1
|
Deng C, Aldali F, Luo H, Chen H. Regenerative rehabilitation: a novel multidisciplinary field to maximize patient outcomes. MEDICAL REVIEW (2021) 2024; 4:413-434. [PMID: 39444794 PMCID: PMC11495474 DOI: 10.1515/mr-2023-0060] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 05/15/2024] [Indexed: 10/25/2024]
Abstract
Regenerative rehabilitation is a novel and rapidly developing multidisciplinary field that converges regenerative medicine and rehabilitation science, aiming to maximize the functions of disabled patients and their independence. While regenerative medicine provides state-of-the-art technologies that shed light on difficult-to-treated diseases, regenerative rehabilitation offers rehabilitation interventions to improve the positive effects of regenerative medicine. However, regenerative scientists and rehabilitation professionals focus on their aspects without enough exposure to advances in each other's field. This disconnect has impeded the development of this field. Therefore, this review first introduces cutting-edge technologies such as stem cell technology, tissue engineering, biomaterial science, gene editing, and computer sciences that promote the progress pace of regenerative medicine, followed by a summary of preclinical studies and examples of clinical investigations that integrate rehabilitative methodologies into regenerative medicine. Then, challenges in this field are discussed, and possible solutions are provided for future directions. We aim to provide a platform for regenerative and rehabilitative professionals and clinicians in other areas to better understand the progress of regenerative rehabilitation, thus contributing to the clinical translation and management of innovative and reliable therapies.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongmei Luo
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Sun J, He L, An Q, Ye X, Ma J, Yan J, Xie X, Sun X, Niu Y, Cao W. Graphene/ chitosan tubes inoculated with dental pulp stem cells promotes repair of facial nerve injury. Front Chem 2024; 12:1417763. [PMID: 38887698 PMCID: PMC11180760 DOI: 10.3389/fchem.2024.1417763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Introduction: Facial nerve injury significantly impacts both the physical and psychological] wellbeing of patients. Despite advancements, there are still limitations associated with autografts transplantation. Consequently, there is an urgent need for effective artificial grafts to address these limitations and repair injuries. Recent years have witnessed the recognition of the beneficial effects of chitosan (CS) and graphene in the realm of nerve repair. Dental pulp stem cells (DPSCs) hold great promise due to their high proliferative and multi-directional differentiation capabilities. Methods: In this study, Graphene/CS (G/CST) composite tubes were synthesized and their physical, chemical and biological properties were evaluated, then DPSCs were employed as seed cells and G/CST as a scaffold to investigate their combined effect on promoting facial nerve injury repair. Results and Disscussion: The experimental results indicate that G/CST possesses favorable physical and chemical properties, along with good cyto-compatibility. making it suitable for repairing facial nerve transection injuries. Furthermore, the synergistic application of G/CST and DPSCs significantly enhanced the repair process for a 10 mm facial nerve defect in rabbits, highlighting the efficacy of graphene as a reinforcement material and DPSCs as a functional material in facial nerve injury repair. This approach offers an effective treatment strategy and introduces a novel concept for clinically managing facial nerve injuries.
Collapse
Affiliation(s)
- Jingxuan Sun
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Lina He
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Qi An
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xu Ye
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Jinjie Ma
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Jing Yan
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xiaoqi Xie
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xiangyu Sun
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Yumei Niu
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Wenxin Cao
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology, Harbin, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, China
| |
Collapse
|
3
|
Chen F, Che Z, Liu Y, Luo P, Xiao L, Song Y, Wang C, Dong Z, Li M, Tipoe GL, Yang M, Lv Y, Zhang H, Wang F, Xiao J. Invigorating human MSCs for transplantation therapy via Nrf2/DKK1 co-stimulation in an acute-on-chronic liver failure mouse model. Gastroenterol Rep (Oxf) 2024; 12:goae016. [PMID: 38529014 PMCID: PMC10963075 DOI: 10.1093/gastro/goae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/27/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
Background Since boosting stem cell resilience in stressful environments is critical for the therapeutic efficacy of stem cell-based transplantations in liver disease, this study aimed to establish the efficacy of a transient plasmid-based preconditioning strategy for boosting the capability of mesenchymal stromal cells (MSCs) for anti-inflammation/antioxidant defenses and paracrine actions in recipient hepatocytes. Methods Human adipose mesenchymal stem cells (hADMSCs) were subjected to transfer, either with or without the nuclear factor erythroid 2-related factor 2 (Nrf2)/Dickkopf1 (DKK1) genes, followed by exposure to TNF-α/H2O2. Mouse models were subjected to acute chronic liver failure (ACLF) and subsequently injected with either transfected or untransfected MSCs. These hADMSCs and ACLF mouse models were used to investigate the interaction between Nrf2/DKK1 and the hepatocyte receptor cytoskeleton-associated protein 4 (CKAP4). Results Activation of Nrf2 and DKK1 enhanced the anti-stress capacity of MSCs in vitro. In a murine model of ACLF, transient co-overexpression of Nrf2 and DKK1 via plasmid transfection improved MSC resilience against inflammatory and oxidative assaults, boosted MSC transplantation efficacy, and promoted recipient liver regeneration due to a shift from the activation of the anti-regenerative IFN-γ/STAT1 pathway to the pro-regenerative IL-6/STAT3 pathway in the liver. Importantly, the therapeutic benefits of MSC transplantation were nullified when the receptor CKAP4, which interacts with DKK1, was specifically removed from recipient hepatocytes. However, the removal of the another receptor low-density lipoprotein receptor-related protein 6 (LRP6) had no impact on the effectiveness of MSC transplantation. Moreover, in long-term observations, no tumorigenicity was detected in mice following transplantation of transiently preconditioned MSCs. Conclusions Co-stimulation with Nrf2/DKK1 safely improved the efficacy of human MSC-based therapies in murine models of ACLF through CKAP4-dependent paracrine mechanisms.
Collapse
Affiliation(s)
- Feng Chen
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Yingxia Liu
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - Pingping Luo
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Lu Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Yali Song
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Cunchuan Wang
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Zhiyong Dong
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Mianhuan Li
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - George L Tipoe
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Min Yang
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - Yi Lv
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, School of Life Sciences, Fujian Normal University, Fuzhou, Fujian, P. R. China
| | - Hong Zhang
- Department of Surgery, The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, Guangdong, P. R. China
| | - Fei Wang
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
- Department of Surgery, The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, Guangdong, P. R. China
| |
Collapse
|
4
|
Iqbal M, Shams S, Rafiq H, Khan M, Khan S, Sadique Khattak U, Afridi SG, Bibi F, Abdulkareem AA, Naseer MI. Combinatorial Therapeutic Potential of Stem Cells and Benzimidazol Derivatives for the Reduction of Liver Fibrosis. Pharmaceuticals (Basel) 2023; 16:306. [PMID: 37259449 PMCID: PMC9965641 DOI: 10.3390/ph16020306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 12/31/2023] Open
Abstract
(1) Background: Liver fibrosis is currently one of the top ten causes of death worldwide. Stem cells transplantation using mesenchymal stem cells (MSCs) is an alternative therapy which is used in the place of organ transplant, due to the incapacity of stem cells to endure oxidative stress in the damage site, thus affecting the healing process. The present study aimed to enhance the therapeutic potential of MSCs using combined therapy, along with the novel synthetic compounds of benzimidazol derivatives. (2) Methods: Eighteen compound series (benzimidazol derivatives) were screened against liver fibrosis using an in vitro CCl4-induced injury model on cultured hepatocytes. IC50 values were calculated on the bases of LDH assay and cell viability assay. (3) Results: Among the eighteen compounds, compounds (10), (14) and (18) were selected on the basis of IC50 value, and compound (10) was the most potent and had the lowest IC50 value in the LDH assay (8.399 ± 0.23 uM) and cell viability assay (4.73 ± 0.37 uM). Next, these compounds were combined with MSCs using an in vitro hepatocytes injury culture and in vivo rat fibrotic model. The effect of the MSCs + compounds treatment on injured hepatocytes was evaluated using LDH assay, cell viability assay, GSH assay and real-time PCR analysis and immuno-staining for caspase-3. Significant reductions in LDH level, caspase-3 and apoptotic marker genes were noted in MSCs + compounds-treated injured hepatocytes. In vivo data also showed the increased homing of the MSCs, along with compounds after transplantation. Real-time PCR analysis and TUNEL assay results also support our study. (4) Conclusions: It was concluded that compounds (10), (14) and (18) can be used in combination with MSCs to reduce liver fibrosis.
Collapse
Affiliation(s)
- Maryam Iqbal
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Sulaiman Shams
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Huma Rafiq
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Shahid Khan
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Umer Sadique Khattak
- College of Veterinary Sciences, The University of Agriculture, Peshawar 25130, Khyber Pakhtunkhwa, Pakistan
| | - Sahib Gul Afridi
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Fehmida Bibi
- Special Infectious Agents Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Angham Abdulrhman Abdulkareem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Imran Naseer
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
5
|
Pu L, Luo Y, Wen Z, Dai Y, Zheng C, Zhu X, Qin L, Zhang C, Liang H, Zhang J, Guo L, Wang L. GPX2 Gene Affects Feed Efficiency of Pigs by Inhibiting Fat Deposition and Promoting Muscle Development. Animals (Basel) 2022; 12:ani12243528. [PMID: 36552449 PMCID: PMC9774625 DOI: 10.3390/ani12243528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
GPX2 has been recognized as a potential candidate gene for feed efficiency in pigs. This article aimed to elucidate polymorphism of GPX2 associated with feed efficiency and its related molecular mechanism. In this study, seven single nucleotide polymorphisms (SNP) of GPX2 were found among 383 Duroc pigs. In addition, seven SNPs and ALGA0043483 (PorcineSNP60 BeadChip data in 600 Duroc pigs), which are near the GPX2 gene, were identified in one haplotypes block. Furthermore, associated studies showed that the genotype of GPX2 has significant association with weaning weight and 100 kg BF in Duroc pigs. In addition, the AG had no effect when the backfat became thinner, and the FCR and RFI traits had a tendency to decrease in the G3 + TT combination genotype, accompanied by an increase of GPX2 expression in backfat and muscle tissues. At the cellular level, the adipocyte proliferation and ability of adipogenic differentiation were reduced, and the lipid degradation increased in 3T3-L1 when there was overexpression of GPX2. In contrast, overexpression of the GPX2 gene can promote the muscle cell proliferation and myogenic differentiation in C2C12 cells. In other words, GPX2 has the effect of reducing fat deposition and promoting muscle development, and it is a candidate gene for backfat and feed efficiency.
