1
|
Cui A, Li S, Li Y, Yang D, Huang J, Wang X, Song N, Chen F, Chen S, Xiang M. Nitric oxide-mediated the therapeutic properties of induced pluripotent stem cell for paraquat-induced acute lung injury. Front Immunol 2023; 14:1136290. [PMID: 37275899 PMCID: PMC10232993 DOI: 10.3389/fimmu.2023.1136290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
The mortality rate associated with acute lung injury (ALI) and its severe form, acute respiratory distress syndrome, is high. Induced pluripotent stem cell (iPSC) therapy is a potential treatment method for ALI, but its therapeutic efficacy is limited in injured lungs. Nitric oxide (NO) has various physiological actions. The current study investigated the effect of iPSCs pretreated with NO donors in paraquat (PQ)-induced ALI mouse model. Male C57BL/6 mice were intraperitoneally injected with PQ, followed by infusion of phosphate-buffered saline, iPSCs, L-arginine pretreated iPSCs, or Nitro-L-arginine methylester (L-NAME) pretreated iPSCs through the tail veins. Histopathological changes, pulmonary microvascular permeability, and inflammatory cytokine levels were analyzed after 3 or 28 d. The effects on iPSC proliferation, migration, and adhesion were evaluated in vitro. More L-arginine-pretreated iPSCs were selectively trafficked into the injured pulmonary tissue of mice with LPS-induced ALI, drastically diminishing the histopathologic changes and inflammatory cytokine levels (IL-1β and IL-6). There was also markedly improved pulmonary microvascular permeability and pulmonary function. The NO inhibitor abolished the protective effects of iPSCs. In addition, the ability of L-arginine to promote the proliferation and migration of iPSCs was decreased by L-NAME pretreatment, suggesting that NO might mediate the therapeutic benefits of iPSC. The improvement of the iPSC physiological changes by the endogenous gaseous molecule NO reduces lung injury severity. L-Arginine represents a pharmacologically important strategy for enhancing the therapeutic potential of iPSCs.
Collapse
Affiliation(s)
- Anfeng Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Pathology, The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shirui Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yijun Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dawei Yang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jiongwei Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Health Management Center Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuemeng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nana Song
- Fudan Zhang Jiang Institute, Shanghai, China
| | - Fuchen Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meng Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Fudan Zhang Jiang Institute, Shanghai, China
| |
Collapse
|
2
|
Koudan EV, Zharkov MN, Gerasimov MV, Karshieva SS, Shirshova AD, Chrishtop VV, Kasyanov VA, Levin AA, Parfenov VA, Karalkin PA, Pereira FDAS, Petrov SV, Pyataev NA, Khesuani YD, Mironov VA, Sukhorukov GB. Magnetic Patterning of Tissue Spheroids Using Polymer Microcapsules Containing Iron Oxide Nanoparticles. ACS Biomater Sci Eng 2021; 7:5206-5214. [PMID: 34610738 DOI: 10.1021/acsbiomaterials.1c00805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Magnetic tissue engineering is one of the rapidly emerging and promising directions of tissue engineering and biofabrication where the magnetic field is employed as temporal removal support or scaffold. Iron oxide nanoparticles are used to label living cells and provide the desired magnetic properties. Recently, polymer microcapsules loaded with iron oxide nanoparticles have been proposed as a novel approach to designing magnetic materials with high local concentrations. These microcapsules can be readily internalized and retained intracellularly for a long time in various types of cells. The low cytotoxicity of these microcapsules was previously shown in 2D cell culture. This paper has demonstrated that cells containing these nontoxic nanomaterials can form viable 3D tissue spheroids for the first time. The spheroids retained labeled fluorescent microcapsules with magnetic nanoparticles without a detectable cytotoxic effect. The high concentration of packed nanoparticles inside the microcapsules enables the evident magnetic properties of the labeled spheroids to be maintained. Finally, magnetic spheroids can be effectively used for magnetic patterning and biofabrication of tissue-engineering constructs.
