed prooxidant activity of these drugs determined by hydroethidine. Further, we revealed that apocynin inhibits PI3K/Akt pathway with its downstream transcriptional factor Nanog. Opposite to this, apocynin augmented activity of canonical Wnt signaling. On the contrary, diphenyleneiodonium activated both PI3K/Akt and Erk signaling pathways without affecting Wnt. Our data indicates limits and possible unexpected interactions of NOXs inhibitors with intracellular signaling pathways.
Collapse
|
55
|
Critical Roles of Reactive Oxygen Species in Age-Related Impairment in Ischemia-Induced Neovascularization by Regulating Stem and Progenitor Cell Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:7095901. [PMID: 26697140 PMCID: PMC4677240 DOI: 10.1155/2016/7095901] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/12/2015] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) regulate bone marrow microenvironment for stem and progenitor cells functions including self-renewal, differentiation, and cell senescence. In response to ischemia, ROS also play a critical role in mediating the mobilization of endothelial progenitor cells (EPCs) from the bone marrow to the sites of ischemic injury, which contributes to postnatal neovascularization. Aging is an unavoidable biological deteriorative process with a progressive decline in physiological functions. It is associated with increased oxidative stress and impaired ischemia-induced neovascularization. This review discusses the roles of ROS in regulating stem and progenitor cell function, highlighting the impact of unbalanced ROS levels on EPC dysfunction and the association with age-related impairment in ischemia-induced neovascularization. Furthermore, it discusses strategies that modulate the oxidative levels of stem and progenitor cells to enhance the therapeutic potential for elderly patients with cardiovascular disease.
Collapse
|
56
|
Nuclear Nox4 Role in Stemness Power of Human Amniotic Fluid Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:101304. [PMID: 26273418 PMCID: PMC4529982 DOI: 10.1155/2015/101304] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 11/25/2022]
Abstract
Human amniotic fluid stem cells (AFSC) are an attractive source for cell therapy due to their multilineage differentiation potential and accessibility advantages. However the clinical application of human stem cells largely depends on their capacity to expand in vitro, since there is an extensive donor-to-donor heterogeneity. Reactive oxygen species (ROS) and cellular oxidative stress are involved in many physiological and pathophysiological processes of stem cells, including pluripotency, proliferation, differentiation, and stress resistance. The mode of action of ROS is also dependent on the localization of their target molecules. Thus, the modifications induced by ROS can be separated depending on the cellular compartments they affect. NAD(P)H oxidase family, particularly Nox4, has been known to produce ROS in the nucleus. In the present study we show that Nox4 nuclear expression (nNox4) depends on the donor and it correlates with the expression of transcription factors involved in stemness regulation, such as Oct4, SSEA-4, and Sox2. Moreover nNox4 is linked with the nuclear localization of redox sensitive transcription factors, as Nrf2 and NF-κB, and with the differentiation potential. Taken together, these results suggest that nNox4 regulation may have important effects in stem cell capability through modulation of transcription factors and DNA damage.
Collapse
|
57
|
Wanet A, Arnould T, Najimi M, Renard P. Connecting Mitochondria, Metabolism, and Stem Cell Fate. Stem Cells Dev 2015; 24:1957-71. [PMID: 26134242 PMCID: PMC4543487 DOI: 10.1089/scd.2015.0117] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As sites of cellular respiration and energy production, mitochondria play a central role in cell metabolism. Cell differentiation is associated with an increase in mitochondrial content and activity and with a metabolic shift toward increased oxidative phosphorylation activity. The opposite occurs during reprogramming of somatic cells into induced pluripotent stem cells. Studies have provided evidence of mitochondrial and metabolic changes during the differentiation of both embryonic and somatic (or adult) stem cells (SSCs), such as hematopoietic stem cells, mesenchymal stem cells, and tissue-specific progenitor cells. We thus propose to consider those mitochondrial and metabolic changes as hallmarks of differentiation processes. We review how mitochondrial biogenesis, dynamics, and function are directly involved in embryonic and SSC differentiation and how metabolic and sensing pathways connect mitochondria and metabolism with cell fate and pluripotency. Understanding the basis of the crosstalk between mitochondria and cell fate is of critical importance, given the promising application of stem cells in regenerative medicine. In addition to the development of novel strategies to improve the in vitro lineage-directed differentiation of stem cells, understanding the molecular basis of this interplay could lead to the identification of novel targets to improve the treatment of degenerative diseases.
Collapse
Affiliation(s)
- Anaïs Wanet
- 1 Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur) , Namur, Belgium
| | - Thierry Arnould
- 1 Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur) , Namur, Belgium
| | - Mustapha Najimi
- 2 Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Clinique et Expérimentale (IREC), Université Catholique de Louvain , Brussels, Belgium
| | - Patricia Renard
- 1 Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur) , Namur, Belgium
| |
Collapse
|
58
|
Ryu JM, Lee HJ, Jung YH, Lee KH, Kim DI, Kim JY, Ko SH, Choi GE, Chai II, Song EJ, Oh JY, Lee SJ, Han HJ. Regulation of Stem Cell Fate by ROS-mediated Alteration of Metabolism. Int J Stem Cells 2015; 8:24-35. [PMID: 26019752 PMCID: PMC4445707 DOI: 10.15283/ijsc.2015.8.1.24] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 04/14/2015] [Indexed: 02/06/2023] Open
Abstract
Stem cells have attracted much attention due to their distinct features that support infinite self-renewal and differentiation into the cellular derivatives of three lineages. Recent studies have suggested that many stem cells both embryonic and adult stem cells reside in a specialized niche defined by hypoxic condition. In this respect, distinguishing functional differences arising from the oxygen concentration is important in understanding the nature of stem cells and in controlling stem cell fate for therapeutic purposes. ROS act as cellular signaling molecules involved in the propagation of signaling and the translation of environmental cues into cellular responses to maintain cellular homeostasis, which is mediated by the coordination of various cellular processes, and to adapt cellular activity to available bioenergetic sources. Thus, in this review, we describe the physiological role of ROS in stem cell fate and its effect on the metabolic regulation of stem cells.
Collapse
Affiliation(s)
- Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ki Hoon Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Dah Ihm Kim
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Jeong Yeon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - So Hee Ko
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ing Ing Chai
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Eun Ju Song
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ji Young Oh
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
59
|
Li N, An L, Hang H. Increased sensitivity of DNA damage response-deficient cells to stimulated microgravity-induced DNA lesions. PLoS One 2015; 10:e0125236. [PMID: 25915950 PMCID: PMC4411073 DOI: 10.1371/journal.pone.0125236] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/22/2015] [Indexed: 12/02/2022] Open
Abstract
Microgravity is a major stress factor that astronauts have to face in space. In the past, the effects of microgravity on genomic DNA damage were studied, and it seems that the effect on genomic DNA depends on cell types and the length of exposure time to microgravity or simulated microgravity (SMG). In this study we used mouse embryonic stem (MES) and mouse embryonic fibroblast (MEF) cells to assess the effects of SMG on DNA lesions. To acquire the insight into potential mechanisms by which cells resist and/or adapt to SMG, we also included Rad9-deleted MES and Mdc1-deleted MEF cells in addition to wild type cells in this study. We observed significant SMG-induced DNA double strand breaks (DSBs) in Rad9-/- MES and Mdc1-/- MEF cells but not in their corresponding wild type cells. A similar pattern of DNA single strand break or modifications was also observed in Rad9-/- MES. As the exposure to SMG was prolonged, Rad9-/- MES cells adapted to the SMG disturbance by reducing the induced DNA lesions. The induced DNA lesions in Rad9-/- MES were due to SMG-induced reactive oxygen species (ROS). Interestingly, Mdc1-/- MEF cells were only partially adapted to the SMG disturbance. That is, the induced DNA lesions were reduced over time, but did not return to the control level while ROS returned to a control level. In addition, ROS was only partially responsible for the induced DNA lesions in Mdc1-/- MEF cells. Taken together, these data suggest that SMG is a weak genomic DNA stress and can aggravate genomic instability in cells with DNA damage response (DDR) defects.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory for Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lili An
- Key Laboratory for Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- * E-mail: (HYH); (LLA)
| | - Haiying Hang
- Key Laboratory for Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- * E-mail: (HYH); (LLA)
| |
Collapse
|
60
|
Sun X, Pang L, Shi M, Huang J, Wang Y. HIF2α induces cardiomyogenesis via Wnt/β-catenin signaling in mouse embryonic stem cells. J Transl Med 2015; 13:88. [PMID: 25889500 PMCID: PMC4399227 DOI: 10.1186/s12967-015-0447-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/27/2015] [Indexed: 11/10/2022] Open
Abstract
Background Embryonic stem cells (ESCs) are pluripotent stem cells and can differentiate into cardiomyocytes when cultured in appropriate conditions. The function of hypoxia-inducible factors (HIFs) has been identified in directing the formation of cardiac lineages. The purpose of this study was to investigate the ability of HIF2α to induce differentiation of ESCs into cardiomyocytes and to explore the potential underlying molecular mechanisms. Methods Cardiac differentiation from mouse ESCs was analyzed using the “hanging drop” method, and success was determined by assaying the numbers of beating embryoid bodies and the expression level of cardiac markers. The expression of HIF2α was then manipulated during cardiac differentiation with piggyBac transposon and the lentivirus system. The underlying mechanism was finally examined via administering selective inhibitors of the Wnt/β-catenin signaling pathway. Results Overexpressing HIF2α can significantly drive mouse ESCs to form cardiomyocytes. Contrarily, knockdown of HIF2α inhibits the emergence of cardiac cells. In addition, the cardiomyogenesis-promoting effect of HIF2α occurred by increasing the protein level of β-catenin, an effector that contributes to cardiac differentiation at an early stage of ESC differentiation. Conclusion HIF2α has a cardiomyogenesis-promoting effect in ESCs via enhancing the activation of the Wnt/β-catenin signaling pathway. Our results may be beneficial for generating and applying cardiomyocytes from ESCs safely and effectively in the future. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0447-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaotian Sun
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Liewen Pang
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Meng Shi
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Jiechun Huang
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| | - Yiqing Wang
- Department of Cardiothoracic surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Road, Shanghai, 200040, PR, China.