Collapse
Affiliation(s)
- Lei Pu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: (L.P.); (L.W.)
| | - Yunyan Luo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Zuochen Wen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Yuxin Dai
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Chunting Zheng
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Xueli Zhu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Lei Qin
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Chunguang Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Hong Liang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Jianbin Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Liang Guo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Lixian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: (L.P.); (L.W.)
| |
Collapse
|
6
|
Administration of Melatonin in Diabetic Retinopathy Is Effective and Improves the Efficacy of Mesenchymal Stem Cell Treatment. Stem Cells Int 2022; 2022:6342594. [PMID: 35450343 PMCID: PMC9017455 DOI: 10.1155/2022/6342594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 03/26/2022] [Indexed: 11/29/2022] Open
Abstract
Stem cell transplantation is a promising therapeutic technique for the treatment of a variety of diseases; nevertheless, stem cell therapy may not always work as well as it could. The goal of this study was to test the hypothesis that employing a powerful antioxidant like melatonin improves stem cell transplantation success and potentiates stem cell function in the therapy of diabetic retinopathy. For this purpose, 50 adult male rats were divided into the following: control group: this group received 0.5 ml of 0.1 M of sodium citrate buffer (pH = 4.5) (intraperitoneal (I.P.)). The confirmed diabetic rats were divided into 4 groups: diabetic group: confirmed diabetic rats received no treatments with a regular follow of the blood glucose profile for 8 weeks; melatonin group: confirmed diabetic rats received melatonin (5 mg/kg/day); stem cell group: the confirmed diabetic rats were given intravitreal injection of stem cells (2 μl cell suspension of stem cells (3 × 104 cells/μl)); and melatonin+stem cell group: confirmed diabetic rats received melatonin (5 mg/kg/day), orally once daily for 8 weeks, and 2 μl cell suspension of stem cells (3 × 104 cells/μl) was carefully injected into the vitreous cavity. Our results showed that administration of melatonin and/or stem cell restored the retinal oxidative/antioxidant redox and reduced retinal inflammatory mediators. Coadministration of melatonin and stem cells enhanced the number of transplanted stem cells in the retinal tissue and significantly reduced retinal BDEF, VEGF, APOA1, and RBP4 levels as compared to melatonin and/or stem alone. We may conclude that rats treated with melatonin and stem cells had their retinal oxidative/antioxidant redox values restored to normal and their histological abnormalities reduced. These findings support the hypothesis that interactions with the BDEF, VEGF, APOA1, and RBP4 signaling pathways are responsible for these effects.
Collapse
|
7
|
Hu XM, Zhang Q, Zhou RX, Wu YL, Li ZX, Zhang DY, Yang YC, Yang RH, Hu YJ, Xiong K. Programmed cell death in stem cell-based therapy: Mechanisms and clinical applications. World J Stem Cells 2021; 13:386-415. [PMID: 34136072 PMCID: PMC8176847 DOI: 10.4252/wjsc.v13.i5.386] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based therapy raises hopes for a better approach to promoting tissue repair and functional recovery. However, transplanted stem cells show a high death percentage, creating challenges to successful transplantation and prognosis. Thus, it is necessary to investigate the mechanisms underlying stem cell death, such as apoptotic cascade activation, excessive autophagy, inflammatory response, reactive oxygen species, excitotoxicity, and ischemia/hypoxia. Targeting the molecular pathways involved may be an efficient strategy to enhance stem cell viability and maximize transplantation success. Notably, a more complex network of cell death receives more attention than one crucial pathway in determining stem cell fate, highlighting the challenges in exploring mechanisms and therapeutic targets. In this review, we focus on programmed cell death in transplanted stem cells. We also discuss some promising strategies and challenges in promoting survival for further study.
Collapse
Affiliation(s)
- Xi-Min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rui-Xin Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yan-Lin Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Zhi-Xin Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Dan-Yi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yi-Chao Yang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rong-Hua Yang
- Department of Burns, Fo Shan Hospital of Sun Yat-Sen University, Foshan 528000, Guangdong Province, China
| | - Yong-Jun Hu
- Department of Cardiovascular Medicine, Hunan People's Hospital (the First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
8
|
Heterospheroid formation improves therapeutic efficacy of mesenchymal stem cells in murine colitis through immunomodulation and epithelial regeneration. Biomaterials 2021; 271:120752. [PMID: 33730631 DOI: 10.1016/j.biomaterials.2021.120752] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022]
Abstract
Tissue repairing capacity and immunomodulatory effects of mesenchymal stem cells (MSCs) have been extensively utilized for treating various inflammatory disorders; however, inconsistent efficacy and therapeutic outcomes due to low survival rate after transplantation often restrain their clinical potential. To overcome these limitations, 3-dimensional culture (3D-culture) was established to augment stemness and paracrine functions of MSCs, although hypoxic stress at the core often leads to unexpected cell death. Thus, we designed a novel strategy to improve the microenvironment of MSCs by creating heterospheroids (HS) consisting of MSCs and quercetin (QUR)-loaded microspheres (MSCHS), to achieve local drug delivery to the cells. Notably, MSCHS exhibited resistance for senescence-associated phenotype and oxidative stress-induced apoptosis compared to 3D-cultured MSCs (MSC3D), as well as to 2D-cultured cells (MSC2D) in vitro. In a murine model of colitis, MSC3D and MSCHS exhibited enhanced anti-inflammatory impact than MSC2Dvia attenuating neutrophil infiltration and regulating helper T cell (Th) polarization into Th1 and Th17 cells. Interestingly, MSCHS provided better therapeutic outcomes compared to MSC3D, partially due to their enhanced survival capacity in vivo. Moreover, we found that MSC-derived paracrine factor, prostaglandin E2 (PGE2), can directly drive the epithelial regeneration process by inducing specialized tissue-repairing cell generation using the intestinal organoid culture. Importantly, MSC3D and MSCHS displayed an outstanding regeneration-inducing potency compared to MSC2D owing to their superior PGE2 secretion. Taken together, we suggest a convergent strategy of MSCHS formation with reactive oxygen species (ROS) scavenger, QUR, which can maximize the inflammation-attenuating and tissue-repairing capacity of MSCs, as well as the engraftment efficiency after transplantation.
Collapse
|
9
|
Identification of a Sesquiterpene Lactone from Arctium lappa Leaves with Antioxidant Activity in Primary Human Muscle Cells. Molecules 2021; 26:molecules26051328. [PMID: 33801315 PMCID: PMC7958318 DOI: 10.3390/molecules26051328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
Many pathologies affecting muscles (muscular dystrophies, sarcopenia, cachexia, renal insufficiency, obesity, diabetes type 2, etc.) are now clearly linked to mechanisms involving oxidative stress. In this context, there is a growing interest in exploring plants to find new natural antioxidants to prevent the appearance and the development of these muscle disorders. In this study, we investigated the antioxidant properties of Arctium lappa leaves in a model of primary human muscle cells exposed to H2O2 oxidative stress. We identified using bioassay-guided purification, onopordopicrin, a sesquiterpene lactone as the main molecule responsible for the antioxidant activity of A. lappa leaf extract. According to our findings, onopordopicrin inhibited the H2O2-mediated loss of muscle cell viability, by limiting the production of free radicals and abolishing DNA cellular damages. Moreover, we showed that onopordopicrin promoted the expression of the nuclear factor-erythroid-2-related factor 2 (Nrf2) downstream target protein heme oxygenase-1 (HO-1) in muscle cells. By using siRNA, we demonstrated that the inhibition of the expression of Nrf2 reduced the protective effect of onopordopicrin, indicating that the activation of the Nrf2/HO-1 signaling pathway mediates the antioxidant effect of onopordopicrin in primary human muscle cells. Therefore, our results suggest that onopordopicrin may be a potential therapeutic molecule to fight against oxidative stress in pathological specific muscle disorders.
Collapse
|
10
|
Awad K, Ahuja N, Fiedler M, Peper S, Wang Z, Aswath P, Brotto M, Varanasi V. Ionic Silicon Protects Oxidative Damage and Promotes Skeletal Muscle Cell Regeneration. Int J Mol Sci 2021; 22:E497. [PMID: 33419056 PMCID: PMC7825403 DOI: 10.3390/ijms22020497] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
Volumetric muscle loss injuries overwhelm the endogenous regenerative capacity of skeletal muscle, and the associated oxidative damage can delay regeneration and prolong recovery. This study aimed to investigate the effect of silicon-ions on C2C12 skeletal muscle cells under normal and excessive oxidative stress conditions to gain insights into its role on myogenesis during the early stages of muscle regeneration. In vitro studies indicated that 0.1 mM Si-ions into cell culture media significantly increased cell viability, proliferation, migration, and myotube formation compared to control. Additionally, MyoG, MyoD, Neurturin, and GABA expression were significantly increased with addition of 0.1, 0.5, and 1.0 mM of Si-ion for 1 and 5 days of C2C12 myoblast differentiation. Furthermore, 0.1-2.0 mM Si-ions attenuated the toxic effects of H2O2 within 24 h resulting in increased cell viability and differentiation. Addition of 1.0 mM of Si-ions significantly aid cell recovery and protected from the toxic effect of 0.4 mM H2O2 on cell migration. These results suggest that ionic silicon may have a potential effect in unfavorable situations where reactive oxygen species is predominant affecting cell viability, proliferation, migration, and differentiation. Furthermore, this study provides a guide for designing Si-containing biomaterials with desirable Si-ion release for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kamal Awad
- Department of Materials Science and Engineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (K.A.); (P.A.)
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Neelam Ahuja
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Matthew Fiedler
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Sara Peper
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
- Department of Bioengineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Zhiying Wang
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Pranesh Aswath
- Department of Materials Science and Engineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (K.A.); (P.A.)
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Venu Varanasi
- Department of Materials Science and Engineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (K.A.); (P.A.)
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| |
Collapse
|
11
|
Yosef B, Zhou Y, Mouschouris K, Poteracki J, Soker S, Criswell T. N-Acetyl-L-Cysteine Reduces Fibrosis and Improves Muscle Function After Acute Compartment Syndrome Injury. Mil Med 2020; 185:25-34. [PMID: 32074330 DOI: 10.1093/milmed/usz232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Upon injury, skeletal muscle undergoes a multiphase process beginning with degeneration of the damaged tissue, which is accompanied by inflammation and finally regeneration. One consequence of an injured microenvironment is excessive production of reactive oxygen species, which results in attenuated regeneration and recovery of function ultimately leading to fibrosis and disability. The objective of this research was to test the potential of the antioxidant, N-Acetyl-L-Cysteine (NAC), as a mediator of reactive oxygen species damage that results from traumatic muscle injury in order to support repair and regeneration of wounded muscle tissue and improve function recovery. MATERIALS AND METHODS Adult female Lewis rats were subjected to compartment syndrome injury as previously published by our group. Rats received intramuscular injections of NAC or vehicle at 24, 48, and 72 hours postinjury. Muscle function, tissue fibrosis, and the expression of myogenic and angiogenic markers were measured. RESULTS Muscle function was significantly improved, and tissue fibrosis was significantly decreased in NAC-treated muscles. CONCLUSIONS These results suggest that NAC treatment of skeletal muscle after injury may be a viable option for the prevention of long-term fibrosis and scar formation, facilitating recovery of muscle function.
Collapse
Affiliation(s)
- Benyam Yosef
- Department of Cardiac Surgery, Brigham and Young Women's Hospital, 75 Francis St., Boston, MA 02115
| | - Yu Zhou
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Medical Center Blvd, Winston-Salem, NC 27157
| | - Kathryn Mouschouris
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Medical Center Blvd, Winston-Salem, NC 27157
| | - James Poteracki
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Medical Center Blvd, Winston-Salem, NC 27157
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Medical Center Blvd, Winston-Salem, NC 27157
| | - Tracy Criswell
- Department of Cardiac Surgery, Brigham and Young Women's Hospital, 75 Francis St., Boston, MA 02115.,Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Health, Medical Center Blvd, Winston-Salem, NC 27157
| |
Collapse
|
12
|
Islam D, Huang Y, Fanelli V, Delsedime L, Wu S, Khang J, Han B, Grassi A, Li M, Xu Y, Luo A, Wu J, Liu X, McKillop M, Medin J, Qiu H, Zhong N, Liu M, Laffey J, Li Y, Zhang H. Identification and Modulation of Microenvironment Is Crucial for Effective Mesenchymal Stromal Cell Therapy in Acute Lung Injury. Am J Respir Crit Care Med 2020; 199:1214-1224. [PMID: 30521764 DOI: 10.1164/rccm.201802-0356oc] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rationale: There are controversial reports on applications of mesenchymal stromal cells (MSCs) in patients with acute respiratory distress syndrome (ARDS). Objectives: We hypothesized that lung microenvironment was the main determinant of beneficial versus detrimental effects of MSCs during ARDS. Methods: Lung proteome was profiled in three models of injury induced by acid instillation and/or mechanical ventilation in mice. Human gene of glutathione peroxidase-1 was delivered before MSC administration; or MSCs carrying human gene of IL-10 or hepatocyte growth factor were administered after lung injury. An inhibitory cocktail against IL-6, fibronectin, and oxidative stress was used in in vitro studies using human small airway epithelial cells and human MSCs after exposure to plasma of patients with ARDS. Measurements and Main Results: Distinct proteomic profiles were observed in three lung injury models. Administration of MSCs protected lung from ventilator-induced injury, whereas it worsened acid-primed lung injuries associated with fibrotic development in lung environment that had high levels of IL-6 and fibronectin along with low antioxidant capacity. Correction of microenvironment with glutathione peroxidase-1, or treatment with MSCs carrying human gene of IL-10 or hepatocyte growth factor after acid-primed injury, reversed the detrimental effects of native MSCs. Proteomic profiles obtained in the mouse models were also similarly observed in human ARDS. Treatment with the inhibitory cocktail in samples of patients with ARDS retained protective effects of MSCs in small airway epithelial cells. Conclusions: MSCs can be beneficial or detrimental depending on microenvironment at the time of administration. Identification of potential beneficiaries seems to be crucial to guide MSC therapy in ARDS.