Collapse
Affiliation(s)
- Elizaveta V Koudan
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia
| | - Mikhail N Zharkov
- National Research Ogarev Mordovia State University, Bolshevistskaya Str. 68-1, Saransk 430005, Russia
| | - Mikhail V Gerasimov
- National Research Ogarev Mordovia State University, Bolshevistskaya Str. 68-1, Saransk 430005, Russia
| | - Saida Sh Karshieva
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia.,Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Kashirskoe Highway 23, Moscow 115478, Russia
| | | | - Vladimir V Chrishtop
- SCAMT Institute, ITMO University, Lomonosova Str. 9, Saint Petersburg 191002, Russia
| | | | - Aleksandr A Levin
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia
| | - Vladislav A Parfenov
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia
| | - Pavel A Karalkin
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia.,I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Bolshaya Pirogovskaya Str. 2-4, Moscow 119991, Russia.,P. Hertsen Moscow Oncology Research Institute, National Medical Research Radiological Centre, 2 Botkinskiy proezd 3, Moscow 125284, Russia
| | - Frederico D A S Pereira
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia
| | - Stanislav V Petrov
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia
| | - Nikolay A Pyataev
- National Research Ogarev Mordovia State University, Bolshevistskaya Str. 68-1, Saransk 430005, Russia
| | - Yusef D Khesuani
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia
| | - Vladimir A Mironov
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Kashirskoe Highway 68-2, Moscow 115409, Russia.,I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Bolshaya Pirogovskaya Str. 2-4, Moscow 119991, Russia
| | - Gleb B Sukhorukov
- School of Engineering and Material Science, Queen Mary University of London,Mile End Road, London E1 4NS, United Kingdom.,Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia
| |
Collapse
|
3
|
Dutta N, Ghosh S, Nelson VK, Sareng HR, Majumder C, Mandal SC, Pal M. Andrographolide upregulates protein quality control mechanisms in cell and mouse through upregulation of mTORC1 function. Biochim Biophys Acta Gen Subj 2021; 1865:129885. [PMID: 33639218 DOI: 10.1016/j.bbagen.2021.129885] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Heat shock response (HSR), a component of cellular protein quality control mechanisms, is defective in different neurodegenerative conditions such as Parkinson's disease (PD). Forced upregulation of heat shock factor 1 (HSF1), an HSR master regulator, showed therapeutic promise in PD models. Many of the reported small-molecule HSF1 activators have limited functions. Therefore, identification and understanding the molecular bases of action of new HSF1 activating molecules is necessary. METHOD We used a cell-based reporter system to screen Andrographis paniculata leaf extract to isolate andrographolide as an inducer of HSF1 activity. The andrographolide activity was characterized by analyzing its role in different protein quality control mechanisms. RESULT We find that besides ameliorating the PD in MPTP-treated mice, andrographolide upregulated different machineries controlled by HSF1 and NRF2 in both cell and mouse brain. Andrographolide achieves these functions through mTORC1 activated via p38 MAPK and ERK pathways. NRF2 activation is reflected in the upregulation of proteasome as well as autophagy pathways. We further show that NRF2 activation is mediated through mTORC1 driven phosphorylation of p62/sequestosome 1. Studies with different cell types suggested that andrographolide-mediated induction of ROS level underlies all these activities in agreement with the upregulation of mTORC1 and NRF2-antioxidant pathway in mice. CONCLUSION Andrographolide through upregulating HSF1 activity ameliorates protein aggregation induced cellular toxicity. GENERAL SIGNIFICANCE Our results provide a reasonable basis for use of andrographolide in the therapy regimen for the treatment of PD.
Collapse
Affiliation(s)
- Naibedya Dutta
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Vinod K Nelson
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Hossainoor R Sareng
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Chirantan Majumder
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Subhash C Mandal
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India.
| |
Collapse
|
4
|
Intracellular Reactive Oxygen Species Mediate the Therapeutic Effect of Induced Pluripotent Stem Cells for Acute Kidney Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1609638. [PMID: 32308798 PMCID: PMC7136790 DOI: 10.1155/2020/1609638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/03/2020] [Accepted: 02/19/2020] [Indexed: 11/17/2022]
Abstract
Aims Treatment for acute kidney injury (AKI) is challenging. Induced pluripotent stem cells (iPSCs) have great therapeutic potential. This study sought to determine whether iPSCs attenuate AKI and the role of reactive oxygen species (ROS). Results We intravenously injected isogenic iPSCs into mice 2 h after renal ischemia-reperfusion injury (IRI). The cells were selectively trafficked to ischemia/reperfusion-injured kidney where they decreased kidney ROS and inflammatory cytokines and improved kidney function and morphology. Pretreating the cells with ROS inhibitors before administration decreased iPSC engraftment and abolished the protective effect of iPSCs. In contrast, pretreating iPSCs with hydrogen peroxide increased iPSC engraftment and therapeutic effect. Although the intravenously administered iPSCs trafficked to the IRI kidney, the cells did not differentiate into proximal or distal tubular epithelial cells. In vitro, the capabilities of the iPSC-released substances to promote proliferation and decrease apoptosis of renal epithelial cells were increased by ROS pretreatment of iPSCs. Moreover, pretreatment of the iPSCs with ROS inhibitor had the opposite effect. Similarly, moderate concentrations of ROS increased while ROS inhibitors decreased iPSC mobility, adhesion to the extracellular matrix, and mitochondrial metabolism. Innovation and Conclusion. iPSCs decreased renal ischemia/reperfusion injury mainly through iPSC-released substances. The therapeutic effect, mitochondrial metabolism, mobility, and kidney trafficking of iPSCs were ROS dependent.