| |
Collapse
|
61
|
Lee JE, Cho KE, Lee KE, Kim J, Bae YS. Nox4-mediated cell signaling regulates differentiation and survival of neural crest stem cells. Mol Cells 2014; 37:907-11. [PMID: 25410908 PMCID: PMC4275708 DOI: 10.14348/molcells.2014.0244] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/23/2014] [Indexed: 11/27/2022] Open
Abstract
The function of reactive oxygen species (ROS) as second messengers in cell differentiation has been demonstrated only for a limited number of cell types. Here, we used a well-established protocol for BMP2-induced neuronal differentiation of neural crest stem cells (NCSCs) to examine the function of BMP2-induced ROS during the process. We first show that BMP2 indeed induces ROS generation in NCSCs and that blocking ROS generation by pretreatment of cells with diphenyleneiodonium (DPI) as NADPH oxidase (Nox) inhibitor inhibits neuronal differentiation. Among the ROS-generating Nox isozymes, only Nox4 was expressed at a detectable level in NCSCs. Nox4 appears to be critical for survival of NCSCs at least in vitro as down-regulation by RNA interference led to apoptotic response from NCSCs. Interestingly, development of neural crest-derived peripheral neural structures in Nox4-/- mouse appears to be grossly normal, although Nox4-/- embryos were born at a sub-Mendelian ratio and showed delayed over-all development. Specifically, cranial and dorsal root ganglia, derived from NCSCs, were clearly present in Nox4-/- embryo at embryonic days (E) 9.5 and 10.5. These results suggest that Nox4-mediated ROS generation likely plays important role in fate determination and differentiation of NCSCs, but other Nox isozymes play redundant function during embryogenesis.
Collapse
Affiliation(s)
- Ji-Eun Lee
- Department of Life Science and GT5 Program, Ewha Womans University, Seoul 120-750,
Korea
| | - Kyu Eun Cho
- Department of Life Science and GT5 Program, Ewha Womans University, Seoul 120-750,
Korea
| | - Kyung Eun Lee
- Department of Life Science and GT5 Program, Ewha Womans University, Seoul 120-750,
Korea
| | - Jaesang Kim
- Department of Life Science and GT5 Program, Ewha Womans University, Seoul 120-750,
Korea
| | - Yun Soo Bae
- Department of Life Science and GT5 Program, Ewha Womans University, Seoul 120-750,
Korea
| |
Collapse
|
62
|
Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr Rev 2014; 35:851-905. [PMID: 25141152 DOI: 10.1210/er.2014-1045] [Citation(s) in RCA: 707] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Decidualization denotes the transformation of endometrial stromal fibroblasts into specialized secretory decidual cells that provide a nutritive and immunoprivileged matrix essential for embryo implantation and placental development. In contrast to most mammals, decidualization of the human endometrium does not require embryo implantation. Instead, this process is driven by the postovulatory rise in progesterone levels and increasing local cAMP production. In response to falling progesterone levels, spontaneous decidualization causes menstrual shedding and cyclic regeneration of the endometrium. A growing body of evidence indicates that the shift from embryonic to maternal control of the decidual process represents a pivotal evolutionary adaptation to the challenge posed by invasive and chromosomally diverse human embryos. This concept is predicated on the ability of decidualizing stromal cells to respond to individual embryos in a manner that either promotes implantation and further development or facilitates early rejection. Furthermore, menstruation and cyclic regeneration involves stem cell recruitment and renders the endometrium intrinsically capable of adapting its decidual response to maximize reproductive success. Here we review the endocrine, paracrine, and autocrine cues that tightly govern this differentiation process. In response to activation of various signaling pathways and genome-wide chromatin remodeling, evolutionarily conserved transcriptional factors gain access to the decidua-specific regulatory circuitry. Once initiated, the decidual process is poised to transit through distinct phenotypic phases that underpin endometrial receptivity, embryo selection, and, ultimately, resolution of pregnancy. We discuss how disorders that subvert the programming, initiation, or progression of decidualization compromise reproductive health and predispose for pregnancy failure.
Collapse
Affiliation(s)
- Birgit Gellersen
- Endokrinologikum Hamburg (B.G.), 20251 Hamburg, Germany; and Division of Reproductive Health (J.J.B.), Warwick Medical School, University of Warwick, Coventry CV4 7AL, United Kingdom
| | | |
Collapse
|
63
|
Redox regulation of cardiomyocyte cell cycling via an ERK1/2 and c-Myc-dependent activation of cyclin D2 transcription. J Mol Cell Cardiol 2014; 79:54-68. [PMID: 25450615 PMCID: PMC4312357 DOI: 10.1016/j.yjmcc.2014.10.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 01/09/2023]
Abstract
Adult mammalian cardiomyocytes have a very limited capacity to proliferate, and consequently the loss of cells after cardiac stress promotes heart failure. Recent evidence suggests that administration of hydrogen peroxide (H2O2), can regulate redox-dependent signalling pathway(s) to promote cardiomyocyte proliferation in vitro, but the potential relevance of such a pathway in vivo has not been tested. We have generated a transgenic (Tg) mouse model in which the H2O2-generating enzyme, NADPH oxidase 4 (Nox4), is overexpressed within the postnatal cardiomyocytes, and observed that the hearts of 1-3week old Tg mice pups are larger in comparison to wild type (Wt) littermate controls. We demonstrate that the cardiomyocytes of Tg mouse pups have increased cell cycling capacity in vivo as determined by incorporation of 5-bromo-2'-deoxyuridine. Further, microarray analyses of the transcriptome of these Tg mouse hearts suggested that the expression of cyclin D2 is significantly increased. We investigated the molecular mechanisms which underlie this more proliferative phenotype in isolated neonatal rat cardiomyocytes (NRCs) in vitro, and demonstrate that Nox4 overexpression mediates an H2O2-dependent activation of the ERK1/2 signalling pathway, which in turn phosphorylates and activates the transcription factor c-myc. This results in a significant increase in cyclin D2 expression, which we show to be mediated, at least in part, by cis-acting c-myc binding sites within the proximal cyclin D2 promoter. Overexpression of Nox4 in NRCs results in an increase in their proliferative capacity that is ablated by the silencing of cyclin D2. We further demonstrate activation of the ERK1/2 signalling pathway, increased phosphorylation of c-myc and significantly increased expression of cyclin D2 protein in the Nox4 Tg hearts. We suggest that this pathway acts to maintain the proliferative capacity of cardiomyocytes in Nox4 Tg pups in vivo and so delays their exit from the cell cycle after birth.
Collapse
|
64
|
Perales-Clemente E, Folmes CDL, Terzic A. Metabolic regulation of redox status in stem cells. Antioxid Redox Signal 2014; 21:1648-59. [PMID: 24949895 PMCID: PMC4174422 DOI: 10.1089/ars.2014.6000] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Metabolism-dependent generation of reactive oxygen species (ROS) and associated oxidative damage have been traditionally linked to impaired homeostasis and cellular death. Beyond the adverse effects of ROS accumulation, increasing evidence implicates redox status as a regulator of vital cellular processes. RECENT ADVANCES Emerging studies on the molecular mechanisms guiding stem cell fate decisions indicate a role for energy metabolism in regulating the fundamental ability of maintaining stemness versus undergoing lineage-specific differentiation. Stem cells have evolved protective metabolic phenotypes to minimize reactive oxygen generation through oxidative metabolism and support antioxidant scavenging through glycolysis and the pentose phosphate pathway. CRITICAL ISSUES While the dynamics in ROS generation has been correlated with stem cell function, the intimate mechanisms by which energy metabolism regulates ROS to impact cellular fate remain to be deciphered. FUTURE DIRECTIONS Decoding the linkage between nutrient sensing, energy metabolism, and ROS in regulating cell fate decisions would offer a redox-dependent strategy to regulate stemness and lineage specification.
