1
|
Seow KS, Ling APK. Mesenchymal stem cells as future treatment for cardiovascular regeneration and its challenges. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:73. [PMID: 39118948 PMCID: PMC11304428 DOI: 10.21037/atm-23-1936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 12/04/2023] [Indexed: 08/10/2024]
Abstract
Cardiovascular diseases (CVDs), particularly stroke and myocardial infarction (MI) contributed to the leading cause of death annually among the chronic diseases globally. Despite the advancement of technology, the current available treatments mainly served as palliative care but not treating the diseases. However, the discovery of mesenchymal stem cells (MSCs) had gained a consideration to serve as promising strategy in treating CVDs. Recent evidence also showed that MSCs are the strong candidate to be used as stem cell therapy involving cardiovascular regeneration due to its cardiomyogenesis, anti-inflammatory and immunomodulatory properties, antifibrotic effects and neovascularization capacity. Besides, MSCs could be used for cellular cardiomyoplasty with its transdifferentiation of MSCs into cardiomyocytes, paracrine effects, microvesicles and exosomes as well as mitochondrial transfer. The safety and efficacy of utilizing MSCs have been described in well-established preclinical and clinical studies in which the accomplishment of MSCs transplantation resulted in further improvement of the cardiac function. Tissue engineering could enhance the desired properties and therapeutic effects of MSCs in cardiovascular regeneration by genome-editing, facilitating the cell delivery and retention, biomaterials-based scaffold, and three-dimensional (3D)-bioprinting. However, there are still obstacles in the use of MSCs due to the complexity and versatility of MSCs, low retention rate, route of administration and the ethical and safety issues of the use of MSCs. The aim of this review is to highlight the details of therapeutic properties of MSCs in treating CVDs, strategies to facilitate the therapeutic effects of MSCs through tissue engineering and the challenges faced using MSCs. A comprehensive review has been done through PubMed and National Center for Biotechnology Information (NCBI) from the year of 2010 to 2021 based on some specific key terms such as 'mesenchymal stem cells in cardiovascular disease', 'mesenchymal stem cells in cardiac regeneration', 'mesenchymal stem cells facilitate cardiac repairs', 'tissue engineering of MSCs' to include relevant literature in this review.
Collapse
Affiliation(s)
- Ke Sin Seow
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Xu D, Fu J, Liu X, Hong Y, Chen X, Li S, Hou J, Zhang K, Zhou C, Zeng C, Zheng G, Wu H, Wang T. ELABELA-APJ Axis Enhances Mesenchymal Stem Cell Proliferation and Migration via the METTL3/PI3K/AKT Pathway. Acta Naturae 2024; 16:111-118. [PMID: 38698964 PMCID: PMC11062101 DOI: 10.32607/actanaturae.17863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/13/2024] [Indexed: 05/05/2024] Open
Abstract
Mesenchymal stem cells (MSCs) possess a strong therapeutic potential in regenerative medicine. ELABELA (ELA) is a 32 amino acid peptide that binds to the apelin peptide jejunum receptor (APJ) to regulate cell proliferation and migration. The aim of this study was to investigate the function of ELA vis-a-vis the MSC proliferation and migration, and further explore the underlying mechanism. We demonstrated that the exogenous supplement of ELA boosts the proliferation and migration ability of MSCs, alongside improved in vitro cell viability. These capabilities were rendered moot upon APJ knockdown. In addition, ELA (5-20 μM) was shown to upregulate the expression of METTL3 in a concentrationdependent pattern, a capacity which was suppressed by APJ reduction, whereas the downregulation of METTL3 expression blocked the beneficial effects induced by ELA. ELA was also observed to upregulate the phosphorylation level of AKT. This ELA-induced activation of the PI3K/AKT pathway, however, is inhibited with knockdown of METTL3. Our data indicate that ELA could act as a promoter of MSC proliferation and migration in vitro through the APJ receptor, something which might be attributed to the activation of the METTL3/PI3K/AKT signaling pathway. Therefore, ELA is a candidate for optimizing MSC-based cell therapy, while METTL3 is a potential target for its promoting action on MSCs.
Collapse
Affiliation(s)
- D. Xu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - J. Fu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - X. Liu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510120 China
| | - Y. Hong
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - X. Chen
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - S. Li
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - J. Hou
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510120 China
| | - K. Zhang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510120 China
| | - C. Zhou
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - C. Zeng
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510120 China
| | - G. Zheng
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510120 China
| | - H. Wu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| | - T. Wang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003 China
| |
Collapse
|
3
|
Li H, Wang S, Hui Y, Ren Y, Li J, Lan X, Wang Y. The implication of blue light-emitting diode on mesenchymal stem cells: a systematic review. Lasers Med Sci 2023; 38:267. [PMID: 37981584 DOI: 10.1007/s10103-023-03908-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 11/21/2023]
Abstract
The application of blue light (400-480 nm) in photobiotherapy remains controversial. This systematic review aimed to collect and analyze the biological effects of blue light-emitting diode (LED) on mesenchymal stem cells (MSCs). Inclusion and exclusion criteria were formulated, and relevant English articles from January 1982 to September 2022 were searched in PubMed, Scopus, and Web of Science. Nine articles with a medium (n = 4) to low (n = 5) risk of bias were included. Most of the MSCs reported were derived from human tissue; only one article used MSCs derived from mouse. The wavelength of the LED used was in the 400-480 nm range, and the irradiation modes were continuous (n = 8) and pulse waves (n = 1). A chiral polarizer was used in one such study in which the irradiance was 14 mW/cm2 and the irradiation time was 24 h. The energy densities used in other studies were between 0.378 and 72 J/cm2, and the irradiation times were between 10 and 3600 s. Blue LED light can inhibit proliferation and promote differentiation of MSCs in an appropriate energy density range, which may be related to the activation of transient receptor potential vanilloid 1 (TRPV1). Additionally, polarized light may reduce the toxic effects of blue light on MSCs. However, the heterogeneity of the design schemes and LED parameters, as well as the small number of studies, limited the conclusiveness of the review. Therefore, further studies are needed to determine the optimal irradiation strategy for promoting MSC function.
Collapse
Affiliation(s)
- Hao Li
- Department of Preventive Health Care, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Shifen Wang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Yining Hui
- Southwest Medical University School of Clinical Medicine, Luzhou, 646000, China
| | - Yajiao Ren
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Jiaxin Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Yao Wang
- Department of Preventive Health Care, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China.
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China.
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
4
|
Mansano BSDM, da Rocha VP, Teixeira ILA, de Oliveira HA, Vieira SS, Antonio EL, Tucci PJF, Serra AJ. Light-emitting Diode Can Enhance the Metabolism and Paracrine Action of Mesenchymal Stem Cells. Photochem Photobiol 2023; 99:1420-1428. [PMID: 36807286 DOI: 10.1111/php.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
This study investigated the influence of red light-emitting diodes (LED, 630 nm) on different irradiation parameters and the number of applications on mesenchymal stem cells derived from adipose tissue (AdMSCs) metabolism and paracrine factors. The AdMSCs were irradiated with a LEDbox device (output power: 2452.5 mW; laser beam: 163.5 cm2 ; irradiance: 15 mW cm-2 ) using radiant exposures of 0.5, 2, and 4 J cm-2 , respectively. AdMSCs were irradiated once or every 48 h up to three irradiations. All molecular analyses were performed 24 h after the last irradiation. LED did not induce changes in cell count, DNA damage, and oxidative stress. A significant repercussion of the LED has been noticed after three irradiations with 4 J cm-2 . AdMSCs had higher levels of IL-6, IGF-1, and NOx index. A higher ATP content and MMT/Resazurin assay were identified in AdMSCs irradiated three times with 4 J cm-2 . Mitochondrial basal respiration, maximal respiration and proton leak under metabolic stress were reduced by 0.5 and 2 J cm-2 irradiations. These data showed that three LED irradiations with 4 J cm-2 may be a suitable parameter for future AdMSCs therapy because of its improved metabolic activity, ATP content, and IL-6, IGF-1, and nitric oxide secretion.