Collapse
|
5
|
Guan F, Huang T, Wang X, Xing Q, Gumpper K, Li P, Song J, Tan T, Yang GL, Zang X, Zhang J, Wang Y, Yang Y, Liu Y, Zhang Y, Yang B, Ma J, Ma S. The TRIM protein Mitsugumin 53 enhances survival and therapeutic efficacy of stem cells in murine traumatic brain injury. Stem Cell Res Ther 2019; 10:352. [PMID: 31779687 PMCID: PMC6883632 DOI: 10.1186/s13287-019-1433-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/22/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a common neurotrauma leading to brain dysfunction and death. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) hold promise in the treatment of TBI. However, their efficacy is modest due to low survival and differentiation under the harsh microenvironment of the injured brain. MG53, a member of TRIM family protein, plays a vital role in cell and tissue damage repair. The present study aims to test whether MG53 preserves hUC-MSCs against oxidative stress and enhances stem cell survival and efficacy in TBI treatment. Methods In this study, we performed a series of in vitro and in vivo experiments in hUC-MSCs and mice to define the function of MG53 enhancing survival, neurogenesis, and therapeutic efficacy of stem cells in murine traumatic brain injury. Results We found that recombinant human MG53 (rhMG53) protein protected hUC-MSCs against H2O2-induced oxidative damage and stimulated hUC-MSC proliferation and migration. In a mouse model of contusion-induced TBI, intravenous administration of MG53 protein preserved the survival of transplanted hUC-MSCs, mitigated brain edema, reduced neurological deficits, and relieved anxiety and depressive-like behaviors. Co-treatment of MG53 and hUC-MSCs enhanced neurogenesis by reducing apoptosis and improving PI3K/Akt-GSK3β signaling. Conclusion MG53 enhances the efficacy of hUC-MSCs in the recovery of TBI, indicating that such adjunctive therapy may provide a novel strategy to lessen damage and optimize recovery for brain injury.
Collapse
Affiliation(s)
- Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.,Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Tuanjie Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xinxin Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Kristyn Gumpper
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Peng Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jishi Song
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | | | - Xingxing Zang
- Department of Microbiology and Immunology, Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Jiewen Zhang
- Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Yuming Wang
- Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Yunlei Yang
- Department of Medicine and Neuroscience, Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Yashi Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
6
|
Noll B, Bahrani Mougeot F, Brennan MT, Mougeot JLC. Telomere erosion in Sjögren's syndrome: A multi-tissue comparative analysis. J Oral Pathol Med 2019; 49:63-71. [PMID: 31529714 DOI: 10.1111/jop.12961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Acinar progenitor cells within salivary glands have decreased regenerative capacity and exhibit shorter telomeres in primary Sjögren's syndrome (pSS) patients. We investigated whether DNA of saliva, PBMCs, and labial salivary gland (LSG) biopsy tissue have shorter telomeres in pSS compared to controls. mRNA expression of genes associated with pSS pathogenesis (ETS1, LEF1, and MMP9), telomere DNA damage response (ATM), senescence (CDKN2A), telomerase inhibition (IFN-y, TGFβ1), and the shelterin complex (TPP1, POT1) were assessed in LSG tissue by qRT-PCR to examine potential defects in telomere maintenance. METHODS Relative telomere length in DNA of saliva, PBMCs, and LSGs from non-pSS sicca and pSS patients was measured using qPCR. Saliva DNA telomere length was further compared to healthy controls. Expression of genes affecting telomere maintenance was analyzed in LSGs using qRT-PCR. RESULTS Primary Sjögren's syndrome patients have shorter telomeres in saliva DNA (n = 21) than healthy controls (n = 27) (P = .0035). ATM mRNA expression was higher in pSS LSG tissue (n = 16) vs non-pSS sicca patients (n = 13) (P = .0283) and strongly correlated with LEF1, TPP1, and POT1 (P < .01, r > 0.6). CONCLUSIONS Patients with pSS exhibited significant telomere erosion in saliva DNA. Overexpression of ATM in LSGs could represent a compensatory response to telomere shortening. The role of LEF1 in telomere erosion remains to be elucidated.