Collapse
Affiliation(s)
- Diana Islam
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yongbo Huang
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Vito Fanelli
- 2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Anesthesia and Critical Care and
| | - Luisa Delsedime
- 4 Department of Pathology, University of Turin, Turin, Italy
| | - Sulong Wu
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Julie Khang
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Bing Han
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Alice Grassi
- 2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Manshu Li
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yonghao Xu
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Alice Luo
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jianfeng Wu
- 2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Xiaoqing Liu
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Montey McKillop
- 5 Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeffery Medin
- 5 Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Haibo Qiu
- 6 Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Nanshan Zhong
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mingyao Liu
- 7 Department of Surgery, University Health Network, Toronto, Ontario, Canada.,8 Department of Medicine.,9 Department of Physiology
| | - John Laffey
- 10 Department of Anesthesia and Intensive Care Medicine, National University of Ireland, Galway, Ireland
| | - Yimin Li
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Haibo Zhang
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada.,8 Department of Medicine.,9 Department of Physiology.,11 Interdepartmental Division of Critical Care Medicine, and.,12 Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
13
|
Maharajan N, Vijayakumar K, Jang CH, Cho GW. Caloric restriction maintains stem cells through niche and regulates stem cell aging. J Mol Med (Berl) 2019; 98:25-37. [DOI: 10.1007/s00109-019-01846-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
|
14
|
Regmi S, Pathak S, Thanh TP, Nguyen TT, Sung JH, Yook S, Kim JO, Yong CS, Choi I, Doh KO, Park PH, Park JB, Seo Y, Kim BK, Lee DM, Moon IJ, Kim HS, Jeong JH. Intraportally delivered stem cell spheroids localize in the liver and protect hepatocytes against GalN/LPS-induced fulminant hepatic toxicity. Stem Cell Res Ther 2019; 10:230. [PMID: 31615539 PMCID: PMC6794806 DOI: 10.1186/s13287-019-1337-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/29/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Systemic inflammatory response syndrome (SIRS) is common in severe fulminant hepatic failure (FHF) and has a high mortality rate (20–50%) due to irreversible cerebral edema or sepsis. Stem cell-based treatment has emerged as a promising alternative therapeutic strategy to prolong the survival of patients suffering from FHF via the inhibition of SIRS due to their immunomodulatory effects. Methods 3D spheroids of adipose-derived mesenchymal stem cells (3D-ADSC) were prepared by the hanging drop method. The efficacy of the 3D-ADSC to rescue FHF was evaluated in a d-galactosamine/lipopolysaccharide (GalN/LPS)-induced mouse model of FHF via intraportal transplantation of the spheroids. Results Intraportally delivered 3D-ADSC better engrafted and localized into the damaged livers compared to 2D-cultured adipose-derived mesenchymal stem cells (2D-ADSC). Transplantation of 3D-ADSC rescued 50% of mice from FHF-induced lethality, whereas only 20% of mice survived when 2D-ADSC were transplanted. The improved transplantation outcomes correlated with the enhanced immunomodulatory effect of 3D-ADSC in the liver microenvironment. Conclusion The study shows that the transplantation of optimized 3D-ADSC can efficiently ameliorate GalN/LPS-induced FHF due to improved viability, resistance to exogenous ROS, and enhanced immunomodulatory effects of 3D-ADSC. Electronic supplementary material The online version of this article (10.1186/s13287-019-1337-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shobha Regmi
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Shiva Pathak
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Tung Pham Thanh
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Tiep Tien Nguyen
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, 42415, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Kyoung-Oh Doh
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, 42415, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yoojin Seo
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea.,Institute for Translational Dental Sciences, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Bieong-Kil Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, 42415, Republic of Korea
| | - Dong-Mok Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Gyeongbuk, 38822, Republic of Korea
| | - Ik-Jae Moon
- WELGENE Inc., Gyeongsan, 38695, Republic of Korea
| | - Hyung-Sik Kim
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea. .,Institute for Translational Dental Sciences, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan-si, Gyeongbuk-do, 38541, Republic of Korea.
| |
Collapse
|
15
|
Huard J. Stem cells, blood vessels, and angiogenesis as major determinants for musculoskeletal tissue repair. J Orthop Res 2019; 37:1212-1220. [PMID: 29786150 DOI: 10.1002/jor.24058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/21/2018] [Indexed: 02/04/2023]
Abstract
This manuscript summarizes 20 years of research from my laboratories at the University of Pittsburgh and more recently, at the University of Texas Health Science Center at Houston and the Steadman Philippon Research Institute in Vail, Colorado. The discovery of muscle-derived stem cells (MDSCs) did not arise from a deliberate search to find a novel population of muscle cells with high regenerative potential, but instead was conceived in response to setbacks encountered while working in muscle cell transplantation for Duchenne muscular dystrophy (DMD). DMD is a devastating inherited X-linked muscle disease characterized by progressive muscle weakness due to lack of dystrophin expression in muscle fiber sarcolemma.1 Although the transplantation of normal myoblasts into dystrophin-deficient muscle can restore dystrophin, this approach has been hindered by limited survival (less than 1%) of the injected cells.1 The fact that 99% of the cells were not surviving implantation was seen as a major weakness with this technology by most. My research team decided to investigate which cells represent the 1% of the cells surviving post-implantation. We have subsequently confirmed that the few cells which exhibit high survival post-implantation also display stem cell characteristics, and were termed "muscle-derived stem cells" or MDSCs. Herein, I will describe the origin of these MDSCs, the mechanisms of MDSC action during tissue repair, and finally the development of therapeutic strategies to improve regeneration and repair of musculoskeletal tissues. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1212-1220, 2019.
Collapse
Affiliation(s)
- Johnny Huard
- Department of Orthopaedic Surgery, and The Brown Foundation Institute of Molecular Medicine Center for Tissue Engineering and Aging Research, McGovern Medical School, The University of Texas Health Science Center, 1881 East Road, 3SCR6.3618, Houston, Texas, 77054.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, 181 W. Meadow Drive, Suite 1000, Vail, Colorado, 81657
| |
Collapse
|
16
|
Schreurs M, Suttorp CM, Mutsaers HAM, Kuijpers-Jagtman AM, Von den Hoff JW, Ongkosuwito EM, Carvajal Monroy PL, Wagener FADTG. Tissue engineering strategies combining molecular targets against inflammation and fibrosis, and umbilical cord blood stem cells to improve hampered muscle and skin regeneration following cleft repair. Med Res Rev 2019; 40:9-26. [PMID: 31104334 PMCID: PMC6972684 DOI: 10.1002/med.21594] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 12/18/2022]
Abstract
Cleft lip with or without cleft palate is a congenital deformity that occurs in about 1 of 700 newborns, affecting the dentition, bone, skin, muscles and mucosa in the orofacial region. A cleft can give rise to problems with maxillofacial growth, dental development, speech, and eating, and can also cause hearing impairment. Surgical repair of the lip may lead to impaired regeneration of muscle and skin, fibrosis, and scar formation. This may result in hampered facial growth and dental development affecting oral function and lip and nose esthetics. Therefore, secondary surgery to correct the scar is often indicated. We will discuss the molecular and cellular pathways involved in facial and lip myogenesis, muscle anatomy in the normal and cleft lip, and complications following surgery. The aim of this review is to outline a novel molecular and cellular strategy to improve musculature and skin regeneration and to reduce scar formation following cleft repair. Orofacial clefting can be diagnosed in the fetus through prenatal ultrasound screening and allows planning for the harvesting of umbilical cord blood stem cells upon birth. Tissue engineering techniques using these cord blood stem cells and molecular targeting of inflammation and fibrosis during surgery may promote tissue regeneration. We expect that this novel strategy improves both muscle and skin regeneration, resulting in better function and esthetics after cleft repair.
Collapse
Affiliation(s)
- Michaël Schreurs
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - C Maarten Suttorp
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | | | - Johannes W Von den Hoff
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Edwin M Ongkosuwito
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Paola L Carvajal Monroy
- Department of Oral & Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Frank A D T G Wagener
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Regmi S, Pathak S, Kim JO, Yong CS, Jeong JH. Mesenchymal stem cell therapy for the treatment of inflammatory diseases: Challenges, opportunities, and future perspectives. Eur J Cell Biol 2019; 98:151041. [PMID: 31023504 DOI: 10.1016/j.ejcb.2019.04.002] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising alternative agents for the treatment of inflammatory disorders due to their immunomodulatory functions, and several clinical trials on MSC-based products are currently being conducted. In this review, we discuss recent progress made on the use of MSCs as immunomodulatory agents, developmental challenges posed by MSC-based therapy, and the strategies being used to overcome these challenges. In this context, current understanding of the mechanisms responsible for MSC interactions with the immune system and the molecular responses of MSCs to inflammatory signals are discussed. The immunosuppressive activities of MSCs are initiated by cell-to-cell contact and the release of immuno-regulatory molecules. By doing so, MSCs can inhibit the proliferation and function of T cells, natural killer cells, B cells, and dendritic cells, and can also increase the proliferation of regulatory T cells. However, various problems, such as low transplanted cell viability, poor homing and engraftment into injured tissues, MSC heterogeneity, and lack of adequate information on optimum MSC doses impede clinical applications. On the other hand, it has been shown that the immunomodulatory activities and viabilities of MSCs might be enhanced by 3D-cultured systems, genetic modifications, preconditioning, and targeted-delivery.
Collapse
Affiliation(s)
- Shobha Regmi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Shiva Pathak
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
18
|
Karabulutoglu M, Finnon R, Imaoka T, Friedl AA, Badie C. Influence of diet and metabolism on hematopoietic stem cells and leukemia development following ionizing radiation exposure. Int J Radiat Biol 2018; 95:452-479. [PMID: 29932783 DOI: 10.1080/09553002.2018.1490042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The review aims to discuss the prominence of dietary and metabolic regulators in maintaining hematopoietic stem cell (HSC) function, long-term self-renewal, and differentiation. RESULTS Most adult stem cells are preserved in a quiescent, nonmotile state in vivo which acts as a "protective state" for stem cells to reduce endogenous stress provoked by DNA replication and cellular respiration as well as exogenous environmental stress. The dynamic balance between quiescence, self-renewal and differentiation is critical for supporting a functional blood system throughout life of an organism. Stress-conditions, for example ionizing radiation exposure can trigger the blood forming HSCs to proliferate and migrate through extramedullary tissues to expand the number of HSCs and increase hematopoiesis. In addition, a wealth of investigation validated that deregulation of this balance plays a critical pathogenic role in various different hematopoietic diseases including the leukemia development. CONCLUSION The review summarizes the current knowledge on how alterations in dietary and metabolic factors could alter the risk of leukemia development following ionizing radiation exposure by inhibiting or even reversing the leukemic progression. Understanding the influence of diet, metabolism, and epigenetics on radiation-induced leukemogenesis may lead to the development of practical interventions to reduce the risk in exposed populations.