Collapse
Affiliation(s)
| | - Vitor Pocani da Rocha
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | | | - Stella Souza Vieira
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
- Base Hospital Foundation, Medicine School of São José do Rio Preto, Sao Paulo, SP, Brazil
| | - Ednei Luiz Antonio
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
| | | | - Andrey Jorge Serra
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo, SP, Brazil
| |
Collapse
|
5
|
Bumroongthai K, Kavanagh DPJ, Genever P, Kalia N. Improving vasculoprotective effects of MSCs in coronary microvessels - benefits of 3D culture, sub-populations and heparin. Front Immunol 2023; 14:1257497. [PMID: 37954606 PMCID: PMC10635425 DOI: 10.3389/fimmu.2023.1257497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/02/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction Opening occluded coronary arteries in patients with myocardial infarction (MI) damages the delicate coronary microvessels through a process called myocardial ischaemia-reperfusion injury. Although mesenchymal stromal cells (MSCs) have the potential to limit this injury, clinical success remains limited. This may be due to (i) poor MSC homing to the heart (ii) infused MSCs, even if derived from the same site, being a heterogeneous population with varying therapeutic efficacy and (iii) conventional 2D culture of MSCs decreasing their homing and beneficial properties. This study investigated whether 3D culture of two distinctly different bone marrow (BM)-derived MSC sub-populations could improve their homing and coronary vasculoprotective efficacy. Methods Intravital imaging of the anaesthetised mouse beating heart was used to investigate the trafficking and microvascular protective effects of two clonally-derived BM-derived MSC lines, namely CD317neg MSCs-Y201 and CD317pos MSCs-Y202, cultured using conventional monolayer and 3D hanging drop methods. Results 3D culture consistently improved the adhesive behaviour of MSCs-Y201 to various substrates in vitro. However, it was their differential ability to reduce neutrophil events within the coronary capillaries and improve ventricular perfusion in vivo that was most remarkable. Moreover, dual therapy combined with heparin further improved the vasculoprotection afforded by 3D cultured MSCs-Y201 by also modifying platelet as well as neutrophil recruitment, which subsequently led to the greatest salvage of viable myocardium. Therapeutic benefit could mechanistically be explained by reductions in coronary endothelial oxidative stress and intercellular adhesion molecule-1 (ICAM-1)/vascular cell adhesion molecule-1 (VCAM-1) expression. However, since this was noted by both 2D and 3D cultured MSCs-Y201, therapeutic benefit is likely explained by the fact that 3D cultured MSCs-Y201 were the most potent sub-population at reducing serum levels of several pro-inflammatory cytokines. Conclusion This novel study highlights the importance of not only 3D culture, but also of a specific CD317neg MSC sub-population, as being critical to realising their full coronary vasculoprotective potential in the injured heart. Since the smallest coronary blood vessels are increasingly recognised as a primary target of reperfusion injury, therapeutic interventions must be able to protect these delicate structures from inflammatory cells and maintain perfusion in the heart. We propose that relatively feasible technical modifications in a specific BM-derived MSC sub-population could achieve this.
Collapse
Affiliation(s)
- Kobkaew Bumroongthai
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dean P. J. Kavanagh
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Genever
- Department of Biology, University of York, York, United Kingdom
| | - Neena Kalia
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
6
|
El-Husseiny HM, Mady EA, El-Dakroury WA, Doghish AS, Tanaka R. Stimuli-responsive hydrogels: smart state of-the-art platforms for cardiac tissue engineering. Front Bioeng Biotechnol 2023; 11:1174075. [PMID: 37449088 PMCID: PMC10337592 DOI: 10.3389/fbioe.2023.1174075] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Biomedicine and tissue regeneration have made significant advancements recently, positively affecting the whole healthcare spectrum. This opened the way for them to develop their applications for revitalizing damaged tissues. Thus, their functionality will be restored. Cardiac tissue engineering (CTE) using curative procedures that combine biomolecules, biomimetic scaffolds, and cells plays a critical part in this path. Stimuli-responsive hydrogels (SRHs) are excellent three-dimensional (3D) biomaterials for tissue engineering (TE) and various biomedical applications. They can mimic the intrinsic tissues' physicochemical, mechanical, and biological characteristics in a variety of ways. They also provide for 3D setup, adequate aqueous conditions, and the mechanical consistency required for cell development. Furthermore, they function as competent delivery platforms for various biomolecules. Many natural and synthetic polymers were used to fabricate these intelligent platforms with innovative enhanced features and specialized capabilities that are appropriate for CTE applications. In the present review, different strategies employed for CTE were outlined. The light was shed on the limitations of the use of conventional hydrogels in CTE. Moreover, diverse types of SRHs, their characteristics, assembly and exploitation for CTE were discussed. To summarize, recent development in the construction of SRHs increases their potential to operate as intelligent, sophisticated systems in the reconstruction of degenerated cardiac tissues.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Eman A. Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Walaa A. El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Egypt
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
7
|
Wang W, Tayier B, Guan L, Yan F, Mu Y. Pre-transplantation of Bone Marrow Mesenchymal Stem Cells Amplifies the Therapeutic Effect of Ultrasound-Targeted Microbubble Destruction-Mediated Localized Combined Gene Therapy in Post-Myocardial Infarction Heart Failure Rats. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:830-845. [PMID: 35246339 DOI: 10.1016/j.ultrasmedbio.2022.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Although stem cell transplantation and single-gene therapy have been intensively discussed separately as treatments for myocardial infarction (MI) hearts and have exhibited ideal therapeutic efficiency in animal models, clinical trials turned out to be disappointing. Here, we deliver sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) and connexin 43 (Cx43) genes simultaneously via an ultrasound-targeted microbubble destruction (UTMD) approach to chronic MI hearts that have been pre-treated with bone marrow mesenchymal stem cells (BMSCs) to amplify cardiac repair. First, biotinylated microbubbles (BMBs) were fabricated, and biotinylated recombinant adenoviruses carrying the SERCA2a or Cx43 gene were conjugated to the surface of self-assembled BMBs to form SERCA2a-BMBs, Cx43-BMBs or dual gene-loaded BMBs. Then, the general characteristics of these bubbles, including particle size, concentration, contrast signal and gene loading capacity, were examined. Second, a rat myocardial infarction model was created by ligating the left anterior descending coronary artery and injecting BMSCs into the infarct and border zones. Four weeks later, co-delivery of SERCA2a and Cx43 genes to the infarcted heart were delivered together to the infarcted heart using the UTMD approach. Cardiac mechano-electrical function was determined 4 wk after gene transfection, and the infarcted hearts were collected for myocardial infarct size measurement and detection of expression of SERCA2a, Cx43 and cardiac-specific markers. Finally, to validate the role of BMSC transplantation, MI rats transplanted or not with BMSCs were transfected with SERCA2a and Cx43, and the cardiac mechano-electrical function of these two groups of rats was recorded and compared. General characteristics of the self-assembled gene-loaded BMBs were qualified, and the gene loading rate was satisfactory. The self-assembled gene-loaded BMBs were in microscale and exhibit satisfactory dual-gene loading capacity. High transfection efficiency was achieved under ultrasound irradiation in vitro. In addition, rats in which SERCA2a and Cx43 were overexpressed simultaneously had the best contractile function and electrical stability among all experimental groups. Immunofluorescence assay revealed that the levels of SERCA2a and/or Cx43 proteins were significantly elevated, especially in the border zone. Moreover, compared with rats that did not receive BMSCs, rats pre-treated with BMSCs have better mechano-electrical function after transfection with SERCA2a and Cx43. Collectively, we report a promising cardiac repair strategy for post-MI hearts that exploits the providential advantages of stem cell therapy and UTMD-mediated localized co-delivery of specific genes.
Collapse
Affiliation(s)
- Wei Wang
- Department of Echocardiography, Xinjiang Medical University First Affiliated Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Baihetiya Tayier
- Department of Echocardiography, Xinjiang Medical University First Affiliated Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Lina Guan
- Department of Echocardiography, Xinjiang Medical University First Affiliated Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuming Mu
- Department of Echocardiography, Xinjiang Medical University First Affiliated Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China.
| |
Collapse
|
8
|
A 3D Mathematical Model of Coupled Stem Cell-Nutrient Dynamics in Myocardial Regeneration Therapy. J Theor Biol 2022; 537:111023. [PMID: 35041851 DOI: 10.1016/j.jtbi.2022.111023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/04/2021] [Accepted: 01/09/2022] [Indexed: 11/23/2022]
Abstract
Stem cell therapy is a promising treatment for the regeneration of myocardial tissue injured by an ischemic event. Mathematical modeling of myocardial regeneration via stem cell therapy is a challenging task, since the mechanisms underlying the processes involved in the treatment are not yet fully understood. Many aspects must be accounted for, such as the spread of stem cells and nutrients, chemoattraction, cell proliferation, stages of cell maturation, differentiation, angiogenesis, stochastic effects, just to name a few. In this paper we propose a 3D mathematical model with a free boundary that aims to provide a qualitative description of some main aspects of the stem cell regenerative therapy in a simplified scenario. The paper mainly focuses on the description of the shrinking of the necrotic core during treatment. The stem cell and nutrients dynamics are described through coupled reaction-diffusion problems. Proliferation, chemoattraction, tissue regeneration and nutrient consumption are included in the model.