Collapse
|
65
|
Kimura W, Muralidhar S, Canseco DC, Puente B, Zhang CC, Xiao F, Abderrahman YH, Sadek HA. Redox signaling in cardiac renewal. Antioxid Redox Signal 2014; 21:1660-73. [PMID: 25000143 PMCID: PMC4175032 DOI: 10.1089/ars.2014.6029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Utilizing oxygen (O2) through mitochondrial oxidative phosphorylation enables organisms to generate adenosine triphosphate (ATP) with a higher efficiency than glycolysis, but it results in increased reactive oxygen species production from mitochondria, which can result in stem cell dysfunction and senescence. RECENT ADVANCES In the postnatal organism, the hematopoietic system represents a classic example of the role of stem cells in cellular turnover and regeneration. However, in other organs such as the heart, both the degree and source of cellular turnover have been heavily contested. CRITICAL ISSUES Although recent evidence suggests that the major source of the limited cardiomyocyte turnover in the adult heart is cardiomyocyte proliferation, the identity and potential role of undifferentiated cardiac progenitor cells remain controversial. Several types of cardiac progenitor cells have been identified, and several studies have identified an important role of redox and metabolic regulation in survival and differentiation of cardiac progenitor cells. Perhaps a simple way to approach these controversies is to focus on the multipotentiality characteristics of a certain progenitor population, and not necessarily its ability to give rise to all cell types within the heart. In addition, it is important to note that cycling cells in the heart may express markers of differentiation or may be truly undifferentiated, and for the purpose of this review, we will refer to these cycling cells as progenitors. FUTURE DIRECTIONS We propose that hypoxia, redox signaling, and metabolic phenotypes are major regulators of cardiac renewal, and may prove to be important therapeutic targets for heart regeneration.
Collapse
Affiliation(s)
- Wataru Kimura
- 1 Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center , Dallas, Texas
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Fosen KM, Thom SR. Hyperbaric oxygen, vasculogenic stem cells, and wound healing. Antioxid Redox Signal 2014; 21:1634-47. [PMID: 24730726 PMCID: PMC4175035 DOI: 10.1089/ars.2014.5940] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/14/2014] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Oxidative stress is recognized as playing a role in stem cell mobilization from peripheral sites and also cell function. RECENT ADVANCES This review focuses on the impact of hyperoxia on vasculogenic stem cells and elements of wound healing. CRITICAL ISSUES Components of the wound-healing process in which oxidative stress has a positive impact on the various cells involved in wound healing are highlighted. A slightly different view of wound-healing physiology is adopted by departing from the often used notion of sequential stages: hemostatic, inflammatory, proliferative, and remodeling and instead organizes the cascade of wound healing as overlapping events or waves pertaining to reactive oxygen species, lactate, and nitric oxide. This was done because hyperoxia has effects of a number of cell signaling events that converge to influence cell recruitment/chemotaxis and gene regulation/protein synthesis responses which mediate wound healing. FUTURE DIRECTIONS Our alternative perspective of the stages of wound healing eases recognition of the multiple sites where oxidative stress has an impact on wound healing. This aids the focus on mechanistic events and the interplay among various cell types and biochemical processes. It also highlights the areas where additional research is needed.
Collapse
Affiliation(s)
- Katina M. Fosen
- Department of Emergency Medicine, Institute for Environmental Medicine, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Stephen R. Thom
- Department of Emergency Medicine, University of Maryland, Baltimore, Maryland
| |
Collapse
|
67
|
Puente BN, Kimura W, Muralidhar SA, Moon J, Amatruda JF, Phelps KL, Grinsfelder D, Rothermel BA, Chen R, Garcia JA, Santos CX, Thet S, Mori E, Kinter MT, Rindler PM, Zacchigna S, Mukherjee S, Chen DJ, Mahmoud AI, Giacca M, Rabinovitch PS, Aroumougame A, Shah AM, Szweda LI, Sadek HA. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell 2014; 157:565-79. [PMID: 24766806 DOI: 10.1016/j.cell.2014.03.032] [Citation(s) in RCA: 686] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/17/2014] [Accepted: 03/21/2014] [Indexed: 12/15/2022]
Abstract
The mammalian heart has a remarkable regenerative capacity for a short period of time after birth, after which the majority of cardiomyocytes permanently exit cell cycle. We sought to determine the primary postnatal event that results in cardiomyocyte cell-cycle arrest. We hypothesized that transition to the oxygen-rich postnatal environment is the upstream signal that results in cell-cycle arrest of cardiomyocytes. Here, we show that reactive oxygen species (ROS), oxidative DNA damage, and DNA damage response (DDR) markers significantly increase in the heart during the first postnatal week. Intriguingly, postnatal hypoxemia, ROS scavenging, or inhibition of DDR all prolong the postnatal proliferative window of cardiomyocytes, whereas hyperoxemia and ROS generators shorten it. These findings uncover a protective mechanism that mediates cardiomyocyte cell-cycle arrest in exchange for utilization of oxygen-dependent aerobic metabolism. Reduction of mitochondrial-dependent oxidative stress should be an important component of cardiomyocyte proliferation-based therapeutic approaches.
Collapse
Affiliation(s)
- Bao N Puente
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wataru Kimura
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shalini A Muralidhar
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jesung Moon
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James F Amatruda
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kate L Phelps
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David Grinsfelder
- Department of Clinical Science, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Beverly A Rothermel
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rui Chen
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph A Garcia
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Celio X Santos
- Cardiovascular Division, King's College London BHF Centre of Research Excellence, School of Medicine, James Black Centre, London SE5 9NU, UK
| | - SuWannee Thet
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eiichiro Mori
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael T Kinter
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Paul M Rindler
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Serena Zacchigna
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Shibani Mukherjee
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David J Chen
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ahmed I Mahmoud
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, MA 02115, USA
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | | | - Asaithamby Aroumougame
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ajay M Shah
- Department of Clinical Science, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Luke I Szweda
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hesham A Sadek
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
68
|
Jin J, Xuan QK, Zhou LJ, Shi CM, Song GX, Sheng YH, Qian LM. Dynamic mitochondrial changes during differentiation of P19 embryonic carcinoma cells into cardiomyocytes. Mol Med Rep 2014; 10:761-6. [PMID: 24920049 DOI: 10.3892/mmr.2014.2315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 05/02/2014] [Indexed: 11/06/2022] Open
Abstract
Murine P19 embryonal carcinoma cells are multipotent cells that can differentiate into cardiomyocytes when treated with dimethyl sulfoxide. This experimental model provides an invaluable tool to study different aspects of cardiac differentiation, such as the function of cardiac‑specific transcription factors and signaling pathways, and the regulation of contractile protein expression. The role of mitochondria during cardiac differentiation is unclear. In this context, we have examined the mitochondrial-related changes in undifferentiated and differentiated P19 cells. We observed that mitochondrial DNA content sharply decreased in P19 cell aggregates compared to undifferentiated cells, accompanied by decreased levels of adenosine triphosphate (ATP) and reactive oxygen species (ROS). Following the aggregation stage, the mitochondrial DNA content reached its highest level on day 7 of the differentiation process, with the intracellular ROS level showing a trend to increase, similar to cellular ATP production. In conclusion, our study on differentiating P19 embryonal carcinoma cells provides new insights into the role of mitochondria in the differentiation of P19 stem cells into beating cardiomyocytes.