Collapse
Affiliation(s)
- Melis Karabulutoglu
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK.,b CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Rosemary Finnon
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| | - Tatsuhiko Imaoka
- c Department of Radiation Effects Research, National Institute of Radiological Sciences , National Institutes for Quantum and Radiological Science and Technology , Chiba , Japan
| | - Anna A Friedl
- d Department of Radiation Oncology , University Hospital, LMU Munich , Munich , Germany
| | - Christophe Badie
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| |
Collapse
|
19
|
Abstract
Stem cell aging is a process in which stem cells progressively lose their ability to self-renew or differentiate, succumb to senescence or apoptosis, and eventually become functionally depleted. Unresolved oxidative stress and concomitant oxidative damages of cellular macromolecules including nucleic acids, proteins, lipids, and carbohydrates have been recognized to contribute to stem cell aging. Excessive production of reactive oxygen species and insufficient cellular antioxidant reserves compromise cell repair and metabolic homeostasis, which serves as a mechanistic switch for a variety of aging-related pathways. Understanding the molecular trigger, regulation, and outcomes of those signaling networks is critical for developing novel therapies for aging-related diseases by targeting stem cell aging. Here we explore the key features of stem cell aging biology, with an emphasis on the roles of oxidative stress in the aging process at the molecular level. As a concept of cytoprotection of stem cells in transplantation, we also discuss how systematic enhancement of endogenous antioxidant capacity before or during graft into tissues can potentially raise the efficacy of clinical therapy. Finally, future directions for elucidating the control of oxidative stress and developing preventive/curative strategies against stem cell aging are discussed.
Collapse
Affiliation(s)
- Feng Chen
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Yingxia Liu
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Nai-Kei Wong
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Jia Xiao
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.,2 Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- 3 GMH Institute of CNS Regeneration, Guangdong Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Li M, Lv Y, Chen F, Wang X, Zhu J, Li H, Xiao J. Co-stimulation of LPAR 1 and S1PR 1/3 increases the transplantation efficacy of human mesenchymal stem cells in drug-induced and alcoholic liver diseases. Stem Cell Res Ther 2018; 9:161. [PMID: 29898789 PMCID: PMC6000942 DOI: 10.1186/s13287-018-0860-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/13/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
Background One of the major obstacles facing stem cell therapy is the limited number of functional stem cells available after transplantation due to the harsh microenvironment surrounding the damaged tissue. The aim of this study was to delineate the mechanistic involvement of lysophosphatidic acid receptors (LPARs) and sphingosine-1-phosphate receptors (S1PRs) in the regulation of anti-stress and transplantation efficacy of stem cells. Methods Human adipose-derived mesenchymal stem cells (hADMSCs) were treated with chemical toxin or ethanol to induce cell stress. Lysophosphatidic acid (LPA) and/or sphingosine-1-phosphate (S1P) were co-treated to examine their protective effects and mechanisms on stem cell damage. Acute liver failure and alcoholic liver disease murine models were also established to test the transplantation efficacy of hADMSCs with or without LPA/S1P pre-incubation. Results Co-stimulation of LPAR1 by LPA and S1PR1/3 by S1P synergistically enhanced the anti-stress ability of hADMSCs induced by chemical or ethanol incubation in vitro. Downstream pathways involved in this process included the Gi protein (but not the G12/13 proteins), the RAS/ERK pathway, and the PI3K/Akt pathway. Upon cell injury, the nuclear translocation of nuclear factor-kappa B (NF-κB) was promoted to facilitate the activation of downstream pro-inflammatory gene transcription, which was ameliorated by co-treatment with LPA and/or S1P. Increased secretion of interleukin (IL)-10 from stem cells by LPA and/or S1P seemed to be one of the major protective mechanisms since blocking IL-10 expression significantly aggravated stress-induced cell damage. In a drug-induced acute liver failure model and a chronic alcoholic liver disease model, pre-conditioning with LPA and/or S1P significantly enhanced the survival ratio and the therapeutic efficacy of hADMSCs in mice, including ameliorating histological damage, oxidative stress, inflammation, fibrosis, lipid metabolism dysfunction, and enhancing alcohol metabolizing enzyme activity. Importantly, supplementing LPA and/or S1P did not alter the basic characteristics of the hADMSCs nor induce tumour formation after cell transplantation. Conclusions Co-use of LPA and S1P represents a novel and safe strategy to enhance stem cell transplantation efficacy for future drug- and alcoholic-related liver disease therapies. Electronic supplementary material The online version of this article (10.1186/s13287-018-0860-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mianhuan Li
- Department of Gastroenterology, Clinical Medicine Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China.,State Key Discipline of Infectious Diseases, Department of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, People's Republic of China
| | - Yi Lv
- Department of Gastroenterology, Clinical Medicine Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China
| | - Feng Chen
- State Key Discipline of Infectious Diseases, Department of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, People's Republic of China
| | - Xiaoyan Wang
- Department of Gastroenterology, Clinical Medicine Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China
| | - Jiang Zhu
- JM Medical (Shenzhen), LLC, Shenzhen, Shenzhen, 518000, People's Republic of China
| | - Hao Li
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Centre, Guangzhou, 510060, People's Republic of China.
| | - Jia Xiao
- Department of Gastroenterology, Clinical Medicine Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China. .,State Key Discipline of Infectious Diseases, Department of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, People's Republic of China. .,School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong, Special Administrative Region of China.
| |
Collapse
|
21
|
Guo J, Zhou H, Wang J, Liu W, Cheng M, Peng X, Qin H, Wei J, Jin P, Li J, Zhang X. Nano vanadium dioxide films deposited on biomedical titanium: a novel approach for simultaneously enhanced osteogenic and antibacterial effects. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:58-74. [PMID: 29560740 DOI: 10.1080/21691401.2018.1452020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Vanadium is a trace element in the human body, and vanadium compounds have a promising future in biological and medical applications due to their various biological activities and low toxicity. Herein, a novel pure vanadium dioxide (VO2) nanofilm was deposited on a substrate of biomedical titanium by magnetron sputtering. The antibacterial effect of VO2 against the methicillin-resistant Staphylococcus aureus (MRSA) was validated in vitro and in vivo. Moreover, the biocompatibility of VO2 and its osteogenic effects were systematically illustrated. A possible osteogenic mechanism involving the amelioration of highly reactive oxygen species (ROS) levels were investigated. According to the results of our present and previous studies, the simultaneous antibacterial and osteogenic effects of VO2 are attributed to its differential regulation of ROS levels in rat bone marrow mesenchymal stem cells (rBMSCs) and bacteria. This study is the first to report the simultaneous effects of VO2 on bactericidal and osteogenic activities through its differential modification of ROS activity in eukaryotic (rBMSCs) and prokaryotic (MRSA) cells. The findings in this work may yield a deeper understanding of the biological activities of vanadium compounds while also paving the way for the further investigation and application of VO2 in biological and medical materials.
Collapse
Affiliation(s)
- Jinxiao Guo
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Huaijuan Zhou
- b State Key Laboratory of High Performance Ceramics and Superfine Microstructure , Shanghai Institute of Ceramics, Chinese Academy of Sciences , Shanghai , China
| | - Jiaxing Wang
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Wei Liu
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Mengqi Cheng
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Xiaochun Peng
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Hui Qin
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Jianfeng Wei
- c Department of Histology and Embryology, School of Basic Medical Sciences , Xuzhou Medical University , Xuzhou , China
| | - Ping Jin
- b State Key Laboratory of High Performance Ceramics and Superfine Microstructure , Shanghai Institute of Ceramics, Chinese Academy of Sciences , Shanghai , China
| | - Jinhua Li
- d Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pok Fu Lam , Hong Kong, China
| | - Xianlong Zhang
- a Department of Orthopaedics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
22
|
Abstract
Stem cell therapy is a promising approach to the treatment of ischemic heart disease via replenishing cell loss after myocardial infarction. Both preclinical studies and clinical trials have indicated that cardiac function improved consistently, but very modestly after cell-based therapy. This mainly attributed to low cell survival rate, engraftment and functional integration, which became the major challenges to regenerative medicine. In recent years, several new cell types have been developed to regenerate cardiomyocytes and novel delivery approaches helped to increase local cell retention. New strategies, such as cell pretreatment, gene-based therapy, tissue engineering, extracellular vesicles application and immunologic regulation, have surged and brought about improved cell survival and functional integration leading to better therapeutic effects after cell transplantation. In this review, we summarize these new strategies targeting at challenges of cardiac regenerative medicine and discuss recent evidences that may hint their effectiveness in the future clinical settings.
Collapse
|
23
|
Echeverri-Ruiz N, Haynes T, Landers J, Woods J, Gemma MJ, Hughes M, Del Rio-Tsonis K. A biochemical basis for induction of retina regeneration by antioxidants. Dev Biol 2017; 433:394-403. [PMID: 29291983 PMCID: PMC5753421 DOI: 10.1016/j.ydbio.2017.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 12/19/2022]
Abstract
The use of antioxidants in tissue regeneration has been studied, but their mechanism of action is not well understood. Here, we analyze the role of the antioxidant N-acetylcysteine (NAC) in retina regeneration. Embryonic chicks are able to regenerate their retina after its complete removal from retinal stem/progenitor cells present in the ciliary margin (CM) of the eye only if a source of exogenous factors, such as FGF2, is present. This study shows that NAC modifies the redox status of the CM, initiates self-renewal of the stem/progenitor cells, and induces regeneration in the absence of FGF2. NAC works as an antioxidant by scavenging free radicals either independently or through the synthesis of glutathione (GSH), and/or by reducing oxidized proteins through a thiol disulfide exchange activity. We dissected the mechanism used by NAC to induce regeneration through the use of inhibitors of GSH synthesis and the use of other antioxidants with different biochemical structures and modes of action, and found that NAC induces regeneration through its thiol disulfide exchange activity. Thus, our results provide, for the first time, a biochemical basis for induction of retina regeneration. Furthermore, NAC induction was independent of FGF receptor signaling, but dependent on the MAPK (pErk1/2) pathway.
Collapse
Affiliation(s)
- Nancy Echeverri-Ruiz
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Tracy Haynes
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Joseph Landers
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Justin Woods
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Michael J Gemma
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Michael Hughes
- Department of Statistics and Statistical Consulting Center, Miami University, Oxford, OH 45056, USA
| | - Katia Del Rio-Tsonis
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| |
Collapse
|
24
|
Le Moal E, Pialoux V, Juban G, Groussard C, Zouhal H, Chazaud B, Mounier R. Redox Control of Skeletal Muscle Regeneration. Antioxid Redox Signal 2017; 27:276-310. [PMID: 28027662 PMCID: PMC5685069 DOI: 10.1089/ars.2016.6782] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/24/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022]
Abstract
Skeletal muscle shows high plasticity in response to external demand. Moreover, adult skeletal muscle is capable of complete regeneration after injury, due to the properties of muscle stem cells (MuSCs), the satellite cells, which follow a tightly regulated myogenic program to generate both new myofibers and new MuSCs for further needs. Although reactive oxygen species (ROS) and reactive nitrogen species (RNS) have long been associated with skeletal muscle physiology, their implication in the cell and molecular processes at work during muscle regeneration is more recent. This review focuses on redox regulation during skeletal muscle regeneration. An overview of the basics of ROS/RNS and antioxidant chemistry and biology occurring in skeletal muscle is first provided. Then, the comprehensive knowledge on redox regulation of MuSCs and their surrounding cell partners (macrophages, endothelial cells) during skeletal muscle regeneration is presented in normal muscle and in specific physiological (exercise-induced muscle damage, aging) and pathological (muscular dystrophies) contexts. Recent advances in the comprehension of these processes has led to the development of therapeutic assays using antioxidant supplementation, which result in inconsistent efficiency, underlying the need for new tools that are aimed at precisely deciphering and targeting ROS networks. This review should provide an overall insight of the redox regulation of skeletal muscle regeneration while highlighting the limits of the use of nonspecific antioxidants to improve muscle function. Antioxid. Redox Signal. 27, 276-310.