Collapse
|
9
|
Overexpression of NMNAT3 improves mitochondrial function and enhances anti-oxidative stress of bone marrow mesenchymal stem cells via the NAD+-Sirt3 pathway. Biosci Rep 2022; 42:230593. [PMID: 34981121 PMCID: PMC8762348 DOI: 10.1042/bsr20211005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/28/2021] [Accepted: 12/22/2021] [Indexed: 12/06/2022] Open
Abstract
Oxidative stress damage is a common problem in bone marrow mesenchymal stem cell (BMSC) transplantation. Under stress conditions, the mitochondrial function of BMSCs is disrupted, which accelerates senescence and apoptosis of BMSCs, ultimately leading to poor efficacy. Therefore, improving mitochondrial function and enhancing the anti-oxidative stress capacity of BMSCs may be an effective way of improving the survival rate and curative effect of BMSCs. In this study, we have confirmed that overexpression of nicotinamide mononucleotide adenylyl transferase 3 (NMNAT3) improves mitochondrial function and resistance to stress-induced apoptosis in BMSCs. We further revealed the mechanism of NMNAT3-mediated resistance to stress-induced apoptosis in BMSCs. We increased the level of nicotinamide adenine dinucleotide (NAD+) by overexpressing NMNAT3 in BMSCs and found that it could significantly increase the activity of silent mating type information regulation 2 homolog 3 (Sirt3) and significantly decrease the acetylation levels of Sirt3-dependent deacetylation-related proteins isocitrate dehydrogenase 2 (Idh2) and Forkhead-box protein O3a (FOXO3a). These findings show that NMNAT3 may increase the activity of Sirt3 by increasing NAD+ levels. Our results confirm that the NMNAT3-NAD+-Sirt3 axis is a potential mechanism for improving mitochondrial function and enhancing anti-oxidative stress of BMSCs. In this study, we take advantage of the role of NMNAT3 in inhibiting stress-induced apoptosis of BMSCs and provide new methods and ideas for breaking through the bottleneck of transplantation efficacy of BMSCs in the clinic.
Collapse
|
10
|
Tsubosaka M, Maruyama M, Huang EE, Zhang N, Utsunomiya T, Gao Q, Shen H, Li X, Kushioka J, Hirata H, Yao Z, Yang YP, Goodman SB. Effect on Osteogenic Differentiation of Genetically Modified IL4 or PDGF-BB Over-Expressing and IL4-PDGF-BB Co-Over-Expressing Bone Marrow-Derived Mesenchymal Stromal Cells In Vitro. Bioengineering (Basel) 2021; 8:bioengineering8110165. [PMID: 34821731 PMCID: PMC8614682 DOI: 10.3390/bioengineering8110165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022] Open
Abstract
The use of genetically modified (GM) mesenchymal stromal cells (MSCs) and preconditioned MSCs (pMSCs) may provide further opportunities to improve the outcome of core decompression (CD) for the treatment of early-stage osteonecrosis of the femoral head (ONFH). GM interleukin-4 (IL4) over-expressing MSCs (IL4-MSCs), platelet-derived growth factor (PDGF)-BB over-expressing MSCs (PDGF-BB-MSCs), and IL4-PDGF-BB co-over-expressing MSCs (IL4-PDGF-BB-MSCs) and their respective pMSCs were used in this in vitro study and compared with respect to cell proliferation and osteogenic differentiation. IL4-MSCs, PDGF-BB-MSCs, IL4-PDGF-BB-MSCs, and each pMSC treatment significantly increased cell proliferation compared to the MSC group alone. The percentage of Alizarin red-stained area in the IL4-MSC and IL4-pMSC groups was significantly lower than in the MSC group. However, the percentage of Alizarin red-stained area in the PDGF-BB-MSC group was significantly higher than in the MSC and PDGF-BB-pMSC groups. The percentage of Alizarin red-stained area in the IL4-PDGF-BB-pMSC was significantly higher than in the IL4-PDGF-BB-MSC group. There were no significant differences in the percentage of Alizarin red-stained area between the MSC and IL4-PDGF-BB-pMSC groups. The use of PDGF-BB-MSCs or IL4-PDGF-BB-pMSCs increased cell proliferation. Furthermore, PDGF-BB-MSCs promoted osteogenic differentiation. The addition of GM MSCs may provide a useful supplementary cell-based therapy to CD for treatment of ONFH.
Collapse
Affiliation(s)
- Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
- Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-650-498-4343
| |
Collapse
|
11
|
Pharmacological Preconditioning Improves the Viability and Proangiogenic Paracrine Function of Hydrogel-Encapsulated Mesenchymal Stromal Cells. Stem Cells Int 2021; 2021:6663467. [PMID: 34367293 PMCID: PMC8342149 DOI: 10.1155/2021/6663467] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/20/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
The efficacy of cell therapy is limited by low retention and survival of transplanted cells in the target tissues. In this work, we hypothesize that pharmacological preconditioning with celastrol, a natural potent antioxidant, could improve the viability and functions of mesenchymal stromal cells (MSC) encapsulated within an injectable scaffold. Bone marrow MSCs from rat (rMSC) and human (hMSC) origin were preconditioned for 1 hour with celastrol 1 μM or vehicle (DMSO 0.1% v/v), then encapsulated within a chitosan-based thermosensitive hydrogel. Cell viability was compared by alamarBlue and live/dead assay. Paracrine function was studied first by quantifying the proangiogenic growth factors released, followed by assessing scratched HUVEC culture wound closure velocity and proliferation of HUVEC when cocultured with encapsulated hMSC. In vivo, the proangiogenic activity was studied by evaluating the neovessel density around the subcutaneously injected hydrogel after one week in rats. Preconditioning strongly enhanced the viability of rMSC and hMSC compared to vehicle-treated cells, with 90% and 75% survival versus 36% and 58% survival, respectively, after 7 days in complete media and 80% versus 64% survival for hMSC after 4 days in low serum media (p < 0.05). Celastrol-treated cells increased quantities of proangiogenic cytokines compared to vehicle-pretreated cells, with a significant 3.0-fold and 1.8-fold increase of VEGFa and SDF-1α, respectively (p < 0.05). The enhanced paracrine function of preconditioned MSC was demonstrated by accelerated growth and wound closure velocity of injured HUVEC monolayer (p < 0.05) in vitro. Moreover, celastrol-treated cells, but not vehicle-treated cells, led to a significant increase of neovessel density in the peri-implant region after one week in vivo compared to the control (blank hydrogel). These results suggest that combining cell pretreatment with celastrol and encapsulation in hydrogel could potentiate MSC therapy for many diseases, benefiting particularly ischemic diseases.
Collapse
|
12
|
Cruz-Samperio R, Jordan M, Perriman A. Cell augmentation strategies for cardiac stem cell therapies. Stem Cells Transl Med 2021; 10:855-866. [PMID: 33660953 PMCID: PMC8133336 DOI: 10.1002/sctm.20-0489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) has been the primary cause of death in developed countries, resulting in a major psychological and financial burden for society. Current treatments for acute MI are directed toward rapid restoration of perfusion to limit damage to the myocardium, rather than promoting tissue regeneration and subsequent contractile function recovery. Regenerative cell therapies (CTs), in particular those using multipotent stem cells (SCs), are in the spotlight for treatment post‐MI. Unfortunately, the efficacy of CTs is somewhat limited by their poor long‐term viability, homing, and engraftment to the myocardium. In response, a range of novel SC‐based technologies are in development to provide additional cellular modalities, bringing CTs a step closer to the clinic. In this review, the current landscape of emerging CTs and their augmentation strategies for the treatment post‐MI are discussed. In doing so, we highlight recent advances in cell membrane reengineering via genetic modifications, recombinant protein immobilization, and the utilization of soft biomimetic scaffold interfaces.
Collapse
Affiliation(s)
| | - Millie Jordan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Adam Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
13
|
Enhancing the Therapeutic Potential of Mesenchymal Stem Cells with Light-Emitting Diode: Implications and Molecular Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6663539. [PMID: 33623634 PMCID: PMC7875639 DOI: 10.1155/2021/6663539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/05/2021] [Accepted: 01/22/2021] [Indexed: 01/08/2023]
Abstract
This study evaluated the effects of light-emitting diode (LED) on mesenchymal stem cells (MSCs). An electronic search was conducted in PubMed/MEDLINE, Scopus, and Web of Science database for articles published from 1980 to February 2020. Ten articles met the search criteria and were included in this review. The risk of bias was evaluated to report quality, safety, and environmental standards. MSCs were derived from adipose tissue, bone marrow, dental pulp, gingiva, and umbilical cord. Protocols for cellular irradiation used red and blue light spectrum with variations of the parameters. The LED has been shown to induce greater cellular viability, proliferation, differentiation, and secretion of growth factors. The set of information available leads to proposing a complex signaling cascade for the action of photobiomodulation, including angiogenic factors, singlet oxygen, mitogen-activated protein kinase/extracellular signal-regulated protein kinase, Janus kinase/signal transducer, and reactive oxygen species. In conclusion, although our results suggest that LED can boost MSCs, a nonuniformity in the experimental protocol, bias, and the limited number of studies reduces the power of systematic review. Further research is essential to find the optimal LED irradiation parameters to boost MSCs function and evaluate its impact in the clinical setting.