Collapse
Affiliation(s)
- Jin Jin
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Qin-Kao Xuan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Li-Juan Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Chun-Mei Shi
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P.R. China
| | - Gui-Xian Song
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Yan-Hui Sheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Ling-Mei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China
| |
Collapse
|
69
|
Abstract
SIGNIFICANCE Stem cells are characterized by the properties of self-renewal and the ability to differentiate into multiple cell types, and thus maintain tissue homeostasis. Reactive oxygen species (ROS) are a natural byproduct of aerobic metabolism and have roles in cell signaling. Regulation of ROS has a vital role in maintaining "stemness" and differentiation of the stem cells, as well as in progression of stem-cell-associated diseases. RECENT ADVANCES As of late, much research has been done on the adverse effects of ROS in stem cells. However, recently it has become apparent that in some cases redox status of the stem cell does have a role in maintaining its identity as such. Both pluripotent and multipotent stem cell types have been reported to possess enzymatic and nonenzymatic mechanisms for detoxification of ROS and to correct oxidative damage to the genome as well as the proteome. CRITICAL ISSUES Although context dependent and somewhat varied among different stem cell types, the correlation seems to exist between antioxidant defense level and stem cell fate change (i.e., proliferation, differentiation, and death). Changes in stem cell redox regulation may affect the pathogenesis of various human diseases. FUTURE DIRECTIONS Dissecting the defined roles of ROS in distinct stem cell types will greatly enhance their basic and translational applications. Here, we discuss the various roles of ROS in adult, embryonic, and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Pooja Chaudhari
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | |
Collapse
|
70
|
Murray TV, Ahmad A, Brewer AC. Reactive oxygen at the heart of metabolism. Trends Cardiovasc Med 2014; 24:113-20. [DOI: 10.1016/j.tcm.2013.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 02/04/2023]
|
71
|
Kim SU, Park YH, Kim JM, Sun HN, Song IS, Huang SM, Lee SH, Chae JI, Hong S, Sik Choi S, Choi SC, Lee TH, Kang SW, Rhee SG, Chang KT, Lee SH, Yu DY, Lee DS. Dominant Role of Peroxiredoxin/JNK Axis in Stemness Regulation During Neurogenesis from Embryonic Stem Cells. Stem Cells 2014; 32:998-1011. [DOI: 10.1002/stem.1593] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Redox balance has been suggested as an important determinant of “stemness” in embryonic stem cells (ESCs). In this study, we demonstrate that peroxiredoxin (Prx) plays a pivotal role in maintenance of ESC stemness during neurogenesis through suppression of reactive oxygen species (ROS)-sensitive signaling. During neurogenesis, Prx I and Oct4 are expressed in a mutually dependent manner and their expression is abruptly downregulated by an excess of ROS. Thus, in Prx I−/− or Prx II−/− ESCs, rapid loss of stemness can occur due to spontaneous ROS overload, leading to their active commitment into neurons; however, stemness is restored by the addition of an antioxidant or an inhibitor of c-Jun N-terminal kinase (JNK). In addition, Prx I and Prx II appear to have a tight association with the mechanism underlying the protection of ESC stemness in developing teratomas. These results suggest that Prx functions as a protector of ESC stemness by opposing ROS/JNK cascades during neurogenesis. Therefore, our findings have important implications for understanding of maintenance of ESC stemness through involvement of antioxidant enzymes and may lead to development of an alternative stem cell-based therapeutic strategy for production of high-quality neurons in large quantity. Stem Cells 2014;32:998–1011
Collapse
Affiliation(s)
- Sun-Uk Kim
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics University of Science and Technology (UST), Daejeon, Republic of Korea
- National Primate Research Center, KRIBB, Chungcheongbuk-do, Republic of Korea
- Division of Life Sciences and Biotechnology Korea University, Seoul, Republic of Korea
| | - Young-Ho Park
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics University of Science and Technology (UST), Daejeon, Republic of Korea
- National Primate Research Center, KRIBB, Chungcheongbuk-do, Republic of Korea
| | - Jin-Man Kim
- Department of Pathology, College of Medicine Chungnam National University, Daejeon, Republic of Korea
| | - Hu-Nan Sun
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - In-Sung Song
- Cardiovascular and Metabolic Disease Center Inje University, Busan, Republic of Korea
| | - Song Mei Huang
- Department of Pathology, College of Medicine Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Hee Lee
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jung-Il Chae
- Department of Oral Pharmacology School of Dentistry and Institute of Dental Bioscience BK21 plus project, Chonbuk National University, Jeonju, Republic of Korea
| | - Su Hong
- Division of Life Sciences and Biotechnology Korea University, Seoul, Republic of Korea
| | - Sung Sik Choi
- Division of Life Sciences and Biotechnology Korea University, Seoul, Republic of Korea
| | - Seung-Cheol Choi
- Division of Life Sciences and Biotechnology Korea University, Seoul, Republic of Korea
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, College of Dentistry Chonnam National University, Gwangju, Republic of Korea
| | - Sang Won Kang
- Division of Life and Pharmaceutical Sciences Ewha Womans University, Seoul, Republic of Korea
| | - Sue Goo Rhee
- Division of Life and Pharmaceutical Sciences Ewha Womans University, Seoul, Republic of Korea
| | - Kyu-Tae Chang
- National Primate Research Center, KRIBB, Chungcheongbuk-do, Republic of Korea
| | - Sang Ho Lee
- Division of Life Sciences and Biotechnology Korea University, Seoul, Republic of Korea
| | - Dae-Yeul Yu
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Dong-Seok Lee
- College of Natural Sciences BK21 plus project, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
72
|
Cho SW, Park JS, Heo HJ, Park SW, Song S, Kim I, Han YM, Yamashita JK, Youm JB, Han J, Koh GY. Dual modulation of the mitochondrial permeability transition pore and redox signaling synergistically promotes cardiomyocyte differentiation from pluripotent stem cells. J Am Heart Assoc 2014; 3:e000693. [PMID: 24627421 PMCID: PMC4187507 DOI: 10.1161/jaha.113.000693] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Cardiomyocytes that differentiate from pluripotent stem cells (PSCs) provide a crucial cellular resource for cardiac regeneration. The mechanisms of mitochondrial metabolic and redox regulation for efficient cardiomyocyte differentiation are, however, still poorly understood. Here, we show that inhibition of the mitochondrial permeability transition pore (mPTP) by Cyclosporin A (CsA) promotes cardiomyocyte differentiation from PSCs. Methods and Results We induced cardiomyocyte differentiation from mouse and human PSCs and examined the effect of CsA on the differentiation process. The cardiomyogenic effect of CsA mainly resulted from mPTP inhibition rather than from calcineurin inhibition. The mPTP inhibitor NIM811, which does not have an inhibitory effect on calcineurin, promoted cardiomyocyte differentiation as much as CsA did, but calcineurin inhibitor FK506 only slightly increased cardiomyocyte differentiation. CsA‐treated cells showed an increase in mitochondrial calcium, mitochondrial membrane potential, oxygen consumption rate, ATP level, and expression of genes related to mitochondrial function. Furthermore, inhibition of mitochondrial oxidative metabolism reduced the cardiomyogenic effect of CsA while antioxidant treatment augmented the cardiomyogenic effect of CsA. Conclusions Our data show that mPTP inhibition by CsA alters mitochondrial oxidative metabolism and redox signaling, which leads to differentiation of functional cardiomyocytes from PSCs.
Collapse
Affiliation(s)
- Sung Woo Cho
- Laboratory of Vascular Biology and Stem Cell, Korea Advanced Institute of Science and Technology
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Lobanok ES, Kvacheva ZB, Pinchuk SV, Volk MV, Mezhevikina LM, Fesenko EE, Volotovski ID. The influence of fibroblast growth factor (FGF2) on cardiomyocytes differentiation of mesenchymal stem cells of bone marrow ex vivo. Biophysics (Nagoya-shi) 2014. [DOI: 10.1134/s0006350914020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
74
|
Piccoli C, Agriesti F, Scrima R, Falzetti F, Di Ianni M, Capitanio N. To breathe or not to breathe: the haematopoietic stem/progenitor cells dilemma. Br J Pharmacol 2014; 169:1652-71. [PMID: 23714011 DOI: 10.1111/bph.12253] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/11/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Adult haematopoietic stem/progenitor cells (HSPCs) constitute the lifespan reserve for the generation of all the cellular lineages in the blood. Although massive progress in identifying the cluster of master genes controlling self-renewal and multipotency has been achieved in the past decade, some aspects of the physiology of HSPCs still need to be clarified. In particular, there is growing interest in the metabolic profile of HSPCs in view of their emerging role as determinants of cell fate. Indeed, stem cells and progenitors have distinct metabolic profiles, and the transition from stem to progenitor cell corresponds to a critical metabolic change, from glycolysis to oxidative phosphorylation. In this review, we summarize evidence, reported in the literature and provided by our group, highlighting the peculiar ability of HSPCs to adapt their mitochondrial oxidative/bioenergetic metabolism to survive in the hypoxic microenvironment of the endoblastic niche and to exploit redox signalling in controlling the balance between quiescence versus active cycling and differentiation. Especial prominence is given to the interplay between hypoxia inducible factor-1, globins and NADPH oxidases in managing the mitochondrial dioxygen-related metabolism and biogenesis in HSPCs under different ambient conditions. A mechanistic model is proposed whereby 'mitochondrial differentiation' is a prerequisite in uncommitted stem cells, paving the way for growth/differentiation factor-dependent processes. Advancing the understanding of stem cell metabolism will, hopefully, help to (i) improve efforts to maintain, expand and manipulate HSPCs ex vivo and realize their potential therapeutic benefits in regenerative medicine; (ii) reprogramme somatic cells to generate stem cells; and (iii) eliminate, selectively, malignant stem cells. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- C Piccoli
- Department of Medical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | | | | | | | | | | |
Collapse
|
75
|
Sandiford SDE, Kennedy KAM, Xie X, Pickering JG, Li SSC. Dual oxidase maturation factor 1 (DUOXA1) overexpression increases reactive oxygen species production and inhibits murine muscle satellite cell differentiation. Cell Commun Signal 2014; 12:5. [PMID: 24410844 PMCID: PMC3895674 DOI: 10.1186/1478-811x-12-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 12/23/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dual oxidase maturation factor 1 (DUOXA1) has been associated with the maturation of the reactive oxygen species (ROS) producing enzyme, dual oxidase 1 (DUOX1) in the adult thyroid. However, ROS have also been implicated in the development of several tissues. We found that activated muscle satellite cells and primary myoblasts isolated from mice express robust levels of DUOXA1 and that its levels are altered as cells differentiate. RESULTS To determine whether DUOXA1 levels affect muscle differentiation, we used an adenoviral construct (pCMV5-DUOXA1-GFP) to drive constitutive overexpression of this protein in primary myoblasts. High levels of DUOXA1 throughout myogenesis resulted in enhanced H2O2 production, fusion defects, reduced expression of early (myogenin) and late (myosin heavy chain) markers of differentiation, and elevated levels of apoptosis compared to control cells infected with an empty adenoviral vector (pCMV5-GFP). DUOXA1 knockdown (using a DUOXA1 shRNA construct) resulted in enhanced differentiation compared to cells subjected to a control shRNA, and subjecting DUOXA1 overexpressing cells to siRNAs targeting DUOX1 or apoptosis signal-regulating kinase 1 (ASK1) rescued the phenotype. CONCLUSIONS This study represents the first to demonstrate the importance of DUOXA1 in skeletal muscle myoblasts and that DUOXA1 overexpression in muscle stem cells induces apoptosis and inhibits differentiation through DUOX1 and ASK1.