Collapse
Affiliation(s)
- Emmeran Le Moal
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Vincent Pialoux
- Laboratoire Interuniversitaire de Biologie de la Motricité, EA7424, Université Claude Bernard Lyon 1, Univ Lyon, Villeurbanne, France
- Institut Universitaire de France, Paris, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Carole Groussard
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Hassane Zouhal
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| |
Collapse
|
25
|
Takizawa H, Fritsch K, Kovtonyuk LV, Saito Y, Yakkala C, Jacobs K, Ahuja AK, Lopes M, Hausmann A, Hardt WD, Gomariz Á, Nombela-Arrieta C, Manz MG. Pathogen-Induced TLR4-TRIF Innate Immune Signaling in Hematopoietic Stem Cells Promotes Proliferation but Reduces Competitive Fitness. Cell Stem Cell 2017; 21:225-240.e5. [DOI: 10.1016/j.stem.2017.06.013] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 04/22/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023]
|
26
|
Effects of Antioxidant Supplements on the Survival and Differentiation of Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5032102. [PMID: 28770021 PMCID: PMC5523230 DOI: 10.1155/2017/5032102] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Although physiological levels of reactive oxygen species (ROS) are required to maintain the self-renewal capacity of stem cells, elevated ROS levels can induce chromosomal aberrations, mitochondrial DNA damage, and defective stem cell differentiation. Over the past decade, several studies have shown that antioxidants can not only mitigate oxidative stress and improve stem cell survival but also affect the potency and differentiation of these cells. Further beneficial effects of antioxidants include increasing genomic stability, improving the adhesion of stem cells to culture media, and enabling researchers to manipulate stem cell proliferation by using different doses of antioxidants. These findings can have several clinical implications, such as improving neurogenesis in patients with stroke and neurodegenerative diseases, as well as improving the regeneration of infarcted myocardial tissue and the banking of spermatogonial stem cells. This article reviews the cellular and molecular effects of antioxidant supplementation to cultured or transplanted stem cells and draws up recommendations for further research in this area.
Collapse
|
27
|
Wu M, Gu L, Gong Q, Sun J, Ma Y, Wu H, Wang Y, Guo G, Li X, Zhu H. Strategies to reduce the intracellular effects of iron oxide nanoparticle degradation. Nanomedicine (Lond) 2017; 12:555-570. [PMID: 28181458 DOI: 10.2217/nnm-2016-0328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have a significant self-renewal capacity and can differentiate into a variety of cell types. Cell labeling is crucial as it is difficult to detect cell fate after transplantation in vivo. MSCs labeled with iron oxide nanoparticles (IONPs), which can be tracked by MRI, have tremendous potential in regenerative medicine and oncological research. As a part of nanoparticle, the iron oxide core is a key aspect that can exhibit adverse or beneficial effects on MSCs labeled for tracking. Some IONPs exhibit adverse effects, such as cytotoxicity and apoptosis, while other IONPs exhibit beneficial functions that can promote both MSC proliferation and homing efficiency. This review reveals the cytotoxic mechanisms and potential functions of the iron oxide core of IONPs in cell labeling as well as strategies for minimizing the intracellular effects of IONPs.
Collapse
Affiliation(s)
- Min Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lei Gu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jiayu Sun
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yiqi Ma
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Haoxing Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yu Wang
- College of Life Science, Sichuan Normal University, Chengdu 610068, China
| | - Gang Guo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xue Li
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Subramani B, Subbannagounder S, Ramanathanpullai C, Palanivel S, Ramasamy R. Impaired redox environment modulates cardiogenic and ion-channel gene expression in cardiac-resident and non-resident mesenchymal stem cells. Exp Biol Med (Maywood) 2017; 242:645-656. [PMID: 28092181 DOI: 10.1177/1535370216688568] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Redox homeostasis plays a crucial role in the regulation of self-renewal and differentiation of stem cells. However, the behavioral actions of mesenchymal stem cells in redox imbalance state remain elusive. In the present study, the effect of redox imbalance that was induced by either hydrogen peroxide (H2O2) or ascorbic acid on human cardiac-resident (hC-MSCs) and non-resident (umbilical cord) mesenchymal stem cells (hUC-MSCs) was evaluated. Both cells were sensitive and responsive when exposed to either H2O2 or ascorbic acid at a concentration of 400 µmol/L. Ascorbic acid pre-treated cells remarkably ameliorated the reactive oxygen species level when treated with H2O2. The endogenous antioxidative enzyme gene (Sod1, Sod2, TRXR1 and Gpx1) expressions were escalated in both MSCs in response to reactive oxygen species elevation. In contrast, ascorbic acid pre-treated hUC-MSCs attenuated considerable anti-oxidative gene (TRXR1 and Gpx1) expressions, but not the hC-MSCs. Similarly, the cardiogenic gene (Nkx 2.5, Gata4, Mlc2a and β-MHC) and ion-channel gene ( IKDR, IKCa, Ito and INa.TTX) expressions were significantly increased in both MSCs on the oxidative state. On the contrary, reduced environment could not alter the ion-channel gene expression and negatively regulated the cardiogenic gene expressions except for troponin-1 in both cells. In conclusion, redox imbalance potently alters the cardiac-resident and non-resident MSCs stemness, cardiogenic, and ion-channel gene expressions. In comparison with cardiac-resident MSC, non-resident umbilical cord-MSC has great potential to tolerate the redox imbalance and positively respond to cardiac regeneration. Impact statement Human mesenchymal stem cells (h-MSCs) are highly promising candidates for tissue repair in cardiovascular diseases. However, the retention of cells in the infarcted area has been a major challenge due to its poor viability and/or low survival rate after transplantation. The regenerative potential of mesenchymal stem cells (MSCs) repudiate and enter into premature senescence via oxidative stress. Thus, various strategies have been attempted to improve the MSC survival in 'toxic' conditions. Similarly, we investigated the response of cardiac resident MSC (hC-MSCs) and non-resident MSCs against the oxidative stress induced by H2O2. Supplementation of ascorbic acid (AA) into MSCs culture profoundly rescued the stem cells from oxidative stress induced by H2O2. Our data showed that the pre-treatment of AA is able to inhibit the cell death and thus preserving the viability and differentiation potential of MSCs.
Collapse
Affiliation(s)
- Baskar Subramani
- 1 Nichi-Asia Life Science Sdn Bhd., Petaling Jaya 47810, Selangor, Malaysia
- 2 Department of Microbiology, Bharathiyar University, Coimbatore, Tamil Nadu 641046, India
| | | | | | - Sekar Palanivel
- 3 Department of Zoology, Arignar Anna Government Arts College, Namakkal, Tamil Nadu 637002, India
| | - Rajesh Ramasamy
- 4 Stem Cell & Immunity Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia
- 5 Stem Cell Research Laboratory, Genetic and Regenerative Medicine Research Center, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
29
|
Liu Y, Xiong Y, Xing F, Gao H, Wang X, He L, Ren C, Liu L, So KF, Xiao J. Precise Regulation of miR-210 Is Critical for the Cellular Homeostasis Maintenance and Transplantation Efficacy Enhancement of Mesenchymal Stem Cells in Acute Liver Failure Therapy. Cell Transplant 2016; 26:805-820. [PMID: 27983913 DOI: 10.3727/096368916x694274] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Stem cell transplantation is a promising clinical strategy to cure acute liver failure. However, a low cell survival ratio after transplantation significantly impairs its therapeutic efficacy. This is partly due to insufficient resistance of transplanted stem cells to severe oxidative and inflammatory stress at the injury sites. In the current study, we demonstrated that a small molecule zeaxanthin dipalmitate (ZD) could enhance the defensive abilities against adverse stresses of human adipose-derived mesenchymal stem cells (hADMSCs) in vitro and increase their therapeutic outcomes of acute liver failure after transplantation in vivo. Treatment with ZD dramatically improved cell survival and suppressed apoptosis, inflammation, and reactive oxygen species (ROS) production of hADMSCs through the PKC/Raf-1/MAPK/NF-κB pathway to maintain a reasonably high expression level of microRNA-210 (miR-210). The regulation loop between miR-210 and cellular/mitochondrial ROS production was found to be linked by the ROS inhibitor iron-sulfur cluster assembly proteins (ISCU). Pretreatment with ZD and stable knockdown of miR-210 significantly improved and impaired the stem cell transplantation efficacy through the alteration of hepatic cell expansion and injury amelioration, respectively. Vehicle treatment with ZD did not pose any adverse effect on cell homeostasis or healthy animal. In conclusion, elevating endogenous antioxidant level of hADMSCs with ZD significantly enhances their hepatic tissue-repairing capabilities. Maintenance of a physiological level of miR-210 is critical for hADMSC homeostasis.
Collapse
|
30
|
The superior regenerative potential of muscle-derived stem cells for articular cartilage repair is attributed to high cell survival and chondrogenic potential. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16065. [PMID: 27990446 PMCID: PMC5129874 DOI: 10.1038/mtm.2016.65] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 06/22/2016] [Indexed: 11/08/2022]
Abstract
Three populations of muscle-derived cells (PP1, PP3, and PP6) were isolated from mouse skeletal muscle using modified preplate technique and retrovirally transduced with BMP4/GFP. In vitro, the PP1 cells (fibroblasts) proliferated significantly slower than the PP3 (myoblasts) and PP6 cells (muscle-derived stem cells); the PP1 and PP6 cells showed a superior rate of survival compared with PP3 cells under oxidative stress; and the PP6 cells showed significantly superior chondrogenic capabilities than PP1 and PP3 cells. In vivo, the PP6 cells promoted superior cartilage regeneration compared with the other muscle-derived cell populations. The cartilage defects in the PP6 group had significantly higher histological scores than those of the other muscle-derived cell groups, and GFP detection revealed that the transplanted PP6 cells showed superior in vivo cell survival and chondrogenic capabilities compared with the PP1 and PP3 cells. PP6 cells (muscle-derived stem cells) are superior to other primary muscle-derived cells for use as a cellular vehicle for BMP4-based ex vivo gene therapy to heal full-thickness osteo-chondral defects. The superiority of the PP6/muscle-derived stem cells appears to be attributable to a combination of increased rate of in vivo survival and superior chondrogenic differentiation capacity.