Collapse
|
14
|
Galow AM, Goldammer T, Hoeflich A. Xenogeneic and Stem Cell-Based Therapy for Cardiovascular Diseases: Genetic Engineering of Porcine Cells and Their Applications in Heart Regeneration. Int J Mol Sci 2020; 21:ijms21249686. [PMID: 33353186 PMCID: PMC7766969 DOI: 10.3390/ijms21249686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases represent a major health concern worldwide with few therapy options for ischemic injuries due to the limited regeneration potential of affected cardiomyocytes. Innovative cell replacement approaches could facilitate efficient regenerative therapy. However, despite extensive attempts to expand primary human cells in vitro, present technological limitations and the lack of human donors have so far prevented their broad clinical use. Cell xenotransplantation might provide an ethically acceptable unlimited source for cell replacement therapies and bridge the gap between waiting recipients and available donors. Pigs are considered the most suitable candidates as a source for xenogeneic cells and tissues due to their anatomical and physiological similarities with humans. The potential of porcine cells in the field of stem cell-based therapy and regenerative medicine is under intensive investigation. This review outlines the current progress and highlights the most promising approaches in xenogeneic cell therapy with a focus on the cardiovascular system.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Correspondence: ; Tel.: +49-38208-68-723
| | - Tom Goldammer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Molecular Biology and Fish Genetics Unit, Faculty of Agriculture and Environmental Sciences, University of Rostock, 18059 Rostock, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
| |
Collapse
|
15
|
Fu J, Chen X, Liu X, Xu D, Yang H, Zeng C, Long H, Zhou C, Wu H, Zheng G, Wu H, Wang W, Wang T. ELABELA ameliorates hypoxic/ischemic-induced bone mesenchymal stem cell apoptosis via alleviation of mitochondrial dysfunction and activation of PI3K/AKT and ERK1/2 pathways. Stem Cell Res Ther 2020. [PMID: 33317626 DOI: 10.1186/s13287-020-0206-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have exerted their brilliant potential to promote heart repair following myocardial infarction. However, low survival rate of MSCs after transplantation due to harsh conditions with hypoxic and ischemic stress limits their therapeutic efficiency in treating cardiac dysfunction. ELABELA (ELA) serves as a peptide hormone which has been proved to facilitate cell growth, survival, and pluripotency in human embryonic stem cells. Although ELA works as an endogenous ligand of a G protein-coupled receptor APJ (Apelin receptor, APLNR), whether APJ is an essential signal for the function of ELA remains elusive. The effect of ELA on apoptosis of MSCs is still vague. OBJECTIVE We studied the role of ELABELA (ELA) treatment on the anti-apoptosis of MSCs in hypoxic/ischemic (H/I) conditions which mimic the impaired myocardial microenvironment and explored the possible mechanisms in vitro. METHODS MSCs were obtained from donated rats weighing between 80~120 g. MSCs were exposed to serum-free and hypoxic (1% O2) environments for 24 h, which mimics hypoxic/ischemic damage in vivo, using serum-containing normoxic conditions (20% O2) as a negative control. MSCs that were exposed to H/I injury with ELA processing were treated by 5 μM of ELA. Cell viability and apoptosis of MSCs were evaluated by CCK8 and flow cytometry, respectively. Mitochondrial function of MSCs was also assessed according to mitochondrial membrane potential (MMP) and ATP content. The protein expression of key kinases of the PI3K/AKT and ERK1/2 signaling pathways involving t-AKT, p-AKT, t-ERK1/2, and p-ERK1/2, as well as apoptosis-related protein expression of Bcl-2, Bax, and cleaved Caspase 3, were monitored by Western blot. RESULTS We found that ELA treatment of H/I-induced MSCs improved overall cell viability, enhanced Bcl/Bax expression, and decreased Caspase 3 activity. ELA inhibited H/I-induced mitochondrial dysfunction by increasing ATP concentration and suppressing the loss of mitochondrial transmembrane potential. However, this anti-apoptotic property of ELA was restrained in APJ-silenced MSCs. Additionally, ELA treatment induced the phosphorylation of AKT and ERK, while the blockade of PI3K/AKT and ERK1/2 pathways with respective inhibitors, LY294002 and U0126, suppressed the action of ELA. CONCLUSION ELA positively affected on the survival of MSCs and exhibited anti-apoptotic characteristics when exposed to hypoxic/ischemic condition in vitro. Also, the function of ELA was correlated with the APJ receptor, reduced mitochondrial damage, and activation of the PI3K/AKT and ERK1/2 signal axes.
Collapse
Affiliation(s)
- Jiaying Fu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Xuxiang Chen
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Xin Liu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Daishi Xu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Huan Yang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Chaotao Zeng
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Huibao Long
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Changqing Zhou
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Haidong Wu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Guanghui Zheng
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Hao Wu
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Wuming Wang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Tong Wang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China.
| |
Collapse
|
16
|
Fu J, Chen X, Liu X, Xu D, Yang H, Zeng C, Long H, Zhou C, Wu H, Zheng G, Wu H, Wang W, Wang T. ELABELA ameliorates hypoxic/ischemic-induced bone mesenchymal stem cell apoptosis via alleviation of mitochondrial dysfunction and activation of PI3K/AKT and ERK1/2 pathways. Stem Cell Res Ther 2020; 11:541. [PMID: 33317626 PMCID: PMC7734864 DOI: 10.1186/s13287-020-02063-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/02/2020] [Indexed: 12/20/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have exerted their brilliant potential to promote heart repair following myocardial infarction. However, low survival rate of MSCs after transplantation due to harsh conditions with hypoxic and ischemic stress limits their therapeutic efficiency in treating cardiac dysfunction. ELABELA (ELA) serves as a peptide hormone which has been proved to facilitate cell growth, survival, and pluripotency in human embryonic stem cells. Although ELA works as an endogenous ligand of a G protein-coupled receptor APJ (Apelin receptor, APLNR), whether APJ is an essential signal for the function of ELA remains elusive. The effect of ELA on apoptosis of MSCs is still vague. Objective We studied the role of ELABELA (ELA) treatment on the anti-apoptosis of MSCs in hypoxic/ischemic (H/I) conditions which mimic the impaired myocardial microenvironment and explored the possible mechanisms in vitro. Methods MSCs were obtained from donated rats weighing between 80~120 g. MSCs were exposed to serum-free and hypoxic (1% O2) environments for 24 h, which mimics hypoxic/ischemic damage in vivo, using serum-containing normoxic conditions (20% O2) as a negative control. MSCs that were exposed to H/I injury with ELA processing were treated by 5 μM of ELA. Cell viability and apoptosis of MSCs were evaluated by CCK8 and flow cytometry, respectively. Mitochondrial function of MSCs was also assessed according to mitochondrial membrane potential (MMP) and ATP content. The protein expression of key kinases of the PI3K/AKT and ERK1/2 signaling pathways involving t-AKT, p-AKT, t-ERK1/2, and p-ERK1/2, as well as apoptosis-related protein expression of Bcl-2, Bax, and cleaved Caspase 3, were monitored by Western blot. Results We found that ELA treatment of H/I-induced MSCs improved overall cell viability, enhanced Bcl/Bax expression, and decreased Caspase 3 activity. ELA inhibited H/I-induced mitochondrial dysfunction by increasing ATP concentration and suppressing the loss of mitochondrial transmembrane potential. However, this anti-apoptotic property of ELA was restrained in APJ-silenced MSCs. Additionally, ELA treatment induced the phosphorylation of AKT and ERK, while the blockade of PI3K/AKT and ERK1/2 pathways with respective inhibitors, LY294002 and U0126, suppressed the action of ELA. Conclusion ELA positively affected on the survival of MSCs and exhibited anti-apoptotic characteristics when exposed to hypoxic/ischemic condition in vitro. Also, the function of ELA was correlated with the APJ receptor, reduced mitochondrial damage, and activation of the PI3K/AKT and ERK1/2 signal axes.
Collapse
Affiliation(s)
- Jiaying Fu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China.,Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Xuxiang Chen
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Xin Liu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China.,Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Daishi Xu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Huan Yang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Chaotao Zeng
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Huibao Long
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Changqing Zhou
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Haidong Wu
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Guanghui Zheng
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Hao Wu
- Department of Emergency, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Wuming Wang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Tong Wang
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, Guangdong, People's Republic of China.
| |
Collapse
|
17
|
Preconditioned and Genetically Modified Stem Cells for Myocardial Infarction Treatment. Int J Mol Sci 2020; 21:ijms21197301. [PMID: 33023264 PMCID: PMC7582407 DOI: 10.3390/ijms21197301] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemic heart disease and myocardial infarction remain leading causes of mortality worldwide. Existing myocardial infarction treatments are incapable of fully repairing and regenerating the infarcted myocardium. Stem cell transplantation therapy has demonstrated promising results in improving heart function following myocardial infarction. However, poor cell survival and low engraftment at the harsh and hostile environment at the site of infarction limit the regeneration potential of stem cells. Preconditioning with various physical and chemical factors, as well as genetic modification and cellular reprogramming, are strategies that could potentially optimize stem cell transplantation therapy for clinical application. In this review, we discuss the most up-to-date findings related to utilizing preconditioned stem cells for myocardial infarction treatment, focusing mainly on preconditioning with hypoxia, growth factors, drugs, and biological agents. Furthermore, genetic manipulations on stem cells, such as the overexpression of specific proteins, regulation of microRNAs, and cellular reprogramming to improve their efficiency in myocardial infarction treatment, are discussed as well.