Collapse
Affiliation(s)
- Shelley DE Sandiford
- Siebens-Drake Research Institute, 1400 Western Road, London, Ontario N6G 2 V4, Canada
| | - Karen AM Kennedy
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Xiaojun Xie
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - J Geoffrey Pickering
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Shawn SC Li
- Siebens-Drake Research Institute, 1400 Western Road, London, Ontario N6G 2 V4, Canada
| |
Collapse
|
76
|
Yang C, Madonna R, Li Y, Zhang Q, Shen WF, McNamara K, Yang YJ, Geng YJ. Simvastatin-enhanced expression of promyogenic nuclear factors and cardiomyogenesis of murine embryonic stem cells. Vascul Pharmacol 2013; 60:8-16. [PMID: 24200505 DOI: 10.1016/j.vph.2013.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/30/2013] [Accepted: 10/28/2013] [Indexed: 12/24/2022]
Abstract
A combination of statin and stem cell therapies has been shown to benefit in experimental models of myocardial infarction. This study tests whether treatment with simvastatin has a direct impact on the cardiomyogenic development of murine embryonic stem cells (ESCs) in embryoid bodies. In a concentration-dependent manner, simvastatin treatment enhanced expression of several promyogenic nuclear transcription factors, including GATA4, Nkx2.5, DTEF-1 and myocardin A. The statin-treated cells also displayed higher levels of cardiac proteins, including myosin, α-actinin, Ryanodine receptor-2, and atrial natriuretic peptide, and they developed synchronized contraction. The statin's promyogenic effect was partially diminished by the addition of the two isoprenoids FPP and GGPP, which are intermediates of cholesterol synthesis. Thus, simvastatin treatment enhances ESC myogenesis during early development perhaps via a mechanism inhibiting the mevalonate-FPP/GGPP pathway.
Collapse
Affiliation(s)
- ChenMin Yang
- The Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Medical School at Houston, Houston, TX USA; The Department of Obstetrics and Gynecology, Ruijin Hospital, Jiao-Tong University Medical School, Shanghai, China; Texas Heart Institute, Houston, TX, USA
| | - Rosalinda Madonna
- The Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Medical School at Houston, Houston, TX USA; Texas Heart Institute, Houston, TX, USA
| | - Yangxin Li
- The Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Medical School at Houston, Houston, TX USA; Texas Heart Institute, Houston, TX, USA
| | - Qi Zhang
- The Department of Cardiovascular Medicine, Ruijin Hospital, Jiao-Tong University Medical School, Shanghai, China
| | - Wei-Feng Shen
- The Department of Cardiovascular Medicine, Ruijin Hospital, Jiao-Tong University Medical School, Shanghai, China
| | - Katharine McNamara
- The Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Medical School at Houston, Houston, TX USA; Texas Heart Institute, Houston, TX, USA
| | - Yue-Jin Yang
- FuWai Cardiovascular Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong-Jian Geng
- The Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Medical School at Houston, Houston, TX USA; Texas Heart Institute, Houston, TX, USA.
| |
Collapse
|
77
|
Geuss LR, Suggs LJ. Making cardiomyocytes: How mechanical stimulation can influence differentiation of pluripotent stem cells. Biotechnol Prog 2013; 29:1089-96. [DOI: 10.1002/btpr.1794] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Laura R. Geuss
- Dept. of Biomedical Engineering; The University of Texas at Austin; Austin TX 78712
| | - Laura J. Suggs
- Dept. of Biomedical Engineering; The University of Texas at Austin; Austin TX 78712
| |
Collapse
|
78
|
Parameswaran S, Kumar S, Verma RS, Sharma RK. Cardiomyocyte culture - an update on the in vitro cardiovascular model and future challenges. Can J Physiol Pharmacol 2013; 91:985-98. [PMID: 24289068 DOI: 10.1139/cjpp-2013-0161] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The success of any work with isolated cardiomyocytes depends on the reproducibility of cell isolation, because the cells do not divide. To date, there is no suitable in vitro model to study human adult cardiac cell biology. Although embryonic stem cells and induced pluripotent stem cells are able to differentiate into cardiomyocytes in vitro, the efficiency of this process is low. Isolation and expansion of human cardiomyocyte progenitor cells from cardiac surgical waste or, alternatively, from fetal heart tissue is another option. However, to overcome various issues related to human tissue usage, especially ethical concerns, researchers use large- and small-animal models to study cardiac pathophysiology. A simple model to study the changes at the cellular level is cultures of cardiomyocytes. Although primary murine cardiomyocyte cultures have their own advantages and drawbacks, alternative strategies have been developed in the last two decades to minimise animal usage and interspecies differences. This review discusses the use of freshly isolated murine cardiomyocytes and cardiomyocyte alternatives for use in cardiac disease models and other related studies.
Collapse
Affiliation(s)
- Sreejit Parameswaran
- a Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | | | | | | |
Collapse
|
79
|
Wu Y, Zhang X, Kang X, Li N, Wang R, Hu T, Xiang M, Wang X, Yuan W, Chen A, Meng D, Chen S. Oxidative stress inhibits adhesion and transendothelial migration, and induces apoptosis and senescence of induced pluripotent stem cells. Clin Exp Pharmacol Physiol 2013; 40:626-34. [DOI: 10.1111/1440-1681.12141] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/30/2013] [Accepted: 06/07/2013] [Indexed: 12/25/2022]
Affiliation(s)
- Yi Wu
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
- Department of Physiology; Ningxia Medical College; Yinchuan Ningxia China
| | - Xueqing Zhang
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Xueling Kang
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Ning Li
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Rong Wang
- Department of Physiology; Ningxia Medical College; Yinchuan Ningxia China
| | - Tiantian Hu
- Department of Physiology; Ningxia Medical College; Yinchuan Ningxia China
| | - Meng Xiang
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Wenjun Yuan
- Department of Physiology; Ningxia Medical College; Yinchuan Ningxia China
| | - Alex Chen
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Dan Meng
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| | - Sifeng Chen
- Department of Physiology and Pathophysiology; Fudan University Shanghai Medical College; Shanghai China
| |
Collapse
|
80
|
Bekhite MM, Figulla HR, Sauer H, Wartenberg M. Static magnetic fields increase cardiomyocyte differentiation of Flk-1+ cells derived from mouse embryonic stem cells via Ca2+ influx and ROS production. Int J Cardiol 2013; 167:798-808. [DOI: 10.1016/j.ijcard.2012.02.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/16/2012] [Accepted: 02/26/2012] [Indexed: 11/16/2022]
|
81
|
Cheng W, Yu Z, Feng L, Wang Y. Perfluorooctane sulfonate (PFOS) induced embryotoxicity and disruption of cardiogenesis. Toxicol In Vitro 2013; 27:1503-12. [DOI: 10.1016/j.tiv.2013.03.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 03/04/2013] [Accepted: 03/25/2013] [Indexed: 10/27/2022]
|
82
|
Affiliation(s)
- Xuejun Yuan
- From the Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- From the Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
83
|
Nadworny AS, Guruju MR, Poor D, Doran RM, Sharma RV, Kotlikoff MI, Davisson RL. Nox2 and Nox4 influence neonatal c-kit(+) cardiac precursor cell status and differentiation. Am J Physiol Heart Circ Physiol 2013; 305:H829-42. [PMID: 23832701 DOI: 10.1152/ajpheart.00761.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Redox status has emerged as critical in modulating stemness and lineage commitment in several precursor cell types. However, a role for redox genes, specifically NADPH oxidases (Nox), in cardiac precursor cells (CPCs) has not been established. We tested whether CPCs marked by type III receptor tyrosine kinase c-kit (c-kit(+)) exhibit a unique NADPH oxidase signature that confers precursor status and whether alterations in this profile are functionally linked to changes in lineage specification. Dihydroethidium (DHE) microfluorography indicated reduced basal reactive oxygen species (ROS) formation within early postnatal c-kit(+) CPCs. Real-time quantitative PCR revealed downregulation of ROS generator Nox2 and its subunit p67(phox) in c-kit(+) CPCs under basal conditions but upregulation of Nox2 and Nox4 over the course of differentiation. Adenoviral silencing of Nox2 and Nox4 increased expression of CPC markers c-kit and Flk-1 and blunted smooth and cardiac muscle differentiation, respectively, while overexpression of Nox2 and Nox4 significantly reduced c-kit expression. These changes were accompanied by altered expression of transcription factors regulating cardiac lineage commitment, Gata6 and Gata4, and cytokine transforming growth factor (TGF)-β1. Similar to other precursor cell types, RT(2)Profiler PCR Arrays revealed that c-kit(+) CPCs also exhibit enhanced antioxidant capacity at the mRNA level. In conclusion, we report that c-kit(+) CPCs demonstrate reduced Nox2 expression and ROS levels and that increases in Nox2 and Nox4 influence their differentiation into mature cells. We speculate that ROS generators Nox2 and Nox4, along with the antioxidant genes identified by PCR Arrays, may be novel targets in CPCs that could prove useful in cell-based therapy of the heart.