Collapse
Affiliation(s)
- Braxton Noll
- Department of Oral Medicine, Carolinas Medical Center - Atrium Health, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Farah Bahrani Mougeot
- Department of Oral Medicine, Carolinas Medical Center - Atrium Health, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Michael T Brennan
- Department of Oral Medicine, Carolinas Medical Center - Atrium Health, Charlotte, NC, USA
| | - Jean-Luc C Mougeot
- Department of Oral Medicine, Carolinas Medical Center - Atrium Health, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
7
|
Ye Q, Feng Y, Wang Z, Jiang W, Qu Y, Zhang C, Zhou A, Xie S, Zou J. Effects of gelsemine on oxidative stress and DNA damage responses of Tetrahymena thermophila. PeerJ 2018; 6:e6093. [PMID: 30581679 PMCID: PMC6292385 DOI: 10.7717/peerj.6093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/10/2018] [Indexed: 01/01/2023] Open
Abstract
Gelsemine is an important toxic substance extracted from Gelsemium elegans, which has a lot of biological functions in cells and organisms, but its toxicity has been rarely reported in Tetrahymena thermophila. In this study, we used the protozoan T. thermophila as an experimental model to investigate the potential toxicity-induced mechanism of gelsemine in the unicellular eukaryote. Our results clearly showed gelsemine inhibited T. thermophila growth in a dose-dependent manner. This exposure also resulted in oxidative stress on T. thermophila cells and antioxidant enzyme levels were significantly altered at high gelsemine levels (p < 0.05). Gelsemine produced a slight apoptotic effect at the highest (0.8 mg/mL) gelsemine level used here (p < 0.05). Furthermore, the toxin-induced DNA damage in a dose-dependent manner. The ultrastructural analysis also revealed mitophagic vacuoles at 0.4 and 0.8 mg/mL levels of gelsemine exposure. Moreover, expressions of oxidative stress-related and MAP kinase genes were significantly changed after exposure to 0.8 mg/mL level of gelsemine (p < 0.05). Altogether, our results clearly show that gelsemine from G. elegans can inhibit the growth via inducing oxidative stress and DNA damage in T. thermophila cells.
Collapse
Affiliation(s)
- Qiao Ye
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yongyong Feng
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhenlu Wang
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wenzhao Jiang
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yuexin Qu
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Chaonan Zhang
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Aiguo Zhou
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shaolin Xie
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jixing Zou
- Healthy Aquaculture Laboratory, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Meng S, Chanda P, Thandavarayan RA, Cooke JP. Transflammation: How Innate Immune Activation and Free Radicals Drive Nuclear Reprogramming. Antioxid Redox Signal 2018; 29:205-218. [PMID: 29634341 PMCID: PMC6003401 DOI: 10.1089/ars.2017.7364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Yamanaka and colleagues galvanized the field of stem cell biology and regenerative medicine by their generation of induced pluripotent stem cells. Evidence is emerging that activation of innate immune signaling is critical for efficient reprogramming to pluripotency and for the nuclear reprogramming occurring in transdifferentiation. Recent Advances: We have shown that innate immune signaling triggers a global change in the expression of epigenetic modifiers to enhance DNA accessibility. In this state of epigenetic plasticity, overexpression of lineage determination factors, and/or environmental cues and paracrine factors, can induce pluripotency, or can direct transdifferentiation to another somatic cell lineage. Accumulating evidence reveals that innate immune activation triggers the generation of reactive oxygen species and reactive nitrogen species, and that these free radicals are required for nuclear reprogramming to pluripotency or for transdifferentiation. CRITICAL ISSUES We have discovered a limb of innate immune signaling that regulates DNA accessibility, in part, by the action of free radicals to induce post-translational modification of epigenetic modifiers. FUTURE DIRECTIONS It is of scientific interest and clinical relevance to understand the mechanisms by which free radicals influence epigenetic plasticity, and how these mechanisms may be therapeutically modulated. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Shu Meng
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| | - Palas Chanda