Collapse
|
18
|
Amini H, Hashemzadeh S, Heidarzadeh M, Mamipour M, Yousefi M, Saberianpour S, Rahbarghazi R, Nouri M, Sokullu E. Cytoprotective and cytofunctional effect of polyanionic polysaccharide alginate and gelatin microspheres on rat cardiac cells. Int J Biol Macromol 2020; 161:969-976. [DOI: 10.1016/j.ijbiomac.2020.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/26/2020] [Accepted: 06/02/2020] [Indexed: 01/13/2023]
|
19
|
Zhang B, Zhao N, Zhang J, Liu Y, Zhu D, Kong Y. Mesenchymal stem cells rejuvenate cardiac muscle through regulating macrophage polarization. Aging (Albany NY) 2020; 11:3900-3908. [PMID: 31212255 PMCID: PMC6628986 DOI: 10.18632/aging.102009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022]
Abstract
We have shown that the effects of transplantation of CD146+ mesenchymal stem cells (MSCs) on myocardial regeneration after myocardial infarction (MI) exceeds the effects of transplantation of MSCs, likely resulting from reduction of aging-associated cellular reactive oxygen species in injured cardiac muscle cells (CMCs). Since the role of macrophages in the MSC-mediated recovery of heart function after MI remains unclear, this question was thus addressed in the current study. We found that transplantation of MSCs did not alter the total number of the macrophages in the injured heart, but induced their polarization towards a M2-phenotype. Moreover, administration of tumor necrosis factor alpha (TNFα) into MSC-transplanted mice, which prevented M2-polarization of macrophages, abolished the effects of MSCs on recovery of heart function and on the reduction of infarcted cardiac tissue. Thus, our data suggest that MSCs may rejuvenate CMCs after ischemic injury at least partially through induction of M2-polarization of macrophages.
Collapse
Affiliation(s)
- Busheng Zhang
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Naishi Zhao
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jing Zhang
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu Liu
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Dan Zhu
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ye Kong
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
20
|
Zhang B, Zhang J, Zhu D, Kong Y. Mesenchymal stem cells rejuvenate cardiac muscle after ischemic injury. Aging (Albany NY) 2020; 11:63-72. [PMID: 30613028 PMCID: PMC6339792 DOI: 10.18632/aging.101718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 12/06/2018] [Indexed: 01/05/2023]
Abstract
Previous studies have shown that transplantation of mesenchymal stem cells (MSCs) enhances myocardial regeneration after myocardial infarction (MI), primarily resulting from the production and release of trophic growth factors and cytokines by MSCs. However, effects of MSCs or a subtype of MSCs on the ageing of injured cardiac muscle cells (CMCs) are limitedly known. Here, we addressed this question. CD146+ MSCs were isolated from total MSCs (tMSCs), and their effects on injured CMCs were assessed. In vivo, transplantation of isogenic CD146+ MSCs into MI-mice increased the proliferation of CMCs and reduced apoptosis of CMCs in a significantly higher degree than transplantation of tMSCs, resulting in significant improvement of the heart function. In vitro, CMCs were co-cultured under hypoxia condition with CD146+MSCs or tMSCs. We found that CD146+MSCs increased the proliferation of CMCs and reduced apoptosis of CMCs in a significantly higher degree, compared to tMSCs, likely resulting from reduction of aging-associated cellular reactive oxygen species in CMCs. Together, these data suggest that MSCs rejuvenate CMCs after ischemic injury and a subtype of MSCs, CD146+ MSCs, appears to have higher potential in coordinating this process.
Collapse
Affiliation(s)
- Busheng Zhang
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jing Zhang
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan Zhu
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ye Kong
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
21
|
Ye B, Weng Y, Lin S, Lin J, Huang Z, Huang W, Cai X. 1,25(OH) 2D 3 Strengthens the Vasculogenesis of Multipotent Mesenchymal Stromal Cells from Rat Bone Marrow by Regulating the PI3K/AKT Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1157-1167. [PMID: 32214801 PMCID: PMC7083642 DOI: 10.2147/dddt.s222244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 03/03/2020] [Indexed: 12/17/2022]
Abstract
Background Multipotent mesenchymal stromal cells (MSCs) have recently been reported to promote vasculogenesis by differentiating into endothelial cells and releasing numerous cytokines and paracrine factors. However, due to low cell activity, their potential for clinical application is not very satisfactory. This study aimed to explore the effects and mechanisms of 1,25-dihydroxyvitamin D (1,25(OH)2D3) on the vasculogenesis of MSCs. Methods MSCs were isolated from the femurs and tibias of rats and characterized by flow cytometry. After treatment with different concentrations of 1,25(OH)2D3 (0 µM, 0.1 µM and 1 µM), the proliferation of MSCs was analyzed by Cell Counting Kit-8 (CCK-8), and the migratory capability was measured by Transwell assays and cell scratch tests. Capillary-like structure formation was observed by using Matrigel. Western blotting was used to detect the expression of FLK-1 and vWF to investigate the differentiation of MSCs into endothelial cells. Western blotting and gelatin zymography were used to detect the expression and activities of VEGF, MMP-2 and MMP-9 secreted by MSCs under the influence of 1,25(OH)2D3. Finally, the VDR antagonist pyridoxal-5-phosphate (P5P) and the PI3K/AKT pathway inhibitor LY294002 were utilized to test the phosphorylation levels of key kinases in the PI3K/AKT pathway by Western blotting and the formation of capillary-like structures in Matrigel. Results The proliferation and migratory capability of MSCs and the ability of MSCs to form a tube-like structure in Matrigel were enhanced after treatment with 1,25(OH)2D3. Moreover, MSCs treated with 1,25(OH)2D3 showed high expression of vWF and Flk-1. There was a significant increase in the expression of VEGF, MMP-2 and MMP-9 secreted by MSCs treated with 1,25(OH)2D3, as well as in the activity of MMP-2 and MMP-9. The phosphorylation level of AKT increased with time after 1,25(OH)2D3 treatment, while LY294002 weakened AKT phosphorylation. In addition, the ability to form capillary-like structures was reduced when the VDR and PI3K/AKT pathways were blocked. Conclusion This study confirmed that 1,25(OH)2D3 treatment can strengthen the ability of MSCs to promote vasculogenesis in vitro, and the mechanism may be related to the activation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Bozhi Ye
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yawen Weng
- Department of Pediatrics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Shuang Lin
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Jiahui Lin
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Zhouqing Huang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Weijian Huang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xueli Cai
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
22
|
Pinto DS, Ahsan T, Serra J, Fernandes-Platzgummer A, Cabral JMS, da Silva CL. Modulation of the in vitro angiogenic potential of human mesenchymal stromal cells from different tissue sources. J Cell Physiol 2020; 235:7224-7238. [PMID: 32037550 DOI: 10.1002/jcp.29622] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stromal cells (MSCs) have been widely exploited for the treatment of several conditions due to their intrinsic regenerative and immunomodulatory properties. MSC have demonstrated to be particularly relevant for the treatment of ischemic diseases, where MSC-based therapies can stimulate angiogenesis and induce tissue regeneration. Regardless of the condition targeted, recent analyses of MSC-based clinical trials have demonstrated limited benefits indicating a need to improve the efficacy of this cell product. Preconditioning MSC ex vivo through microenvironment modulation was found to improve MSC survival rate and thus prolong their therapeutic effect. This workstudy aims at enhancing the in vitro angiogenic capacity of a potential MSC-based medicinal product by comparing different sources of MSC and culture conditions. MSC from three different sources (bone marrow [BM], adipose tissue [AT], and umbilical cord matrix [UCM]) were cultured with xenogeneic-/serum-free culture medium under static conditions and their angiogenic potential was studied. Results indicated a higher in vitro angiogenic capacity of UCM MSC, compared with cells derived from BM and AT. Physicochemical preconditioning of UCM MSC through a microcarrier-based culture platform and low oxygen concentration (2% O2 , compared with atmospheric air) increased the in vitro angiogenic potential of the cultured cells. Envisaging the clinical manufacturing of an allogeneic, off-the-shelf MSC-based product, preconditioned UCM MSC maintain the angiogenic gene expression profile upon cryopreservation and delivery processes in the conditions of our study. These results are expected to contribute to the development of MSC-based therapies in the context of angiogenesis.