Collapse
Affiliation(s)
- Alyson S Nadworny
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York; and
| | | | | | | | | | | | | |
Collapse
|
84
|
Topchiy E, Panzhinskiy E, Griffin WST, Barger SW, Das M, Zawada WM. Nox4-generated superoxide drives angiotensin II-induced neural stem cell proliferation. Dev Neurosci 2013; 35:293-305. [PMID: 23751520 DOI: 10.1159/000350502] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/05/2013] [Indexed: 01/16/2023] Open
Abstract
Reactive oxygen species (ROS) have been reported to affect neural stem cell self-renewal and therefore may be important for normal development and may influence neurodegenerative processes when ROS activity is elevated. To determine if increasing production of superoxide, via activation of NADPH oxidase (Nox), increases neural stem cell proliferation, 100 nM angiotensin II (Ang II) - a strong stimulator of Nox - was applied to cultures of a murine neural stem cell line, C17.2. Twelve hours following a single treatment with Ang II, there was a doubling of the number of neural stem cells. This increase in neural stem cell numbers was preceded by a gradual elevation of superoxide levels (detected by dihydroethidium fluorescence) from the steady state at 0, 5, and 30 min and gradually increasing from 1 h to the maximum at 12 h, and returning to baseline at 24 h. Ang II-dependent proliferation was blocked by the antioxidant N-acetyl-L-cysteine. Confocal microscopy revealed the presence of two sources of intracellular ROS in C17.2 cells: (i) mitochondrial and (ii) extramitochondrial; the latter indicative of the involvement of one or more specific isoforms of Nox. Of the Nox family, mRNA expression for one member, Nox4, is abundant in neural stem cell cultures, and Ang II treatment resulted in elevation of the relative levels of Nox4 protein. SiRNA targeting of Nox4 mRNA reduced both the constitutive and Ang II-induced Nox4 protein levels and attenuated Ang II-driven increases in superoxide levels and stem cell proliferation. Our findings are consistent with our hypothesis that Ang II-induced proliferation of neural stem cells occurs via Nox4-generated superoxide, suggesting that an Ang II/Nox4 axis is an important regulator of neural stem cell self-renewal and as such may fine-tune normal, stress- or disease-modifying neurogenesis.
Collapse
Affiliation(s)
- Elena Topchiy
- Department of Behavioral Neuroscience, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Rd., mail code L470, Portland, OR 97239, USA.
| | | | | | | | | | | |
Collapse
|
85
|
Pendergrass KD, Boopathy AV, Seshadri G, Maiellaro-Rafferty K, Che PL, Brown ME, Davis ME. Acute preconditioning of cardiac progenitor cells with hydrogen peroxide enhances angiogenic pathways following ischemia-reperfusion injury. Stem Cells Dev 2013; 22:2414-24. [PMID: 23544670 DOI: 10.1089/scd.2012.0673] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
There are a limited number of therapies available to prevent heart failure following myocardial infarction. One novel therapy that is currently being pursued is the implantation of cardiac progenitor cells (CPCs); however, their responses to oxidative stress during differentiation have yet to be elucidated. The objective of this study was to determine the effect of hydrogen peroxide (H2O2) treatment on CPC differentiation in vitro, as well as the effect of H2O2 preconditioning before implantation following ischemia-reperfusion (I/R) injury. CPCs were isolated and cloned from adult rat hearts, and then cultured in the absence or presence of H2O2 for 2 or 5 days. CPC survival was assessed with Annexin V, and cellular differentiation was evaluated through mRNA expression for cardiogenic genes. We found that 100 μM H2O2 decreased serum withdrawal-induced apoptosis by at least 45% following both 2 and 5 days of treatment. Moreover, 100 μM H2O2 treatment for 2 days significantly increased endothelial and smooth muscle markers compared to time-matched untreated CPCs. However, continued H2O2 treatment significantly decreased these markers. Left ventricular cardiac function was assessed 28 days after I/R and I/R with the implantation of Luciferase/GFP(+) CPCs, which were preconditioned with 100 μM H2O2 for 2 days. Hearts implanted with Luciferase/GFP(+) CPCs had significant improvement in both positive and negative dP/dT over I/R. Furthermore, cardiac fibrosis was significantly decreased in the preconditioned cells versus both I/R alone and I/R with control cells. We also observed a significant increase in endothelial cell density in the preconditioned CPC hearts compared to untreated CPC hearts, which also coincided with a higher density of Luciferase(+) vessels. These findings suggest that preconditioning of CPCs with H2O2 for 2 days stimulates neoangiogenesis in the peri-infarct area following I/R injury and could be a viable therapeutic option to prevent heart failure.
Collapse
Affiliation(s)
- Karl D Pendergrass
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
86
|
Hirose K, Monzen S, Sato H, Sato M, Aoki M, Hatayama Y, Kawaguchi H, Narita Y, Takai Y, Kashiwakura I. Megakaryocytic differentiation in human chronic myelogenous leukemia K562 cells induced by ionizing radiation in combination with phorbol 12-myristate 13-acetate. JOURNAL OF RADIATION RESEARCH 2013; 54:438-446. [PMID: 23263730 PMCID: PMC3650750 DOI: 10.1093/jrr/rrs125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 11/25/2012] [Accepted: 11/27/2012] [Indexed: 06/01/2023]
Abstract
Differentiation-induction therapy is an attractive approach in leukemia treatment. It has been suggested that the accumulation of intracellular reactive oxygen species (ROS) is involved in megakaryocytic differentiation induced by phorbol 12-myristate 13-acetate (PMA) in the K562 leukemia cell line. Therefore, a ROS-inducible technique could be a powerful method of differentiation induction. Accordingly, we hypothesized that ionizing radiation contributes to the acceleration of megakaryocytic differentiation through the accumulation of intracellular ROS in leukemia cells. In the present study, ionizing radiation was shown to promote PMA-induced megakaryocytic differentiation. Cells with high CD41 expression sustained intracellular ROS levels effectively. The enhancement of differentiation by ionizing radiation was found to be regulated through the mitogen-activated protein kinase (MAPK) pathway, involving both extracellular signal-regulated protein kinase 1/2 (ERK1/2) and p38 MAPK. Ionizing radiation also controlled mRNA expression of the oxidative stress response gene heme oxygenase-1 (HO1). Consequently, we concluded that intracellular ROS, increased by ionizing radiation, modulate megakaryocytic differentiation downstream of the MAPK pathway.
Collapse
Affiliation(s)
- Katsumi Hirose
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Satoru Monzen
- Department of Radiological Life Sciences, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| | - Haruka Sato
- Department of Radiological Life Sciences, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| | - Mariko Sato
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Masahiko Aoki
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yoshiomi Hatayama
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hideo Kawaguchi
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yuichiro Narita
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yoshihiro Takai
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Ikuo Kashiwakura
- Department of Radiological Life Sciences, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| |
Collapse
|
87
|
Murray TVA, Smyrnias I, Shah AM, Brewer AC. NADPH oxidase 4 regulates cardiomyocyte differentiation via redox activation of c-Jun protein and the cis-regulation of GATA-4 gene transcription. J Biol Chem 2013; 288:15745-59. [PMID: 23589292 DOI: 10.1074/jbc.m112.439844] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NADPH oxidase 4 (Nox4) generates reactive oxygen species (ROS) that can modulate cellular phenotype and function in part through the redox modulation of the activity of transcription factors. We demonstrate here the potential of Nox4 to drive cardiomyocyte differentiation in pluripotent embryonal carcinoma cells, and we show that this involves the redox activation of c-Jun. This in turn acts to up-regulate GATA-4 expression, one of the earliest markers of cardiotypic differentiation, through a defined and highly conserved cis-acting motif within the GATA-4 promoter. These data therefore suggest a mechanism whereby ROS act in pluripotential cells in vivo to regulate the initial transcription of critical tissue-restricted determinant(s) of the cardiomyocyte phenotype, including GATA-4. The ROS-dependent activation, mediated by Nox4, of widely expressed redox-regulated transcription factors, such as c-Jun, is fundamental to this process.