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| | - Rajarajan A Thandavarayan
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| |
Collapse
|
9
|
Selection of antioxidants against ovarian oxidative stress in mouse model. J Biochem Mol Toxicol 2017; 31. [DOI: 10.1002/jbt.21997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/16/2017] [Accepted: 08/25/2017] [Indexed: 01/01/2023]
|
10
|
Kang X, Wei X, Jiang L, Niu C, Zhang J, Chen S, Meng D. Nox2 and Nox4 regulate self-renewal of murine induced-pluripotent stem cells. IUBMB Life 2016; 68:963-970. [PMID: 27797149 DOI: 10.1002/iub.1574] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/19/2016] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) and redox homeostasis have a pivotal role in the maintenance of stem cell pluripotency and in stem cell self-renewal; however, the mechanisms by which ROS regulate the self-renewal of stem cells have not been thoroughly studied. Here, we evaluated the role of the ROS produced by NADPH oxidase 2 (Nox2) and NADPH oxidase 4 (Nox4) in the self-renewal and stemness of murine induced-pluripotent stem cells (miPSCs). Targeted silencing of Nox2 or Nox4 reduced both NADPH oxidase activity and intracellular ROS levels, as well as alkaline phosphatase activity, the total number of miPSCs, the expression of insulin-like growth factor-1 (IGF-1), IGF-1 receptor, and the phosphorylation of extracellular signal regulated kinase (ERK) 1/2. Nox2/Nox4 overexpression or low, nontoxic concentration of H2 O2 increased cell proliferation in miPSCs. Furthermore, expression of the stemness genes Sox2 and Oct4 was lower in Nox2/Nox4-deficient miPSCs, and higher in Nox2/Nox4-overexpressing miPSCs, than in miPSCs with normal levels of Nox2/Nox4 expression. Collectively, these results suggest that Nox2- and Nox4-derived ROS contribute to stem cell pluripotency maintenance and self-renewal. © 2016 IUBMB Life, 68(12):963-970, 2016.
Collapse
Affiliation(s)
- Xueling Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Cong Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Fang D, Qing Y, Yan S, Chen D, Yan SS. Development and Dynamic Regulation of Mitochondrial Network in Human Midbrain Dopaminergic Neurons Differentiated from iPSCs. Stem Cell Reports 2016; 7:678-692. [PMID: 27666790 PMCID: PMC5063542 DOI: 10.1016/j.stemcr.2016.08.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 02/05/2023] Open
Abstract
Mitochondria are critical to neurogenesis, but the mechanisms of mitochondria in neurogenesis have not been well explored. We fully characterized mitochondrial alterations and function in relation to the development of human induced pluripotent stem cell (hiPSC)-derived dopaminergic (DA) neurons. Following directed differentiation of hiPSCs to DA neurons, mitochondria in these neurons exhibit pronounced changes during differentiation, including mature neurophysiology characterization and functional synaptic network formation. Inhibition of mitochondrial respiratory chains via application of complex IV inhibitor KCN (potassium cyanide) or complex I inhibitor rotenone restricted neurogenesis of DA neurons. These results demonstrated the direct importance of mitochondrial development and bioenergetics in DA neuronal differentiation. Our study also provides a neurophysiologic model of mitochondrial involvement in neurogenesis, which will enhance our understanding of the role of mitochondrial dysfunctions in neurodegenerative diseases. Mitochondria are essential for the development of hiPSC-derived DA neurons Mitochondrial defects suppress maturation and synaptogenesis of DA neurons ROS levels positively correlate to DA neuron maturation and synaptic formation A model of crosstalk of mitochondrial network to neurogenesis of DA neurons
Collapse
Affiliation(s)
- Du Fang
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, School of Pharmacy, University of Kansas, 2099 Constant Avenue, Lawrence, KS 66047, USA
| | - Yu Qing
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, School of Pharmacy, University of Kansas, 2099 Constant Avenue, Lawrence, KS 66047, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Cheng Du 610041, China
| | - Shijun Yan
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, School of Pharmacy, University of Kansas, 2099 Constant Avenue, Lawrence, KS 66047, USA
| | - Doris Chen
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, School of Pharmacy, University of Kansas, 2099 Constant Avenue, Lawrence, KS 66047, USA
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, School of Pharmacy, University of Kansas, 2099 Constant Avenue, Lawrence, KS 66047, USA.