Collapse
Affiliation(s)
- Diogo S Pinto
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Tabassum Ahsan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Joana Serra
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
23
|
Nakao M, Inanaga D, Nagase K, Kanazawa H. Characteristic differences of cell sheets composed of mesenchymal stem cells with different tissue origins. Regen Ther 2019; 11:34-40. [PMID: 31193157 PMCID: PMC6517796 DOI: 10.1016/j.reth.2019.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/28/2018] [Accepted: 01/06/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Stem cell therapy with mesenchymal stem cells (MSCs) has been widely used in many clinical trials, and therapy with MSC sheets shows promise for patients. However, there are few reports characterizing MSC sheets. In the present study, the properties of MSC sheets derived from bone marrow, adipose tissue, and umbilical cord were evaluated. METHODS Cell sheets were fabricated with MSCs from different tissue origins in temperature-responsive cell culture dishes with and without pre-coating of fetal bovine serum (FBS). MSC adhesion behavior in the culture dish was observed. Secretion of cytokines related to cell proliferation and immune regulation from MSC sheets was investigated by ELISA. The adhesion properties of the MSC sheets were investigated by time-lapse microscopy. RESULTS Different cell adhesion and proliferation rates in temperature-responsive cell culture dishes were observed among the three types of MSCs. FBS pre-coating of the dishes enhanced cell attachment and proliferation in all cell types. Harvested cell sheets showed high attachment capacity to tissue culture polystyrene dish surfaces. CONCLUSIONS MSC sheets can be fabricated from MSCs from different tissue origins using temperature-responsive cell culture dishes. The fabricated MSC sheets could be useful in cell transplantation therapies by choosing appropriate types of MSCs that secrete therapeutic cytokines for the targeted diseases.
Collapse
Affiliation(s)
| | | | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo, 105-8512, Japan
| | - Hideko Kanazawa
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo, 105-8512, Japan
| |
Collapse
|
24
|
Hummitzsch L, Zitta K, Rusch R, Cremer J, Steinfath M, Gross J, Fandrich F, Berndt R, Albrecht M. Characterization of the Angiogenic Potential of Human Regulatory Macrophages (Mreg) after Ischemia/Reperfusion Injury In Vitro. Stem Cells Int 2019; 2019:3725863. [PMID: 31341483 PMCID: PMC6614961 DOI: 10.1155/2019/3725863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Ischemia/reperfusion- (I/R-) induced organ damage represents one of the main causes of death worldwide, and new strategies to reduce I/R injury are urgently needed. We have shown that programmable cells of monocytic origin (PCMO) respond to I/R with the release of angiogenic mediators and that transplantation of PCMO results in increased neovascularization. Human regulatory macrophages (Mreg), which are also of monocytic origin, have been successfully employed in clinical transplantation studies due to their immunomodulatory properties. Here, we investigated whether Mreg also possess angiogenic potential in vitro and could represent a treatment option for I/R-associated illnesses. Mreg were differentiated using peripheral blood monocytes from different donors (N = 14) by incubation with M-CSF and human AB serum and stimulation with INF-gamma. Mreg cultures were subjected to 3 h of hypoxia and 24 h of reoxygenation (resembling I/R) or the respective nonischemic control. Cellular resilience, expression of pluripotency markers, secretion of angiogenic proteins, and influence on endothelial tube formation as a surrogate marker for angiogenesis were investigated. Mreg showed resilience against I/R that did not lead to increased cell damage. Mreg express DHRS9 as well as IDO and display a moderate to low expression pattern of several pluripotency genes (e.g., NANOG, OCT-4, and SOX2). I/R resulted in an upregulation of IDO (p < 0.001) while C-MYC and KLF4 were downregulated (p < 0.001 and p < 0.05). Proteome profiling revealed the secretion of numerous angiogenic proteins by Mreg of which several were strongly upregulated by I/R (e.g., MIP-1alpha, 19.9-fold; GM-CSF, 19.2-fold; PTX3, 5.8-fold; IL-1β, 5.2-fold; and MCP-1, 4.7-fold). The angiogenic potential of supernatants from Mreg subjected to I/R remains inconclusive. While Mreg supernatants from 3 donors induced tube formation, 2 supernatants were not effective. We suggest that Mreg may prove beneficial as a cell therapy-based treatment option for I/R-associated illnesses. However, donor characteristics seem to crucially influence the effectiveness of Mreg treatment.
Collapse
Affiliation(s)
- Lars Hummitzsch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Rene Rusch
- Department of Cardiovascular Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Jochen Cremer
- Department of Cardiovascular Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Markus Steinfath
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Justus Gross
- Clinic for Vascular Surgery, Bad Segeberg, Germany
| | - Fred Fandrich
- Department of Applied Cell Therapy, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Rouven Berndt
- Department of Cardiovascular Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
25
|
Karpov AA, Puzanov MV, Ivkin DY, Krasnova MV, Anikin NA, Docshin PM, Moiseeva OM, Galagudza MM. Non-inferiority of microencapsulated mesenchymal stem cells to free cells in cardiac repair after myocardial infarction: A rationale for using paracrine factor(s) instead of cells. Int J Exp Pathol 2019; 100:102-113. [PMID: 31017330 DOI: 10.1111/iep.12312] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 02/06/2019] [Accepted: 02/24/2019] [Indexed: 12/30/2022] Open
Abstract
A major translational barrier to the use of stem cell (SC)-based therapy in patients with myocardial infarction (MI) is the lack of a clear understanding of the mechanism(s) underlying the cardioprotective effect of SCs. Numerous paracrine factors from SCs may account for reduction in infarct size, but myocardial salvage associated with transdifferentiation of SCs into vascular cells as well as cardiomyocyte-like cells may be involved too. In this study, bone marrow-derived rat mesenchymal SC (MSCs) were microencapsulated in alginate preventing viable cell release while supporting their secretory phenotype. The hypothesis on the key role of paracrine factors from MSCs in their cardioprotective activity was tested by comparison of the effect of encapsulated vs free MSCs in the rat model of MI. Intramyocardial administration of both free and encapsulated MSCs after MI caused reduction in scar size (12.1 ± 6.83 and 14.7 ± 4.26%, respectively, vs 21.7 ± 6.88% in controls, P = 0.015 and P = 0.03 respectively). Scar size was not different in animals treated with free and encapsulated MSC (P = 0.637). These data provide evidence that MSC-derived growth factors and cytokines are crucial for cardioprotection elicited by MSC. Administration of either free or encapsulated MSCs was not arrhythmogenic in non-infarcted rats. The consistency of our data with the results of other studies on the major role of MSC secretome components in cardiac protection further support the theory that the use of live, though encapsulated, cells for MI therapy may be replaced with heart-targeted-sustained delivery of growth factors/cytokines.
Collapse
Affiliation(s)
- Andrey A Karpov
- Almazov National Medical Research Centre, St-Petersburg, Russia
| | - Maxim V Puzanov
- Almazov National Medical Research Centre, St-Petersburg, Russia
| | - Dmitry Yu Ivkin
- Saint Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, Russia
| | - Marina V Krasnova
- Saint Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, Russia
| | - Nikita A Anikin
- Department of Pathophysiology, First Pavlov State Medical, University of Saint Petersburg, St. Petersburg, Russia
| | - Pavel M Docshin
- Almazov National Medical Research Centre, St-Petersburg, Russia
| | - Olga M Moiseeva
- Almazov National Medical Research Centre, St-Petersburg, Russia
| | | |
Collapse
|
26
|
Wang P, Zhang C, Li J, Luo L, Zhang S, Dong F, Tang Z, Ni S. Adipose-derived mesenchymal stromal cells improve hemodynamic function in pulmonary arterial hypertension: identification of microRNAs implicated in modulating endothelial function. Cytotherapy 2019; 21:416-427. [PMID: 30904330 DOI: 10.1016/j.jcyt.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 11/18/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial hyperproliferation and dysfunction. Restoration of endothelial function is a common goal of available treatments. In the present study, human adipose-derived mesenchymal stromal cells (ASCs) were co-cultured with monocrotaline pyrrole-treated human pulmonary arterial endothelial cells (HPAECs); increased proliferation of HPAECs and expression of vascular endothelial growth factor (VEGF) were observed. High throughput sequencing results showed that six microRNAs (miMNAs) of ASCs were significantly dysregulated. In monocrotaline-induced PAH rat models, ASC transplantation improved the right ventricle systolic pressure, right ventricle hypertrophy and pulmonary endothelium hyperproliferation, and four of the six miRNAs were validated in the lung tissue samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that these dysregulated miRNAs were involved in the regulation of transcription, signal transduction, negative regulation of cell proliferation through mitogen-activated protein kinase (MAPK) signaling pathway, Wnt signaling pathway, VEGF signaling pathway, cytokine-cytokine receptor interaction, regulation of actin cytoskeleton, transforming growth factor (TGF)-beta signaling pathway and P53 signaling pathway. Our data indicates that the unique six miRNA expression signature could be involved in the PAH endothelial repair by ASCs.
Collapse
Affiliation(s)
- Pengbo Wang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Caixin Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jun Li
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lin Luo
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shuwen Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fulu Dong
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Zhiyuan Tang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China.
| | - Songshi Ni
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
27
|
Functionally Improved Mesenchymal Stem Cells to Better Treat Myocardial Infarction. Stem Cells Int 2018; 2018:7045245. [PMID: 30622568 PMCID: PMC6286742 DOI: 10.1155/2018/7045245] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/10/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death worldwide. Mesenchymal stem cell (MSC) transplantation is considered a promising approach and has made significant progress in preclinical studies and clinical trials for treating MI. However, hurdles including poor survival, retention, homing, and differentiation capacity largely limit the therapeutic effect of transplanted MSCs. Many strategies such as preconditioning, genetic modification, cotransplantation with bioactive factors, and tissue engineering were developed to improve the survival and function of MSCs. On the other hand, optimizing the hostile transplantation microenvironment of the host myocardium is also of importance. Here, we review the modifications of MSCs as well as the host myocardium to improve the efficacy of MSC-based therapy against MI.