Collapse
Affiliation(s)
- Thomas V A Murray
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London SE5 9NU, United Kingdom
| | | | | | | |
Collapse
|
88
|
Reactive oxygen species in vascular formation and development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:374963. [PMID: 23401740 PMCID: PMC3564431 DOI: 10.1155/2013/374963] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/29/2012] [Accepted: 12/29/2012] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are derived from the metabolism of oxygen and are traditionally viewed as toxic byproducts that cause damage to biomolecules. It is now becoming widely acknowledged that ROS are key modulators in a variety of biological processes and pathological states. ROS mediate key signaling transduction pathways by reversible oxidation of certain signaling components and are involved in the signaling of growth factors, G-protein-coupled receptors, Notch, and Wnt and its downstream cascades including MAPK, JAK-STAT, NF-κB, and PI3K/AKT. Vascular formation and development is one of the most important events during embryogenesis and is vital for postnasal tissue repair. In this paper, we will discuss how ROS regulate different steps in vascular development, including smooth muscle cell differentiation, angiogenesis, endothelial progenitor cells recruitment, and vascular cell migration.
Collapse
|
89
|
Urao N, Ushio-Fukai M. Redox regulation of stem/progenitor cells and bone marrow niche. Free Radic Biol Med 2013; 54:26-39. [PMID: 23085514 PMCID: PMC3637653 DOI: 10.1016/j.freeradbiomed.2012.10.532] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 01/19/2023]
Abstract
Bone marrow (BM)-derived stem and progenitor cell functions including self-renewal, differentiation, survival, migration, proliferation, and mobilization are regulated by unique cell-intrinsic and -extrinsic signals provided by their microenvironment, also termed the "niche." Reactive oxygen species (ROS), especially hydrogen peroxide (H(2)O(2)), play important roles in regulating stem and progenitor cell functions in various physiologic and pathologic responses. The low level of H(2)O(2) in quiescent hematopoietic stem cells (HSCs) contributes to maintaining their "stemness," whereas a higher level of H(2)O(2) within HSCs or their niche promotes differentiation, proliferation, migration, and survival of HSCs or stem/progenitor cells. Major sources of ROS are NADPH oxidase and mitochondria. In response to ischemic injury, ROS derived from NADPH oxidase are increased in the BM microenvironment, which is required for hypoxia and hypoxia-inducible factor-1α expression and expansion throughout the BM. This, in turn, promotes progenitor cell expansion and mobilization from BM, leading to reparative neovascularization and tissue repair. In pathophysiological states such as aging, atherosclerosis, heart failure, hypertension, and diabetes, excess amounts of ROS create an inflammatory and oxidative microenvironment, which induces cell damage and apoptosis of stem and progenitor cells. Understanding the molecular mechanisms of how ROS regulate the functions of stem and progenitor cells and their niche in physiological and pathological conditions will lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
90
|
Abstract
Redox signaling refers to the specific and usually reversible oxidation/reduction modification of molecules involved in cellular signaling pathways. In the heart, redox signaling regulates several physiological processes (eg, excitation-contraction coupling) and is involved in a wide variety of pathophysiological and homoeostatic or stress response pathways. Reactive oxygen species involved in cardiac redox signaling may derive from many sources, but NADPH oxidases, as dedicated sources of signaling reactive oxygen species, seem to be especially important. An increasing number of specific posttranslational oxidative modifications involved in cardiac redox signaling are being defined, along with the reactive oxygen species sources that are involved. Here, we review current knowledge on the molecular targets of signaling reactive oxygen species in cardiac cells and their involvement in cardiac physiopathology. Advances in this field may allow the development of targeted therapeutic strategies for conditions such as heart failure as opposed to the general antioxidant approaches that have failed to date.
Collapse
|
91
|
Shatynski KE, Chen H, Kwon J, Williams MS. Decreased STAT5 phosphorylation and GATA-3 expression in NOX2-deficient T cells: role in T helper development. Eur J Immunol 2012; 42:3202-11. [PMID: 22930452 PMCID: PMC3691816 DOI: 10.1002/eji.201242659] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/15/2012] [Accepted: 08/20/2012] [Indexed: 01/26/2023]
Abstract
Absence of phagocyte NADPH oxidase (NOX2) activity causes chronic granulomatous disease (CGD), a primary immunodeficiency characterized by recurrent bacterial infections. In contrast to this innate immune deficit, CGD patients and animal models display a predisposition toward autoimmune disease and enhanced response to Helicobacter pylori and influenza virus infection. These data imply an altered, perhaps augmented, adaptive immune response in CGD. As previous data demonstrated functional NOX2 expression in T cells, our goal here was to determine if NOX2-deficient T cells are inherently altered in their responses. Activation of purified naive CD4(+) T cells from NOX2-deficient mice led to augmented IFN-γ and diminished IL-4 production and an increased ratio of expression of the T(H)1-specific transcription factor T-bet versus the T(H)2-specfic transcription factor GATA-3, consistent with a T(H)1 skewing of naïve T cells. Selective inhibition of TCR-induced STAT5 phosphorylation was identified as a potential mechanism for skewed T helper differentiation. Exposure to antioxidants inhibited, while pro-oxidants augmented T(H)2 cytokine secretion and STAT5 phosphorylation, supporting the redox dependence of these signaling changes. These data suggest that TCR-induced ROS generation from NOX2 activation can regulate the adaptive immune response in a T-cell-inherent fashion, and propose a possible role for redox signaling in T helper differentiation.
Collapse
MESH Headings
- Animals
- Bacterial Infections/enzymology
- Bacterial Infections/genetics
- Bacterial Infections/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- GATA3 Transcription Factor/biosynthesis
- GATA3 Transcription Factor/genetics
- GATA3 Transcription Factor/immunology
- Granulomatous Disease, Chronic/genetics
- Granulomatous Disease, Chronic/immunology
- Granulomatous Disease, Chronic/metabolism
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-4/genetics
- Interleukin-4/immunology
- Interleukin-4/metabolism
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Knockout
- NADPH Oxidase 2
- NADPH Oxidases/genetics
- NADPH Oxidases/immunology
- NADPH Oxidases/metabolism
- Oxidation-Reduction
- Phosphorylation/genetics
- Phosphorylation/immunology
- Reactive Oxygen Species/immunology
- Reactive Oxygen Species/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/immunology
- STAT5 Transcription Factor/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th1 Cells/pathology
- Th2 Cells/immunology
- Th2 Cells/metabolism
Collapse
Affiliation(s)
- Kristen E. Shatynski
- Department of Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 W. Baltimore Street, Baltimore, MD 21201
| | - Haiyan Chen
- Department of Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 W. Baltimore Street, Baltimore, MD 21201
| | - Jaeyul Kwon
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12441 Parklawn Drive, Rockville, MD 20852
| | - Mark S. Williams
- Department of Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 W. Baltimore Street, Baltimore, MD 21201
| |
Collapse
|
92
|
Italiano D, Lena AM, Melino G, Candi E. Identification of NCF2/p67phox as a novel p53 target gene. Cell Cycle 2012. [PMID: 23187810 DOI: 10.4161/cc.22853] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Analysis of microarrays performed in p53-, TAp63α- and ΔNp63α-inducible SaOs-2 cell lines allowed the identification of NCF2 mRNA upregulation in response to p53 induction. NCF2 gene encodes for p67phox, the cytosolic subunit of the NADPH oxidase enzyme complex. The recruitment of p67phox to the cell membrane causes the activation of the NADPH oxidase complex followed by the generation of NADP+ and superoxide from molecular oxygen. The presence of three putative p53 binding sites on the NCF2 promoter was predicted, and the subsequent luciferase and chromatin immunoprecipitation assays showed the activation of NCF2 promoter by p53 and its direct binding in vivo to at least one of the sites, thus confirming the hypothesis. NCF2 upregulation was also confirmed by real-time PCR in several cell lines after p53 activation. NCF2 knockdown by siRNA results in a significant reduction of ROS production and stimulates cell death, suggesting a protective function of Nox2-generated ROS in cells against apoptosis. These results provide insight into the redox-sensitive signaling mechanism that mediates cell survival involving p53 and its novel target NCF2/p67phox.