| |
Collapse
|
12
|
Nox2 contributes to the arterial endothelial specification of mouse induced pluripotent stem cells by upregulating Notch signaling. Sci Rep 2016; 6:33737. [PMID: 27642005 PMCID: PMC5027389 DOI: 10.1038/srep33737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) have a crucial role in stem-cell differentiation; however, the mechanisms by which ROS regulate the differentiation of stem cells into endothelial cells (ECs) are unknown. Here, we determine the role of ROS produced by NADPH oxidase 2 (Nox2) in the endothelial-lineage specification of mouse induced-pluripotent stem cells (miPSCs). When wild-type (WT) and Nox2-knockout (Nox2−/−) miPSCs were differentiated into ECs (miPSC-ECs), the expression of endothelial markers, arterial endothelial markers, pro-angiogenic cytokines, and Notch pathway components was suppressed in the Nox2−/− cells but increased in both WT and Nox2−/− miPSCs when Nox2 expression was upregulated. Higher levels of Nox2 expression increased Notch signaling and arterial EC differentiation, and this increase was abolished by the inhibition of ROS generation or by the silencing of Notch1 expression. Nox2 deficiency was associated with declines in the survival and angiogenic potency of miPSC-ECs, and capillary and arterial density were lower in the ischemic limbs of mice after treatment with Nox2−/− miPSC-ECs than WT miPSC-EC treatment. Taken together, these observations indicate that Nox2-mediated ROS production promotes arterial EC specification in differentiating miPSCs by activating the Notch signaling pathway and contributes to the angiogenic potency of transplanted miPSC-derived ECs.
Collapse
|
13
|
Fang D, Yan S, Yu Q, Chen D, Yan SS. Mfn2 is Required for Mitochondrial Development and Synapse Formation in Human Induced Pluripotent Stem Cells/hiPSC Derived Cortical Neurons. Sci Rep 2016; 6:31462. [PMID: 27535796 PMCID: PMC4989148 DOI: 10.1038/srep31462] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are essential dynamic organelles for energy production. Mitochondria dynamically change their shapes tightly coupled to fission and fusion. Imbalance of fission and fusion can cause deficits in mitochondrial respiration, morphology and motility. Mfn2 (mitofusin 2), a mitochondrial membrane protein that participates in mitochondrial fusion in mammalian cells, contributes to the maintenance and operation of the mitochondrial network. Due to lack of applicable model systems, the mechanisms and involvement of mitochondria in neurogenesis in human brain cells have not been well explored. Here, by employing the human induced pluripotent stem cells (hiPSCs) differentiation system, we fully characterized mitochondrial development, neurogenesis and synapse formation in hiPSCs-derived cortical neurons. Differentiation of hiPSCs to cortical neurons with extended period demonstrates mature neurophysiology characterization and functional synaptic network formation. Mitochondrial respiration, morphology and motility in the differentiated neurons also exhibit pronounced development during differentiation. Mfn2 knock-down results in deficits in mitochondrial metabolism and network, neurogenesis and synapse formation, while Mfn2 overexpression enhances mitochondrial bioenergetics and functions, and promotes the differentiation and maturation of neurons. Together, our data indicate that Mfn2 is essential for human mitochondrial development in neuronal maturation and differentiation, which will enhance our understanding of the role of Mfn2 in neurogenesis.
Collapse
Affiliation(s)
- Du Fang
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacology, University of Kansas, Lawrence, KS, USA
| | - Shijun Yan
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacology, University of Kansas, Lawrence, KS, USA
| | - Qing Yu
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacology, University of Kansas, Lawrence, KS, USA
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Cheng Du, China
| | - Doris Chen
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacology, University of Kansas, Lawrence, KS, USA
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacology, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
14
|
Zhou G, Meng S, Li Y, Ghebre YT, Cooke JP. Optimal ROS Signaling Is Critical for Nuclear Reprogramming. Cell Rep 2016; 15:919-925. [PMID: 27117405 DOI: 10.1016/j.celrep.2016.03.084] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 11/03/2015] [Accepted: 03/23/2016] [Indexed: 01/08/2023] Open
Abstract
Efficient nuclear reprogramming of somatic cells to pluripotency requires activation of innate immunity. Because innate immune activation triggers reactive oxygen species (ROS) signaling, we sought to determine whether there was a role of ROS signaling in nuclear reprogramming. We examined ROS production during the reprogramming of doxycycline (dox)-inducible mouse embryonic fibroblasts (MEFs) carrying the Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc [OSKM]) into induced pluripotent stem cells (iPSCs). ROS generation was substantially increased with the onset of reprogramming. Depletion of ROS via antioxidants or Nox inhibitors substantially decreased reprogramming efficiency. Similarly, both knockdown and knockout of p22(phox)-a critical subunit of the Nox (1-4) complex-decreased reprogramming efficiency. However, excessive ROS generation using genetic and pharmacological approaches also impaired reprogramming. Overall, our data indicate that ROS signaling is activated early with nuclear reprogramming, and optimal levels of ROS signaling are essential to induce pluripotency.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute (HMRI), 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Shu Meng
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute (HMRI), 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Yanhui Li
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute (HMRI), 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Yohannes T Ghebre
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute (HMRI), 6670 Bertner Avenue, Houston, TX 77030, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute (HMRI), 6670 Bertner Avenue, Houston, TX 77030, USA.