Collapse
|
31
|
Therapeutic Strategies for Oxidative Stress-Related Cardiovascular Diseases: Removal of Excess Reactive Oxygen Species in Adult Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2483163. [PMID: 27668035 PMCID: PMC5030421 DOI: 10.1155/2016/2483163] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
Abstract
Accumulating evidence indicates that acute and chronic uncontrolled overproduction of oxidative stress-related factors including reactive oxygen species (ROS) causes cardiovascular diseases (CVDs), atherosclerosis, and diabetes. Moreover ROS mediate various signaling pathways underlying vascular inflammation in ischemic tissues. With respect to stem cell-based therapy, several studies clearly indicate that modulating antioxidant production at cellular levels enhances stem/progenitor cell functionalities, including proliferation, long-term survival in ischemic tissues, and complete differentiation of transplanted cells into mature vascular cells. Recently emerging therapeutic strategies involving adult stem cells, including endothelial progenitor cells (EPCs), for treating ischemic CVDs have highlighted the need to control intracellular ROS production, because it critically affects the replicative senescence of ex vivo expanded therapeutic cells. Better understanding of the complexity of cellular ROS in stem cell biology might improve cell survival in ischemic tissues and enhance the regenerative potentials of transplanted stem/progenitor cells. In this review, we will discuss the nature and sources of ROS, drug-based therapeutic strategies for scavenging ROS, and EPC based therapeutic strategies for treating oxidative stress-related CVDs. Furthermore, we will discuss whether primed EPCs pretreated with natural ROS-scavenging compounds are crucial and promising therapeutic strategies for vascular repair.
Collapse
|
32
|
Abdelwahid E, Kalvelyte A, Stulpinas A, de Carvalho KAT, Guarita-Souza LC, Foldes G. Stem cell death and survival in heart regeneration and repair. Apoptosis 2016; 21:252-68. [PMID: 26687129 PMCID: PMC5200890 DOI: 10.1007/s10495-015-1203-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are major causes of mortality and morbidity. Cardiomyocyte apoptosis disrupts cardiac function and leads to cardiac decompensation and terminal heart failure. Delineating the regulatory signaling pathways that orchestrate cell survival in the heart has significant therapeutic implications. Cardiac tissue has limited capacity to regenerate and repair. Stem cell therapy is a successful approach for repairing and regenerating ischemic cardiac tissue; however, transplanted cells display very high death percentage, a problem that affects success of tissue regeneration. Stem cells display multipotency or pluripotency and undergo self-renewal, however these events are negatively influenced by upregulation of cell death machinery that induces the significant decrease in survival and differentiation signals upon cardiovascular injury. While efforts to identify cell types and molecular pathways that promote cardiac tissue regeneration have been productive, studies that focus on blocking the extensive cell death after transplantation are limited. The control of cell death includes multiple networks rather than one crucial pathway, which underlies the challenge of identifying the interaction between various cellular and biochemical components. This review is aimed at exploiting the molecular mechanisms by which stem cells resist death signals to develop into mature and healthy cardiac cells. Specifically, we focus on a number of factors that control death and survival of stem cells upon transplantation and ultimately affect cardiac regeneration. We also discuss potential survival enhancing strategies and how they could be meaningful in the design of targeted therapies that improve cardiac function.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, 303 E. Chicago Ave., Tarry 14-725, Chicago, IL, 60611, USA.
| | - Audrone Kalvelyte
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Aurimas Stulpinas
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Katherine Athayde Teixeira de Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pequeno Príncipe Faculty, Pelé Pequeno Príncipe Institute, Curitiba, Paraná, 80250-200, Brazil
| | - Luiz Cesar Guarita-Souza
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Parana, Curitiba, Paraná, 80215-901, Brazil
| | - Gabor Foldes
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
33
|
Kozakowska M, Pietraszek-Gremplewicz K, Jozkowicz A, Dulak J. The role of oxidative stress in skeletal muscle injury and regeneration: focus on antioxidant enzymes. J Muscle Res Cell Motil 2016; 36:377-93. [PMID: 26728750 PMCID: PMC4762917 DOI: 10.1007/s10974-015-9438-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are generated in skeletal muscle both during the rest and contractile activity. Myogenic cells are equipped with antioxidant enzymes, like superoxide dismutase, catalase, glutathione peroxidase, γ-glutamylcysteine synthetase and heme oxygenase-1. These enzymes not only neutralise excessive ROS, but also affect myogenic regeneration at several stages: influence post-injury inflammatory reaction, enhance viability and proliferation of muscle satellite cells and myoblasts and affect their differentiation. Finally, antioxidant enzymes regulate also processes accompanying muscle regeneration-induce angiogenesis and reduce fibrosis. Elevated ROS production was also observed in Duchenne muscular dystrophy (DMD), a disease characterised by degeneration of muscle tissue and therefore-increased rate of myogenic regeneration. Antioxidant enzymes are consequently considered as target for therapies counteracting dystrophic symptoms. In this review we present current knowledge regarding the role of oxidative stress and systems of enzymatic antioxidant defence in muscular regeneration after both acute injury and persistent muscular degeneration.
Collapse
Affiliation(s)
- Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Pietraszek-Gremplewicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland. .,Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
34
|
Salem MY, El-Eraky El-Azab N, Helal OK, Gabr Metwaly H, Abd El-Halim Bayoumi HE. Does selenium improve the stem cell therapeutic effect on isoproterenol-induced myocardial infarction in rats? A histological and immunohistochemical study. THE EGYPTIAN JOURNAL OF HISTOLOGY 2015; 38:679-691. [DOI: 10.1097/01.ehx.0000475224.41506.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
35
|
Huang CC, Pan WY, Tseng MT, Lin KJ, Yang YP, Tsai HW, Hwang SM, Chang Y, Wei HJ, Sung HW. Enhancement of cell adhesion, retention, and survival of HUVEC/cbMSC aggregates that are transplanted in ischemic tissues by concurrent delivery of an antioxidant for therapeutic angiogenesis. Biomaterials 2015; 74:53-63. [PMID: 26447555 DOI: 10.1016/j.biomaterials.2015.09.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 01/01/2023]
Abstract
A recurring obstacle in cell-base strategies for treating ischemic diseases is the significant loss of viable cells that is caused by the elevated levels of regional reactive oxygen species (ROS), which ultimately limits therapeutic capacity. In this study, aggregates of human umbilical vein endothelial cells (HUVECs) and cord-blood mesenchymal stem cells (cbMSCs), which are capable of inducing therapeutic angiogenesis, are prepared. We hypothesize that the concurrent delivery of an antioxidant N-acetylcysteine (NAC) may significantly increase cell retention following the transplantation of HUVEC/cbMSC aggregates in a mouse model with hindlimb ischemia. Our in vitro results demonstrate that the antioxidant NAC can restore ROS-impaired cell adhesion and recover the reduced angiogenic potential of HUVEC/cbMSC aggregates under oxidative stress. In the animal study, we found that by scavenging the ROS generated in ischemic tissues, NAC is likely to be able to establish a receptive cell environment in the early stage of cell transplantation, promoting the adhesion, retention, and survival of cells of engrafted aggregates. Therapeutic angiogenesis is therefore enhanced and blood flow recovery and limb salvage are ultimately achieved. The combinatory strategy that uses an antioxidant and HUVEC/cbMSC aggregates may provide a new means of boosting the therapeutic efficacy of cell aggregates for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Chieh-Cheng Huang
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Wen-Yu Pan
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC; Division of Cardiovascular Surgery, Veterans General Hospital-Taichung, and College of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Michael T Tseng
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, USA
| | - Kun-Ju Lin
- Healthy Aging Research Center, Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC; Department of Nuclear Medicine and Center of Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Linkou, Taiwan, ROC
| | - Yi-Pei Yang
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Hung-Wen Tsai
- Division of Cardiovascular Surgery, Veterans General Hospital-Taichung, and College of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, ROC
| | - Yen Chang
- Division of Cardiovascular Surgery, Veterans General Hospital-Taichung, and College of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Hao-Ji Wei
- Division of Cardiovascular Surgery, Veterans General Hospital-Taichung, and College of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC; Division of Cardiovascular Surgery, Chiayi Branch, Veterans General Hospital-Taichung, Chiayi, Taiwan, ROC.
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC.
| |
Collapse
|
36
|
Pisciotta A, Riccio M, Carnevale G, Lu A, De Biasi S, Gibellini L, La Sala GB, Bruzzesi G, Ferrari A, Huard J, De Pol A. Stem cells isolated from human dental pulp and amniotic fluid improve skeletal muscle histopathology in mdx/SCID mice. Stem Cell Res Ther 2015; 6:156. [PMID: 26316011 PMCID: PMC4552417 DOI: 10.1186/s13287-015-0141-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/07/2015] [Accepted: 07/30/2015] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD), caused by a lack of the functional structural protein dystrophin, leads to severe muscle degeneration where the patients are typically wheelchair-bound and die in their mid-twenties from cardiac or respiratory failure or both. The aim of this study was to investigate the potential of human dental pulp stem cells (hDPSCs) and human amniotic fluid stem cells (hAFSCs) to differentiate toward a skeletal myogenic lineage using several different protocols in order to determine the optimal conditions for achieving myogenic commitment and to subsequently evaluate their contribution in the improvement of the pathological features associated with dystrophic skeletal muscle when intramuscularly injected into mdx/SCID mice, an immune-compromised animal model of DMD. METHODS Human DPSCs and AFSCs were differentiated toward myogenic lineage in vitro through the direct co-culture with a myogenic cell line (C2C12 cells) and through a preliminary demethylation treatment with 5-Aza-2'-deoxycytidine (5-Aza), respectively. The commitment and differentiation of both hDPSCs and hAFSCs were evaluated by immunofluorescence and Western blot analysis. Subsequently, hDPSCs and hAFSCs, preliminarily demethylated and pre-differentiated toward a myogenic lineage for 2 weeks, were injected into the dystrophic gastrocnemius muscles of mdx/SCID mice. After 1, 2, and 4 weeks, the gastrocnemius muscles were taken for immunofluorescence and histological analyses. RESULTS Both populations of cells engrafted within the host muscle of mdx/SCID mice and through a paracrine effect promoted angiogenesis and reduced fibrosis, which eventually led to an improvement of the histopathology of the dystrophic muscle. CONCLUSION This study shows that hAFSCs and hDPSCs represent potential sources of stem cells for translational strategies to improve the histopathology and potentially alleviate the muscle weakness in patients with DMD.
Collapse
Affiliation(s)
- Alessandra Pisciotta
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.
| | - Massimo Riccio
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.
| | - Gianluca Carnevale
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.
| | - Aiping Lu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, 450 Technology Drive, Bridgeside Point II, Suite 206, 15219, Pittsburgh, PA, USA.
| | - Sara De Biasi
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.
| | - Lara Gibellini
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.
| | - Giovanni B La Sala
- Department of Obstetrics and Gynecology, Arcispedale Santa Maria Nuova, viale Risorgimento 80, 42123, Reggio Emilia, Italy.
| | - Giacomo Bruzzesi
- Oro-Maxillo-Facial Department, AUSL Baggiovara, via Giardini 1355, 41126, Modena, Baggiovara, Italy.
| | - Adriano Ferrari
- Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, Children Rehabilitation Special Unit, IRCCS Arcispedale Santa Maria Nuova, viale Risorgimento 80, 42123, Reggio Emilia, Italy.