Collapse
|
28
|
Specific Cell (Re-)Programming: Approaches and Perspectives. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:71-115. [PMID: 29071403 DOI: 10.1007/10_2017_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Many disorders are manifested by dysfunction of key cell types or their disturbed integration in complex organs. Thereby, adult organ systems often bear restricted self-renewal potential and are incapable of achieving functional regeneration. This underlies the need for novel strategies in the field of cell (re-)programming-based regenerative medicine as well as for drug development in vitro. The regenerative field has been hampered by restricted availability of adult stem cells and the potentially hazardous features of pluripotent embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Moreover, ethical concerns and legal restrictions regarding the generation and use of ESCs still exist. The establishment of direct reprogramming protocols for various therapeutically valuable somatic cell types has overcome some of these limitations. Meanwhile, new perspectives for safe and efficient generation of different specified somatic cell types have emerged from numerous approaches relying on exogenous expression of lineage-specific transcription factors, coding and noncoding RNAs, and chemical compounds.It should be of highest priority to develop protocols for the production of mature and physiologically functional cells with properties ideally matching those of their endogenous counterparts. Their availability can bring together basic research, drug screening, safety testing, and ultimately clinical trials. Here, we highlight the remarkable successes in cellular (re-)programming, which have greatly advanced the field of regenerative medicine in recent years. In particular, we review recent progress on the generation of cardiomyocyte subtypes, with a focus on cardiac pacemaker cells. Graphical Abstract.
Collapse
|
29
|
Tan QW, Tang SL, Zhang Y, Yang JQ, Wang ZL, Xie HQ, Lv Q. Hydrogel from Acellular Porcine Adipose Tissue Accelerates Wound Healing by Inducing Intradermal Adipocyte Regeneration. J Invest Dermatol 2018; 139:455-463. [PMID: 30195900 DOI: 10.1016/j.jid.2018.08.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/03/2018] [Accepted: 08/09/2018] [Indexed: 02/09/2023]
Abstract
As an important component of the skin, intradermal adipocytes are closely associated with skin homeostasis and wound healing. Although studies have focused on the role of fibroblasts, keratinocytes, and inflammatory cells in wound healing, the role of adipocytes has not been fully investigated. Here, we verified whether the induction of adipocyte regeneration in a wound bed can effectively promote wound healing, finding that the hydrogel from acellular porcine adipose tissue in combination with adipose-derived stem cells can induce in situ adipogenesis in the wound microenvironment. The newly regenerated adipocytes enhanced fibroblast migration, accelerated wound closing, and enhanced wound epithelialization. More importantly, newly formed intact skin structure was observed after treating the wound with adipose-derived stem cell-loaded hydrogel from acellular porcine adipose tissue. These results show that hydrogel from acellular porcine adipose tissue might substantially improve re-epithelialization, angiogenesis, and skin-appendage regeneration, making it a promising therapeutic biomaterial for skin wound healing.
Collapse
Affiliation(s)
- Qiu-Wen Tan
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China; Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Shen-Li Tang
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China
| | - Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Ji-Qiao Yang
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China
| | - Zhu-Le Wang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China.
| | - Qing Lv
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China.
| |
Collapse
|
30
|
Ward MR, Abadeh A, Connelly KA. Concise Review: Rational Use of Mesenchymal Stem Cells in the Treatment of Ischemic Heart Disease. Stem Cells Transl Med 2018; 7:543-550. [PMID: 29665255 PMCID: PMC6052612 DOI: 10.1002/sctm.17-0210] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/22/2018] [Indexed: 12/20/2022] Open
Abstract
The capacity of stem and progenitor cells to stimulate cardiac regeneration has been studied for almost 20 years, with very promising preclinical data and mixed clinical results. Several cell types have been studied, identified by their cell surface markers, differentiation capacity and their secreted growth factors. Bone marrow derived mesenchymal stem cells (MSCs) have been found to have potent regenerative capacity, through multiple mechanisms, including mesoderm lineage differentiation, immunomodulation, and paracrine stimulation. MSCs also secrete exosomes and microvesicles, which themselves contain potent angiogenic cytokines or mRNA molecules with effects on their local milieu. This concise review summarizes the mechanisms of MSC-based cardiac regeneration and highlighting results from molecular and preclinical studies. We also discuss clinical trial results to date, and ongoing studies. Furthermore, we discuss novel approaches for the enhancement of MSC based cardiac regeneration, such as genetic modification. Stem Cells Translational Medicine 2018;7:543-550.
Collapse
Affiliation(s)
- Michael R Ward
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Armin Abadeh
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Zhou H, Tan W, Qiu Z, Song Y, Gao S. A bibliometric analysis in gene research of myocardial infarction from 2001 to 2015. PeerJ 2018; 6:e4354. [PMID: 29456889 PMCID: PMC5813587 DOI: 10.7717/peerj.4354] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/20/2018] [Indexed: 12/23/2022] Open
Abstract
Objectives We aimed to evaluate the global scientific output of gene research of myocardial infarction and explore their hotspots and frontiers from 2001 to 2015, using bibliometric methods. Methods Articles about the gene research of myocardial infarction between 2001 and 2015 were retrieved from the Web of Science Core Collection (WoSCC). We used the bibliometric method and Citespace V to analyze publication years, journals, countries, institutions, research areas, authors, research hotspots, and trends. We plotted the reference co-citation network, and we used key words to analyze the research hotspots and trends. Results We identified 1,853 publications on gene research of myocardial research from 2001 to 2015, and the annual publication number increased with time. Circulation published the highest number of articles. United States ranked highest in the countries with most publications, and the leading institute was Harvard University. Relevant publications were mainly in the field of Cardiovascular system cardiology. Keywords and references analysis indicated that gene expression, microRNA and young women were the research hotspots, whereas stem cell, chemokine, inflammation and cardiac repair were the frontiers. Conclusions We depicted gene research of myocardial infarction overall by bibliometric analysis. Mesenchymal stem cells Therapy, MSCs-derived microRNA and genetic modified MSCs are the latest research frontiers. Related studies may pioneer the future direction of this filed in next few years. Further studies are needed.
Collapse
Affiliation(s)
- Huaqiang Zhou
- Department of Anesthesia, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wulin Tan
- Department of Anesthesia, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zeting Qiu
- Department of Anesthesia, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiyan Song
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shaowei Gao
- Department of Anesthesia, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Abstract
During the past decades, stem cell-based therapy has acquired a promising role in regenerative medicine. The application of novel cell therapeutics for the treatment of cardiovascular diseases could potentially achieve the ambitious aim of effective cardiac regeneration. Despite the highly positive results from preclinical studies, data from phase I/II clinical trials are inconsistent and the improvement of cardiac remodeling and heart performance was found to be quite limited. The major issues which cardiac stem cell therapy is facing include inefficient cell delivery to the site of injury, accompanied by low cell retention and weak effectiveness of remaining stem cells in tissue regeneration. According to preclinical and clinical studies, various stem cells (adult stem cells, embryonic stem cells, and induced pluripotent stem cells) represent the most promising cell types so far. Beside the selection of the appropriate cell type, researchers have developed several strategies to produce “second-generation” stem cell products with improved regenerative capacity. Genetic and nongenetic modifications, chemical and physical preconditioning, and the application of biomaterials were found to significantly enhance the regenerative capacity of transplanted stem cells. In this review, we will give an overview of the recent developments in stem cell engineering with the goal to facilitate stem cell delivery and to promote their cardiac regenerative activity.
Collapse
|
33
|
Hematti P. Role of Extracellular Matrix in Cardiac Cellular Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1098:173-188. [PMID: 30238371 DOI: 10.1007/978-3-319-97421-7_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) is an essential regulator of homeostasis at the cellular, tissue, and organ level. It is now very well known that ECM dynamic remodeling is indispensable not only for normal growth and development but also recovery from tissue injuries. Indeed, abnormal remodeling of the ECM plays a major role in many pathophysiological processes and contributes to many different pathologies including cardiovascular disorders. Recently, cellular therapies have emerged as a potential therapeutic strategy for restoration of lost cardiomyocytes or their rejuvenation after cardiac damage and injuries. Harnessing the biological properties of ECM could be a viable strategy to enhance the therapeutic effects of cellular therapies by improving the engraftment, integration, survival, and functional adaptation of newly transplanted cells in many different platforms. Conversely, transplanted cells could restore the functionality and original composition of damaged ECM by secreting and depositing new ECM or stimulating normal ECM production by cardiac tissue native cells. Although the ultimate role of cell therapy in treatment of cardiac disorders is still a matter of great debate, the potential utility of ECM in improving the therapeutic effect of transplanted cells and vice versa the potential role of cell therapy as a means to restore the structure and functionality of damaged ECM should be carefully considered in implementation of future clinical cardiovascular cell therapy trials.