| |
Collapse
|
15
|
Spigoni V, Cito M, Alinovi R, Pinelli S, Passeri G, Zavaroni I, Goldoni M, Campanini M, Aliatis I, Mutti A, Bonadonna RC, Dei Cas A. Effects of TiO₂ and Co₃O₄ nanoparticles on circulating angiogenic cells. PLoS One 2015; 10:e0119310. [PMID: 25803285 PMCID: PMC4372399 DOI: 10.1371/journal.pone.0119310] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/12/2015] [Indexed: 12/03/2022] Open
Abstract
Background and Aim Sparse evidence suggests a possible link between exposure to airborne nanoparticles (NPs) and cardiovascular (CV) risk, perhaps through mechanisms involving oxidative stress and inflammation. We assessed the effects of TiO2 and Co3O4 NPs in human circulating angiogenic cells (CACs), which take part in vascular endothelium repair/replacement. Methods CACs were isolated from healthy donors’ buffy coats after culturing lymphomonocytes on fibronectin-coated dishes in endothelial medium for 7 days. CACs were pre-incubated with increasing concentration of TiO2 and Co3O4 (from 1 to 100 μg/ml) to test the effects of NP – characterized by Transmission Electron Microscopy – on CAC viability, apoptosis (caspase 3/7 activation), function (fibronectin adhesion assay), oxidative stress and inflammatory cytokine gene expression. Results Neither oxidative stress nor cell death were associated with exposure to TiO2 NP (except at the highest concentration tested), which, however, induced a higher pro-inflammatory effect compared to Co3O4 NPs (p<0.01). Exposure to Co3O4 NPs significantly reduced cell viability (p<0.01) and increased caspase activity (p<0.01), lipid peroxidation end-products (p<0.05) and pro-inflammatory cytokine gene expression (p<0.05 or lower). Notably, CAC functional activity was impaired after exposure to both TiO2 (p<0.05 or lower) and Co3O4 (p<0.01) NPs. Conclusions In vitro exposure to TiO2 and Co3O4 NPs exerts detrimental effects on CAC viability and function, possibly mediated by accelerated apoptosis, increased oxidant stress (Co3O4 NPs only) and enhancement of inflammatory pathways (both TiO2 and Co3O4 NPs). Such adverse effects may be relevant for a potential role of exposure to TiO2 and Co3O4 NPs in enhancing CV risk in humans.