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, 450 Technology Drive, Bridgeside Point II, Suite 206, 15219, Pittsburgh, PA, USA.
| | - Anto De Pol
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.
| |
Collapse
|
37
|
Zeng W, Xiao J, Zheng G, Xing F, Tipoe GL, Wang X, He C, Chen ZY, Liu Y. Antioxidant treatment enhances human mesenchymal stem cell anti-stress ability and therapeutic efficacy in an acute liver failure model. Sci Rep 2015; 5:11100. [PMID: 26057841 PMCID: PMC4460871 DOI: 10.1038/srep11100] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/12/2015] [Indexed: 02/07/2023] Open
Abstract
One of the major problems influencing the therapeutic efficacy of stem cell therapy is the poor cell survival following transplantation. This is partly attributed to insufficient resistance of transplanted stem cells to oxidative and inflammatory stresses at the injured sites. In the current study, we demonstrated the pivotal role of antioxidant levels in human umbilical cord mesenchymal stem cells (hUCMSCs) dynamic in vitro anti-stress abilities against lipopolysaccharide (LPS)/H2O2 intoxication and in vivo therapeutic efficacy in a murine acute liver failure model induced by D-galactosamine/LPS (Gal/LPS) by either reducing the antioxidant levels with diethyl maleate (DEM) or increasing antioxidant levels with edaravone. Both the anti- and pro-oxidant treatments dramatically influenced the survival, apoptosis, and reactive oxygen species (ROS) production of hUCMSCs through the MAPK-PKC-Nrf2 pathway in vitro. When compared with untreated and DEM-treated cells, edaravone-treated hUCMSCs rescued NOD/SCID mice from Gal/LPS-induced death, significantly improved hepatic functions and promoted host liver regeneration. These effects were probably from increased stem cell homing, promoted proliferation, decreased apoptosis and enhanced secretion of hepatocyte growth factor (HGF) under hepatic stress environment. In conclusion, elevating levels of antioxidants in hUCMSCs with edaravone can significantly influence their hepatic tissue repair capacity.
Collapse
Affiliation(s)
- Wen Zeng
- State key Discipline of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.,Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jia Xiao
- State key Discipline of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.,Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China.,Department of Anatomy, The University of Hong Kong, Hong Kong, China
| | - Gang Zheng
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Feiyue Xing
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - George L Tipoe
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
| | - Xiaogang Wang
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Chengyi He
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhi-Ying Chen
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yingxia Liu
- State key Discipline of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| |
Collapse
|
38
|
Stilhano RS, Martins L, Ingham SJM, Pesquero JB, Huard J. Gene and cell therapy for muscle regeneration. Curr Rev Musculoskelet Med 2015; 8:182-187. [PMID: 25899573 DOI: 10.1007/s12178-015-9268-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Skeletal muscle injury and healing are multifactorial processes, involving three steps of healing: (1) degeneration and inflammation, (2) regeneration, and (3) fibrosis. Fibrous tissue hinders the muscle's complete recovery and current therapies fail in achieving total muscle recovery. Gene and cell therapy (or both) are potential future treatments for severe muscular injuries. Stem cells' properties associated with growth factors or/and cytokines can improve muscle healing and permit long-term recovery.
Collapse
Affiliation(s)
- Roberta Sessa Stilhano
- Biophysics Department, Federal University of São Paulo - UNIFESP, Rua Mirassol, 207 - Vila Clementino, 04044-010 São Paulo, Brazil
| | - Leonardo Martins
- Biophysics Department, Federal University of São Paulo - UNIFESP, Rua Mirassol, 207 - Vila Clementino, 04044-010 São Paulo, Brazil
| | | | - João Bosco Pesquero
- Biophysics Department, Federal University of São Paulo - UNIFESP, Rua Mirassol, 207 - Vila Clementino, 04044-010 São Paulo, Brazil
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
39
|
Goichberg P, Chang J, Liao R, Leri A. Cardiac stem cells: biology and clinical applications. Antioxid Redox Signal 2014; 21:2002-17. [PMID: 24597850 PMCID: PMC4208604 DOI: 10.1089/ars.2014.5875] [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: 02/06/2023]
Abstract
SIGNIFICANCE Heart disease is the primary cause of death in the industrialized world. Cardiac failure is dictated by an uncompensated reduction in the number of viable and fully functional cardiomyocytes. While current pharmacological therapies alleviate the symptoms associated with cardiac deterioration, heart transplantation remains the only therapy for advanced heart failure. Therefore, there is a pressing need for novel therapeutic modalities. Cell-based therapies involving cardiac stem cells (CSCs) constitute a promising emerging approach for the replenishment of the lost tissue and the restoration of cardiac contractility. RECENT ADVANCES CSCs reside in the adult heart and govern myocardial homeostasis and repair after injury by producing new cardiomyocytes and vascular structures. In the last decade, different classes of immature cells expressing distinct stem cell markers have been identified and characterized in terms of their growth properties, differentiation potential, and regenerative ability. Phase I clinical trials, employing autologous CSCs in patients with ischemic cardiomyopathy, are being completed with encouraging results. CRITICAL ISSUES Accumulating evidence concerning the role of CSCs in heart regeneration imposes a reconsideration of the mechanisms of cardiac aging and the etiology of heart failure. Deciphering the molecular pathways that prevent activation of CSCs in their environment and understanding the processes that affect CSC survival and regenerative function with cardiac pathologies, commonly accompanied by alterations in redox conditions, are of great clinical importance. FUTURE DIRECTIONS Further investigations of CSC biology may be translated into highly effective and novel therapeutic strategies aiming at the enhancement of the endogenous healing capacity of the diseased heart.
Collapse
Affiliation(s)
- Polina Goichberg
- Departments of Anesthesia and Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | | | | | | |
Collapse
|
40
|
Cho GS, Fernandez L, Kwon C. Regenerative medicine for the heart: perspectives on stem-cell therapy. Antioxid Redox Signal 2014; 21:2018-31. [PMID: 25133793 PMCID: PMC4208610 DOI: 10.1089/ars.2014.6063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Despite decades of progress in cardiovascular biology and medicine, heart disease remains the leading cause of death, and there is no cure for the failing heart. Since heart failure is mostly caused by loss or dysfunction of cardiomyocytes (CMs), replacing dead or damaged CMs with new CMs might be an ideal way to reverse the disease. However, the adult heart is composed mainly of terminally differentiated CMs that have no significant self-regeneration capacity. RECENT ADVANCES Stem cells have tremendous regenerative potential and, thus, current cardiac regenerative research has focused on developing stem cell sources to repair damaged myocardium. CRITICAL ISSUES In this review, we examine the potential sources of cells that could be used for heart therapies, including embryonic stem cells and induced pluripotent stem cells, as well as alternative methods for activating the endogenous regenerative mechanisms of the heart via transdifferentiation and cell reprogramming. We also discuss the current state of knowledge of cell purification, delivery, and retention. FUTURE DIRECTIONS Efforts are underway to improve the current stem cell strategies and methodologies, which will accelerate the development of innovative stem-cell therapies for heart regeneration.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Johns Hopkins University , Baltimore, Maryland
| | | | | |
Collapse
|
41
|
Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat Med 2014; 20:870-80. [PMID: 25100532 DOI: 10.1038/nm.3651] [Citation(s) in RCA: 508] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 07/09/2014] [Indexed: 12/14/2022]
Abstract
Aging tissues experience a progressive decline in homeostatic and regenerative capacities, which has been attributed to degenerative changes in tissue-specific stem cells, stem cell niches and systemic cues that regulate stem cell activity. Understanding the molecular pathways involved in this age-dependent deterioration of stem cell function will be critical for developing new therapies for diseases of aging that target the specific causes of age-related functional decline. Here we explore key molecular pathways that are commonly perturbed as tissues and stem cells age and degenerate. We further consider experimental evidence both supporting and refuting the notion that modulation of these pathways per se can reverse aging phenotypes. Finally, we ask whether stem cell aging establishes an epigenetic 'memory' that is indelibly written or one that can be reset.
Collapse
|
42
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes: multitasking cells in the regeneration of injured, diseased, and aged skeletal muscle. Front Aging Neurosci 2014; 6:245. [PMID: 25278877 PMCID: PMC4166895 DOI: 10.3389/fnagi.2014.00245] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/29/2014] [Indexed: 12/16/2022] Open
Abstract
Pericytes are perivascular cells that envelop and make intimate connections with adjacent capillary endothelial cells. Recent studies show that they may have a profound impact in skeletal muscle regeneration, innervation, vessel formation, fibrosis, fat accumulation, and ectopic bone formation throughout life. In this review, we summarize and evaluate recent advances in our understanding of pericytes' influence on adult skeletal muscle pathophysiology. We also discuss how further elucidating their biology may offer new approaches to the treatment of conditions characterized by muscle wasting.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Maria L Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| |
Collapse
|
43
|
Beckman SA, Sekiya N, Chen WC, Mlakar L, Tobita K, Huard J. The cardiac regenerative potential of myoblasts remains limited despite improving their survival via antioxidant treatment. CELLR4-- REPAIR, REPLACEMENT, REGENERATION, & REPROGRAMMING 2014; 2:e845. [PMID: 28989945 PMCID: PMC5627517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Since myoblasts have been limited by poor cell survival after cellular myoplasty, the major goal of the current study was to determine whether improving myoblast survival with an antioxidant could improve cardiac function after the transplantation of the myoblasts into an acute myocardial infarction. BACKGROUND We previously demonstrated that early myogenic progenitors such as muscle-derived stem cells (MDSCs) exhibited superior cell survival and improved cardiac repair after transplantation into infarcted hearts compared to myoblasts, which we partially attributed to MDSC's higher antioxidant levels. AIM To determine if antioxidant treatment could increase myoblast survival, subsequently improving cardiac function after myoblast transplantation into infarcted hearts. MATERIALS AND METHODS Myoblasts were pre-treated with the antioxidant N-acetylcysteine (NAC) or the glutathione depleter, diethyl maleate (DEM), and injected into infarcted murine hearts. Regenerative potential was monitored by cell survival and cardiac function. RESULTS At early time points, hearts injected with NAC-treated myoblasts exhibited increased donor cell survival, greater cell proliferation, and decreased cellular apoptosis, compared to untreated myoblasts. NAC-treated myoblasts significantly improved cardiac contractility, reduced fibrosis, and increased vascular density compared to DEM-treated myoblasts, but compared to untreated myoblasts, no difference was noted. DISCUSSION While early survival of myoblasts transplanted into infarcted hearts was augmented by NAC pre-treatment, cardiac function remained unchanged compared to non-treated myoblasts. CONCLUSION Despite improving cell survival with NAC treated myoblast transplantation in a MI heart, cardiac function remained similar to untreated myoblasts. These results suggest that the reduced cardiac regenerative potential of myoblasts, when compared to MDSCs, is not only attributable to cell survival but is probably also related to the secretion of paracrine factors by the MDSCs.
Collapse
Affiliation(s)
- Sarah A. Beckman
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Naosumi Sekiya
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - William C.W. Chen
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Department of BioEngineering, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Logan Mlakar
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Kimimassa Tobita
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Department of BioEngineering, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
| |
Collapse
|
44
|
Sicari BM, Dearth CL, Badylak SF. Tissue Engineering and Regenerative Medicine Approaches to Enhance the Functional Response to Skeletal Muscle Injury. Anat Rec (Hoboken) 2013; 297:51-64. [DOI: 10.1002/ar.22794] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/14/2022]
Affiliation(s)
- Brian M. Sicari
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Cellular and Molecular Pathology Graduate Program; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Christopher L. Dearth
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| |
Collapse
|
45
|
Chung JH, Kim YS, Noh K, Lee YM, Chang SW, Kim EC. Deferoxamine promotes osteoblastic differentiation in human periodontal ligament cells via the nuclear factor erythroid 2-related factor-mediated antioxidant signaling pathway. J Periodontal Res 2013; 49:563-73. [PMID: 24111577 DOI: 10.1111/jre.12136] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE Recently it was reported that deferoxamine (DFO), an iron chelator, stimulates bone formation from MG63 and mesenchymal stem cells, but inhibits differentiation in rat calvarial cells; however, the effect of DFO on osteoblastic differentiation in human periodontal ligament cells (hPDLCs) has not been reported. The aim of this study was to investigate the effects and the possible underlying mechanism of DFO on osteoblastic differentiation of hPDLCs. MATERIAL AND METHODS The effect of DFO on osteoblast differentiation was determined by the staining intensity of calcium deposits with Alizarin red and by RT-PCR analysis of the expression of osteoblastic markers. Signal transduction pathways were analyzed by western blotting. RESULTS DFO increased osteogenic differentiation in a concentration-dependent manner by expression of the mRNA for differentiation markers and calcium nodule formation. Exposure of hPDLCs to DFO resulted in increases in the production of reactive oxygen species and in the levels of nuclear factor erythroid 2-related factor (Nrf2) protein in nuclear extractions, as well as a dose-dependent increase in the expression of Nrf2 target genes, including glutathione (GSH), glutathione S-transferase, γ-glutamylcysteine lygase, glutathione reductase and glutathione peroxidase. Pretreatment with Nrf2 small interfering RNA, GSH depletion by buthionine sulfoximine and diethyl maleate, and with antioxidants by N-acetylcysteine and vitamin E, blocked DFO-stimulated osteoblastic differentiation. Furthermore, pretreatment with GSH depletion and antioxidants blocked DFO-induced p38 MAPK, ERK, JNK and nuclear factor-kappaB pathways. CONCLUSION These data indicate, for the first time, that nontoxic DFO promotes osteoblastic differentiation of hPDLCs via modulation of the Nrf2-mediated antioxidant pathway.