Collapse
Affiliation(s)
- Dafne Italiano
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy
| | | | | | | |
Collapse
|
93
|
Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ Res 2012; 111:344-58. [PMID: 22821908 DOI: 10.1161/circresaha.110.227512] [Citation(s) in RCA: 544] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since human embryonic stem cells were first differentiated to beating cardiomyocytes a decade ago, interest in their potential applications has increased exponentially. This has been further enhanced over recent years by the discovery of methods to induce pluripotency in somatic cells, including those derived from patients with hereditary cardiac diseases. Human pluripotent stem cells have been among the most challenging cell types to grow stably in culture, but advances in reagent development now mean that most laboratories can expand both embryonic and induced pluripotent stem cells robustly using commercially available products. However, differentiation protocols have lagged behind and in many cases only produce the cell types required with low efficiency. Cardiomyocyte differentiation techniques were also initially inefficient and not readily transferable across cell lines, but there are now a number of more robust protocols available. Here, we review the basic biology underlying the differentiation of pluripotent cells to cardiac lineages and describe current state-of-the-art protocols, as well as ongoing refinements. This should provide a useful entry for laboratories new to this area to start their research. Ultimately, efficient and reliable differentiation methodologies are essential to generate desired cardiac lineages to realize the full promise of human pluripotent stem cells for biomedical research, drug development, and clinical applications.
Collapse
Affiliation(s)
- Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
94
|
Juurlink BHJ. Dietary Nrf2 activators inhibit atherogenic processes. Atherosclerosis 2012; 225:29-33. [PMID: 22986182 DOI: 10.1016/j.atherosclerosis.2012.08.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 08/23/2012] [Accepted: 08/25/2012] [Indexed: 12/21/2022]
Abstract
Dietary Nrf2 activators increase expression of phase 2 protein genes in cells undergoing oxidative stress resulting in a lowering of oxidative stress. Oxidative stress promotes atherogenic processes through oxidizing low density lipoproteins and promotion of inflammation through activation of nuclear factor kappa B and activation of mitogen-activated protein kinases. Nrf2 activators by decreasing oxidative stress decrease the probability of developing atherosclerotic lesions.
Collapse
Affiliation(s)
- Bernhard H J Juurlink
- College of Medicine, University of Saskatchewan, SK, Canada; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
95
|
Folmes CDL, Nelson TJ, Dzeja PP, Terzic A. Energy metabolism plasticity enables stemness programs. Ann N Y Acad Sci 2012; 1254:82-89. [PMID: 22548573 DOI: 10.1111/j.1749-6632.2012.06487.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Engineering pluripotency through nuclear reprogramming and directing stem cells into defined lineages underscores cell fate plasticity. Acquisition of and departure from stemness are governed by genetic and epigenetic controllers, with modulation of energy metabolism and associated signaling increasingly implicated in cell identity determination. Transition from oxidative metabolism, typical of somatic tissues, into glycolysis is a prerequisite to fuel-proficient reprogramming, directing a differentiated cytotype back to the pluripotent state. The glycolytic metabotype supports the anabolic and catabolic requirements of pluripotent cell homeostasis. Conversely, redirection of pluripotency into defined lineages requires mitochondrial biogenesis and maturation of efficient oxidative energy generation and distribution networks to match demands. The vital function of bioenergetics in regulating stemness and lineage specification implicates a broader role for metabolic reprogramming in cell fate decisions and determinations of tissue regenerative potential.
Collapse
Affiliation(s)
- Clifford D L Folmes
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
96
|
|
97
|
Abstract
The main objective of this review is to examine the role of endogenous reactive oxygen/nitrogen species (ROS) in the aging process. Until relatively recently, ROS were considered to be potentially toxic by-products of aerobic metabolism, which, if not eliminated, may inflict structural damage on various macromolecules. Accrual of such damage over time was postulated to be responsible for the physiological deterioration in the postreproductive phase of life and eventually the death of the organism. This "structural damage-based oxidative stress" hypothesis has received support from the age-associated increases in the rate of ROS production and the steady-state amounts of oxidized macromolecules; however, there are increasing indications that structural damage alone is insufficient to satisfactorily explain the age-associated functional losses. The level of oxidative damage accrued during aging often does not match the magnitude of functional losses. Although experimental augmentation of antioxidant defenses tends to enhance resistance to induced oxidative stress, such manipulations are generally ineffective in the extension of life span of long-lived strains of animals. More recently, in a major conceptual shift, ROS have been found to be physiologically vital for signal transduction, gene regulation, and redox regulation, among others, implying that their complete elimination would be harmful. An alternative notion, advocated here, termed the "redox stress hypothesis," proposes that aging-associated functional losses are primarily caused by a progressive pro-oxidizing shift in the redox state of the cells, which leads to the overoxidation of redox-sensitive protein thiols and the consequent disruption of the redox-regulated signaling mechanisms.
Collapse
Affiliation(s)
- Rajindar S Sohal
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - William C Orr
- Department of Biological Sciences, Southern Methodist University, Dallas, TX 75275, USA
| |
Collapse
|
98
|
Srinivasa K, Kim J, Yee S, Kim W, Choi W. A MAP kinase pathway is implicated in the pseudohyphal induction by hydrogen peroxide in Candica albicans. Mol Cells 2012; 33:183-93. [PMID: 22358510 PMCID: PMC3887715 DOI: 10.1007/s10059-012-2244-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 12/12/2011] [Accepted: 12/14/2011] [Indexed: 12/20/2022] Open
Abstract
Hydrogen peroxide (H(2)O(2)) functions as a ubiquitous intracellular messenger besides as an oxidative stress molecule. This dual role is based on the distinct cellular responses against different concentrations of H(2)O(2). Previously, we demonstrated that both low (> 1 mM) and high (4-10 mM) doses of exogenous H(2)O(2) induce filamentous growth with distinct cell morphology and growth rate in Candida albicans, suggesting the different transcription response. In this study, we revealed that the sub-toxic and toxic levels of H(2)O(2) indeed induced pseudohyphae, but not true hyphae. Supporting this, several hyphae-specific genes that are expressed in true hyphae induced by serum were not detected in either sub-toxic or toxic H(2)O(2) condition. A DNA microarray analysis was conducted to reveal the transcription profiles in cells treated with sub-toxic and toxic conditions of H(2)O(2). Under the sub-toxic condition, a small number of genes involved in cell proliferation and metabolism were up-regulated, whereas a large number of genes were up-regulated in the toxic condition where the genes required for growth and proliferation were selectively restricted. For pseudohyphal induction by sub-toxic H(2)O(2), Cek1 MAPK activating the transcription factor Cph1 was shown to be important. The absence of expression of several hyphae-specific genes known to be downstream targets of Cph1-signaling pathway for true hyphae formation suggests that the Cek1-mediated signaling pathway is not solely responsible for pseudohyphal formation by subtoxic H(2)O(2) and, but instead, complex networking pathway may exists by the activation of different regulators.
Collapse
Affiliation(s)
- Kavitha Srinivasa
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750,
Korea
| | - Jihyun Kim
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750,
Korea
| | - Subog Yee
- Microbial Resources Research Center, Ewha Womans University, Seoul 120-750,
Korea
| | - Wankee Kim
- Institute for Medical Sciences, School of Medicine, Ajou University, Suwon 442-749,
Korea
| | - Wonja Choi
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750,
Korea
- Microbial Resources Research Center, Ewha Womans University, Seoul 120-750,
Korea
| |
Collapse
|
99
|
Kennedy KAM, Sandiford SDE, Skerjanc IS, Li SSC. Reactive oxygen species and the neuronal fate. Cell Mol Life Sci 2012; 69:215-21. [PMID: 21947442 PMCID: PMC11114775 DOI: 10.1007/s00018-011-0807-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 07/29/2011] [Accepted: 08/18/2011] [Indexed: 10/17/2022]
Abstract
Aberrant or elevated levels of reactive oxygen species (ROS) can mediate deleterious cellular effects, including neuronal toxicity and degeneration observed in the etiology of a number of pathological conditions, including Alzheimer's and Parkinson's diseases. Nevertheless, ROS can be generated in a controlled manner and can regulate redox sensitive transcription factors such as NFκB, AP-1 and NFAT. Moreover, ROS can modulate the redox state of tyrosine phosphorylated proteins, thereby having an impact on many transcriptional networks and signaling cascades important for neurogenesis. A large body of literature links the controlled generation of ROS at low-to-moderate levels with the stimulation of differentiation in certain developmental programs such as neurogenesis. In this regard, ROS are involved in governing the acquisition of the neural fate-from neural induction to the elaboration of axons. Here, we summarize and discuss the growing body of literature that describe a role for ROS signaling in neuronal development.
Collapse
Affiliation(s)
- Karen A. M. Kennedy
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1 Canada
| | - Shelley D. E. Sandiford
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1 Canada
| | - Ilona S. Skerjanc
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5 Canada
| | - Shawn S.-C. Li
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1 Canada
| |
Collapse
|
100
|
Abstract
Continuous developmental maturation of cardiomyocytes is essential to meet the functional and metabolic demands of the growing heart. A new study (Hom et al., 2011) reports that embryonic cardiomyocytes are influenced by mitochondrial maturation, such that closure of the mitochondrial permeability transition pore results in decreased levels of reactive oxygen species, thereby inducing differentiation.
Collapse
|