Collapse
Affiliation(s)
- Valentina Spigoni
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Monia Cito
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Rossella Alinovi
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Silvana Pinelli
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Giovanni Passeri
- Department of Clinical and Experimental Medicine. Unit of Andrology, Metabolic Bone Diseases and Endocrinology. University of Parma, Parma, Italy
| | - Ivana Zavaroni
- Department of Clinical and Experimental Medicine. Unit of Diabetes and prevention of associated diseases. University of Parma, Parma, Italy
| | - Matteo Goldoni
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Marco Campanini
- IMEM-CNR (Istituto Materiale per l’Elettronica ed il Magnetismo – Consiglio Nazionale delle Ricerche) Institute, Parma, Italy
| | - Irene Aliatis
- Department of Physics and Earth Sciences. University of Parma, Parma, Italy
| | - Antonio Mutti
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Riccardo C. Bonadonna
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Alessandra Dei Cas
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
- * E-mail:
| |
Collapse
|
16
|
Srivastav S, Basu Ball W, Gupta P, Giri J, Ukil A, Das PK. Leishmania donovani prevents oxidative burst-mediated apoptosis of host macrophages through selective induction of suppressors of cytokine signaling (SOCS) proteins. J Biol Chem 2013; 289:1092-105. [PMID: 24275663 DOI: 10.1074/jbc.m113.496323] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
One of the mechanisms for establishment of infection employed by intra-macrophage pathogen-like Leishmania is inhibition of oxidative burst-mediated macrophage apoptosis to protect their niche for survival and replication. We tried to elucidate the underlying mechanism for this by using H2O2 for induction of apoptosis. Leishmania donovani-infected macrophages were much more resistant to H2O2-mediated apoptosis compared with control. Although infected cells were capable of comparable reactive oxygen species production, there was less activation of the downstream cascade consisting of caspase-3 and -7 and cleaved poly(ADP)-ribose polymerase. Suppressors of cytokine signaling (SOCS) 1 and 3 proteins and reactive oxygen species scavenging enzyme thioredoxin, known to be involved in stabilization of protein-tyrosine phosphatases, were found to be induced during infection. Induction of SOCS proteins may be mediated by Egr1, and silencing of Socs1 and -3 either alone or in combination resulted in reduced thioredoxin levels, enhanced activation of caspases, and increased apoptosis of infected macrophages. The induction of protein-tyrosine phosphatases, thioredoxin, SOCS, and Egr1 in L. donovani-infected macrophages was found to be unaffected by H2O2 treatment. SOCS knocked down cells also displayed decreased parasite survival thus marking reduction in disease progression. Taken together, these results suggest that L. donovani may exploit SOCS for subverting macrophage apoptotic machinery toward establishing its replicative niche inside the host.
Collapse
Affiliation(s)
- Supriya Srivastav
- From the Infectious Diseases and Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032 and
| | | | | | | | | | | |
Collapse
|
17
|
Sangani R, Pandya CD, Bhattacharyya MH, Periyasamy-Thandavan S, Chutkan N, Markand S, Hill WD, Hamrick M, Isales C, Fulzele S. Knockdown of SVCT2 impairs in-vitro cell attachment, migration and wound healing in bone marrow stromal cells. Stem Cell Res 2013; 12:354-63. [PMID: 24365600 DOI: 10.1016/j.scr.2013.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/01/2013] [Accepted: 11/03/2013] [Indexed: 11/28/2022] Open
Abstract
Bone marrow stromal cell (BMSC) adhesion and migration are fundamental to a number of pathophysiologic processes, including fracture and wound healing. Vitamin C is beneficial for bone formation, fracture repair and wound healing. However, the role of the vitamin C transporter in BMSC adhesion, migration and wound healing is not known. In this study, we knocked-down the sodium-dependent vitamin C transporter, SVCT2, the only known transporter of vitamin C in BMSCs, and performed cell adhesion, migration, in-vitro scratch wound healing and F-actin re-arrangement studies. We also investigated the role of oxidative stress on the above processes. Our results demonstrate that both oxidative stress and down-regulation of SVCT2 decreased cell attachment and spreading. A trans-well cell migration assay showed that vitamin C helped in BMSC migration and that knockdown of SVCT2 decreased cell migration. In the in-vitro scratch wound healing studies, we established that oxidative stress dose-dependently impairs wound healing. Furthermore, the supplementation of vitamin C significantly rescued the BMSCs from oxidative stress and increased wound closing. The knockdown of SVCT2 in BMSCs strikingly decreased wound healing, and supplementing with vitamin C failed to rescue cells efficiently. The knockdown of SVCT2 and induction of oxidative stress in cells produced an alteration in cytoskeletal dynamics. Signaling studies showed that oxidative stress phosphorylated members of the MAP kinase family (p38) and that vitamin C inhibited their phosphorylation. Taken together, these results indicate that both the SVCT2 transporter and oxidative stress play a vital role in BMSC attachment, migration and cytoskeletal re-arrangement. BMSC-based cell therapy and modulation of SVCT2 could lead to a novel therapeutic approach that enhances bone remodeling, fracture repair and wound healing in chronic disease conditions.
Collapse
Affiliation(s)
- Rajnikumar Sangani
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Chirayu D Pandya
- Department of Psychiatry and Health Behavior, Georgia Regents University, Augusta, GA 30912, USA
| | | | | | - Norman Chutkan
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Shanu Markand
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA
| | - William D Hill
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Mark Hamrick
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Carlos Isales
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|