Collapse
Affiliation(s)
- J H Chung
- Department of Periodontology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
46
|
N-acetylcysteine-pretreated human embryonic mesenchymal stem cell administration protects against bleomycin-induced lung injury. Am J Med Sci 2013; 346:113-22. [PMID: 23085672 DOI: 10.1097/maj.0b013e318266e8d8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The transplantation of mesenchymal stem cells (MSCs) has been reported to be a promising approach in the treatment of acute lung injury. However, the poor efficacy of transplanted MSCs is one of the serious handicaps in the progress of MSC-based therapy. Therefore, the purpose of this study was to investigate whether the pretreatment of human embryonic MSCs (hMSCs) with an antioxidant, namely N-acetylcysteine (NAC), can improve the efficacy of hMSC transplantation in lung injury. METHODS In vitro, the antioxidant capacity of NAC-pretreated hMSCs was assessed using intracellular reactive oxygen species (ROS) and glutathione assays and cell adhesion and spreading assays. In vivo, the therapeutic potential of NAC-pretreated hMSCs was assessed in a bleomycin-induced model of lung injury in nude mice. RESULTS The pretreatment of hMSCs with NAC improved antioxidant capacity to defend against redox imbalances through the elimination of cellular ROS, increasing cellular glutathione levels, and the enhancement of cell adhesion and spreading when exposed to oxidative stresses in vitro. In addition, the administration of NAC-pretreated hMSCs to nude mice with bleomycin-induced lung injury decreased the pathological grade of lung inflammation and fibrosis, hydroxyproline content and numbers of neutrophils and inflammatory cytokines in bronchoalveolar lavage fluid and apoptotic cells, while enhancing the retention and proliferation of hMSCs in injured lung tissue and improving the survival rate of mice compared with results from untreated hMSCs. CONCLUSIONS The pretreatment of hMSCs with NAC could be a promising therapeutic approach to improving cell transplantation and, therefore, the treatment of lung injury.
Collapse
|
47
|
Saparov A, Chen CW, Beckman SA, Wang Y, Huard J. The role of antioxidation and immunomodulation in postnatal multipotent stem cell-mediated cardiac repair. Int J Mol Sci 2013; 14:16258-79. [PMID: 23924945 PMCID: PMC3759910 DOI: 10.3390/ijms140816258] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/23/2013] [Accepted: 07/30/2013] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress and inflammation play major roles in the pathogenesis of coronary heart disease including myocardial infarction (MI). The pathological progression following MI is very complex and involves a number of cell populations including cells localized within the heart, as well as cells recruited from the circulation and other tissues that participate in inflammatory and reparative processes. These cells, with their secretory factors, have pleiotropic effects that depend on the stage of inflammation and regeneration. Excessive inflammation leads to enlargement of the infarction site, pathological remodeling and eventually, heart dysfunction. Stem cell therapy represents a unique and innovative approach to ameliorate oxidative stress and inflammation caused by ischemic heart disease. Consequently, it is crucial to understand the crosstalk between stem cells and other cells involved in post-MI cardiac tissue repair, especially immune cells, in order to harness the beneficial effects of the immune response following MI and further improve stem cell-mediated cardiac regeneration. This paper reviews the recent findings on the role of antioxidation and immunomodulation in postnatal multipotent stem cell-mediated cardiac repair following ischemic heart disease, particularly acute MI and focuses specifically on mesenchymal, muscle and blood-vessel-derived stem cells due to their antioxidant and immunomodulatory properties.
Collapse
Affiliation(s)
- Arman Saparov
- Nazarbayev University Research and Innovation System, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (J.H.); Tel.: +7-717-270-6140 (A.S.); +1-412-648-2798 (J.H.); Fax: +7-717-270-6054 (A.S.); +1-412-648-4066 (J.H.)
| | - Chien-Wen Chen
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; E-Mail:
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Sarah A. Beckman
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; E-Mail:
| | - Yadong Wang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; E-Mail:
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (J.H.); Tel.: +7-717-270-6140 (A.S.); +1-412-648-2798 (J.H.); Fax: +7-717-270-6054 (A.S.); +1-412-648-4066 (J.H.)
| |
Collapse
|
48
|
Das B, Bayat-Mokhtari R, Tsui M, Lotfi S, Tsuchida R, Felsher DW, Yeger H. HIF-2α suppresses p53 to enhance the stemness and regenerative potential of human embryonic stem cells. Stem Cells 2013; 30:1685-95. [PMID: 22689594 PMCID: PMC3584519 DOI: 10.1002/stem.1142] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human embryonic stem cells (hESCs) have been reported to exert cytoprotective activity in the area of tissue injury. However, hypoxia/oxidative stress prevailing in the area of injury could activate p53, leading to death and differentiation of hESCs. Here we report that when exposed to hypoxia/oxidative stress, a small fraction of hESCs, namely the SSEA3+/ABCG2+ fraction undergoes a transient state of reprogramming to a low p53 and high hypoxia inducible factor (HIF)-2α state of transcriptional activity. This state can be sustained for a period of 2 weeks and is associated with enhanced transcriptional activity of Oct-4 and Nanog, concomitant with high teratomagenic potential. Conditioned medium obtained from the post-hypoxia SSEA3+/ABCG2+ hESCs showed cytoprotection both in vitro and in vivo. We termed this phenotype as the “enhanced stemness” state. We then demonstrated that the underlying molecular mechanism of this transient phenotype of enhanced stemness involved high Bcl-2, fibroblast growth factor (FGF)-2, and MDM2 expression and an altered state of the p53/MDM2 oscillation system. Specific silencing of HIF-2α and p53 resisted the reprogramming of SSEA3+/ABCG2+ to the enhanced stemness phenotype. Thus, our studies have uncovered a unique transient reprogramming activity in hESCs, the enhanced stemness reprogramming where a highly cytoprotective and undifferentiated state is achieved by transiently suppressing p53 activity. We suggest that this transient reprogramming is a form of stem cell altruism that benefits the surrounding tissues during the process of tissue regeneration.
Collapse
Affiliation(s)
- Bikul Das
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
El Haddad M, Jean E, Turki A, Hugon G, Vernus B, Bonnieu A, Passerieux E, Hamade A, Mercier J, Laoudj-Chenivesse D, Carnac G. Glutathione peroxidase 3, a new retinoid target gene, is crucial for human skeletal muscle precursor cell survival. J Cell Sci 2012; 125:6147-56. [PMID: 23132926 DOI: 10.1242/jcs.115220] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Protection of satellite cells from cytotoxic damages is crucial to ensure efficient adult skeletal muscle regeneration and to improve therapeutic efficacy of cell transplantation in degenerative skeletal muscle diseases. It is therefore important to identify and characterize molecules and their target genes that control the viability of muscle stem cells. Recently, we demonstrated that high aldehyde dehydrogenase activity is associated with increased viability of human myoblasts. In addition to its detoxifying activity, aldehyde dehydrogenase can also catalyze the irreversible oxidation of vitamin A to retinoic acid; therefore, we examined whether retinoic acid is important for myoblast viability. We showed that when exposed to oxidative stress induced by hydrogen peroxide, adherent human myoblasts entered apoptosis and lost their capacity for adhesion. Pre-treatment with retinoic acid reduced the cytotoxic damage ex vivo and enhanced myoblast survival in transplantation assays. The effects of retinoic acid were maintained in dystrophic myoblasts derived from facioscapulohumeral patients. RT-qPCR analysis of antioxidant gene expression revealed glutathione peroxidase 3 (Gpx3), a gene encoding an antioxidant enzyme, as a potential retinoic acid target gene in human myoblasts. Knockdown of Gpx3 using short interfering RNA induced elevation in reactive oxygen species and cell death. The anti-cytotoxic effects of retinoic acid were impaired in GPx3-inactivated myoblasts, which indicates that GPx3 regulates the antioxidative effects of retinoic acid. Therefore, retinoid status and GPx3 levels may have important implications for the viability of human muscle stem cells.
Collapse
Affiliation(s)
- Marina El Haddad
- Inserm U1046, Université Montpellier 1, 34295 Montpellier, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gao X, Usas A, Lu A, Tang Y, Wang B, Chen CW, Li H, Tebbets JC, Cummins JH, Huard J. BMP2 is superior to BMP4 for promoting human muscle-derived stem cell-mediated bone regeneration in a critical-sized calvarial defect model. Cell Transplant 2012; 22:2393-408. [PMID: 23244588 DOI: 10.3727/096368912x658854] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Muscle-derived cells have been successfully isolated using a variety of different methods and have been shown to possess multilineage differentiation capacities, including an ability to differentiate into articular cartilage and bone in vivo; however, the characterization of human muscle-derived stem cells (hMDSCs) and their bone regenerative capacities have not been fully investigated. Genetic modification of these cells may enhance their osteogenic capacity, which could potentially be applied to bone regenerative therapies. We found that hMDSCs, isolated by the preplate technique, consistently expressed the myogenic marker CD56, the pericyte/endothelial cell marker CD146, and the mesenchymal stem cell markers CD73, CD90, CD105, and CD44 but did not express the hematopoietic stem cell marker CD45, and they could undergo osteogenic, chondrogenic, adipogenic, and myogenic differentiation in vitro. In order to investigate the osteoinductive potential of hMDSCs, we constructed a retroviral vector expressing BMP4 and GFP and a lentiviral vector expressing BMP2. The BMP4-expressing hMDSCs were able to undergo osteogenic differentiation in vitro and exhibited enhanced mineralization compared to nontransduced cells; however, when transplanted into a calvarial defect, they failed to regenerate bone. Local administration of BMP4 protein and cell pretreatment with N-acetylcysteine (NAC), which improves cell survival, did not enhance the osteogenic capacity of the retro-BMP4-transduced cells. In contrast, lenti-BMP2-transduced hMDSCs not only exhibited enhanced in vitro osteogenic differentiation but also induced robust bone formation and nearly completely healed a critical-sized calvarial defect in CD-1 nude mice 6 weeks following transplantation. Herovici's staining of the regenerated bone demonstrated that the bone matrix contained a large amount of type I collagen. Our findings indicated that the hMDSCs are likely mesenchymal stem cells of muscle origin and that BMP2 is more efficient than BMP4 in promoting the bone regenerative capacity of the hMDSCs in vivo.
Collapse
Affiliation(s)
- Xueqin Gao
- Stem Cell Research Center, Growth and Developmental Laboratory, Department of Orthopaedic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|