Collapse
Affiliation(s)
- Peiman Hematti
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
34
|
Lee H, Woo HM, Kang BJ. Impact of collagen-alginate composition from microbead morphological properties to microencapsulated canine adipose tissue-derived mesenchymal stem cell activities. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 29:1042-1052. [PMID: 29082833 DOI: 10.1080/09205063.2017.1399002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The purpose of this study was to identify the effect of collagen-alginate composition on the size and shape of microbeads and the proliferation and osteogenic properties of microencapsulated canine adipose-derived mesenchymal stem cells (ASCs) in vitro. Canine ASCs were microencapsulated in mixtures of various collagen-alginate compositions using a vibrational technologic encapsulator. The size and shape of the resultant microbeads were measured using a light field microscope and the viability of the microencapsulated canine ASCs was evaluated using a live/dead viability/cytotoxicity kit. Proliferation and osteogenic potentials of microencapsulated canine ASCs were evaluated using an alamarBlue proliferation assay and an alkaline phosphatase assay, respectively. As the collagen ratio increased, the size and size variation of microbeads increased and the shape of microbeads became more irregular. Nonetheless, homogeneous microbeads were created with no significant difference in size and shape, in the range of 0.75% alginate mixed with 0.099% collagen solution in 1.2% alginate solution. There were no significant differences in viability of the ASCs in the various collagen-alginate compositions. Both proliferation and osteogenic properties, in vitro, increased with increasing collagen ratio. Microencapsulation of canine ASCs with appropriate collagen-alginate composition increases cell proliferation and osteogenic properties, in vitro, without significant effects on the shape and size of microbeads and cell viability. Microencapsulation with adequate collagen-alginate composition may produce injectable microbeads that could enhance the therapeutic efficacy of stem cells.
Collapse
Affiliation(s)
- Hyunkyu Lee
- a Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science , Kangwon National University , Chuncheon , Republic of Korea
| | - Heung-Myong Woo
- a Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science , Kangwon National University , Chuncheon , Republic of Korea
| | - Byung-Jae Kang
- a Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science , Kangwon National University , Chuncheon , Republic of Korea
| |
Collapse
|
35
|
Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int J Mol Sci 2017; 18:E2087. [PMID: 28974046 PMCID: PMC5666769 DOI: 10.3390/ijms18102087] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/18/2017] [Accepted: 09/28/2017] [Indexed: 12/11/2022] Open
Abstract
Cell-based therapies have the potential to revolutionize current treatments for diseases with high prevalence and related economic and social burden. Unfortunately, clinical trials have made only modest improvements in restoring normal function to degenerating tissues. This limitation is due, at least in part, to the death of transplanted cells within a few hours after transplant due to a combination of mechanical, cellular, and host factors. In particular, mechanical stress during implantation, extracellular matrix loss upon delivery, nutrient and oxygen deprivation at the recipient site, and host inflammatory response are detrimental factors limiting long-term transplanted cell survival. The beneficial effect of cell therapy for regenerative medicine ultimately depends on the number of administered cells reaching the target tissue, their viability, and their promotion of tissue regeneration. Therefore, strategies aiming at improving viable cell engraftment are crucial for regenerative medicine. Here we review the major factors that hamper successful cell engraftment and the strategies that have been studied to enhance the beneficial effects of cell therapy. Moreover, we provide a perspective on whether mesenchymal stromal cell-derived extracellular vesicle delivery, as a cell-free regenerative approach, may circumvent current cell therapy limitations.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research "Città della Speranza", corso Stati Uniti 4, Padova 35127, Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| |
Collapse
|
36
|
Lin S, Zhang Q, Shao X, Zhang T, Xue C, Shi S, Zhao D, Lin Y. IGF-1 promotes angiogenesis in endothelial cells/adipose-derived stem cells co-culture system with activation of PI3K/Akt signal pathway. Cell Prolif 2017; 50. [PMID: 28960620 DOI: 10.1111/cpr.12390] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/05/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The aim of this study was to investigate the role of insulin-like growth factor-1 (IGF-1) and crosstalk between endothelial cells (ECs) and adipose-derived stem cells (ASCs) in the process of angiogenesis. METHODS A three-dimensional collagen gel used to culture mouse ASCs and mouse ECs in vitro was established. The effects of angiogenesis after exposure to IGF-1 were observed by confocal laser scanning microscopy. Western blotting and qPCR were performed to elucidate the underlying mechanisms. RESULTS IGF-1 treatment promoted the formation of vessel-like structures and the recruitment of ASCs in the three-dimensional collagen gel. The angiogenic genes and proteins in ECs were up-regulated by IGF-1 and in co-culture. Similar changes in the genes and in the proteins were detected in ASCs after exposure to IGF-1 and co-culture. p-Akt expression levels were high in ECs and ASCs after exposure to IGF-1 and co-culture. CONCLUSIONS IGF-1 and co-culture between cells facilitate the process of angiogenesis via the PI3-kinase/Akt signalling pathway. In ECs, IGF-1 stimulates the expression of angiogenesis-related growth factors with the activation of the PI3-kinase/Akt signalling pathway. Co-cultured ECs exposed to excess VEGF-A and other angiogenesis-related growth factors para-secreted from ASCs exhibit high expression of angiogenesis-related genes and proteins. In ASCs, IGF-1 induces the recruitment and function of ASCs by up-regulating the expression of PDGFB, MMPs and α-SMA. Crosstalk with ECs further facilitates changes in ASCs.
Collapse
Affiliation(s)
- Shiyu Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoru Shao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Changyue Xue
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dan Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Improvement of therapeutic effects of mesenchymal stem cells in myocardial infarction through genetic suppression of microRNA-142. Oncotarget 2017; 8:85549-85558. [PMID: 29156740 PMCID: PMC5689630 DOI: 10.18632/oncotarget.20935] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/08/2017] [Indexed: 11/30/2022] Open
Abstract
Transplanted mesenchymal stem cells (MSCs) have been shown to contribute to myocardial repair after myocardial infarction (MI), primarily through production and secretion some growth factors and cytokines related to cell survival and regeneration. Further improvement of the therapeutic potential of MSCs appears to be an attractive strategy for MI treatment. CXC chemokine receptor (CXCR) 7 is the receptor for stromal cell-derived factor-1 (SDF-1), an important chemokine that is essential for tissue repair and angiogenesis. SDF-1/CXCR7 axis plays a critical role in the mobilization, recruitment and function of MSCs during tissue regeneration. Here, we depleted miR-142 that targets CXCR7 in MSCs cells through expression of antisense of miR-142, resulting in enhanced expression of CXCR7 in these miR-142-depleted MSCs (md-MSCs). In vitro, presence of md-MSCs reduced hypoxia-induced cardiac muscle cell apoptosis in a more pronounced manner than MSCs. In vivo, compared to transplantation of MSCs, transplantation of md-MSCs further enhanced cardiac re-vascularization and further improved cardiac functions after MI in mice. Together, our data suggest that depletion of miR-142 in MSCs may improve their therapeutic effects on MI.
Collapse
|
38
|
Gaffney L, Wrona EA, Freytes DO. Potential Synergistic Effects of Stem Cells and Extracellular Matrix Scaffolds. ACS Biomater Sci Eng 2017. [DOI: 10.1021/acsbiomaterials.7b00083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Lewis Gaffney
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Emily A. Wrona
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
39
|
Micheu MM, Dorobantu M. Fifteen years of bone marrow mononuclear cell therapy in acute myocardial infarction. World J Stem Cells 2017; 9:68-76. [PMID: 28491241 PMCID: PMC5405402 DOI: 10.4252/wjsc.v9.i4.68] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/16/2017] [Accepted: 04/19/2017] [Indexed: 02/07/2023] Open
Abstract
In spite of modern treatment, acute myocardial infarction (AMI) still carries significant morbidity and mortality worldwide. Even though standard of care therapy improves symptoms and also long-term prognosis of patients with AMI, it does not solve the critical issue, specifically the permanent damage of cardiomyocytes. As a result, a complex process occurs, namely cardiac remodeling, which leads to alterations in cardiac size, shape and function. This is what has driven the quest for unconventional therapeutic strategies aiming to regenerate the injured cardiac and vascular tissue. One of the latest breakthroughs in this regard is stem cell (SC) therapy. Based on favorable data obtained in experimental studies, therapeutic effectiveness of this innovative therapy has been investigated in clinical settings. Of various cell types used in the clinic, autologous bone marrow derived SCs were the first used to treat an AMI patient, 15 years ago. Since then, we have witnessed an increasing body of data as regards this cutting-edge therapy. Although feasibility and safety of SC transplant have been clearly proved, it’s efficacy is still under dispute. Conducted studies and meta-analysis reported conflicting results, but there is hope for conclusive answer to be provided by the largest ongoing trial designed to demonstrate whether this treatment saves lives. In the meantime, strategies to enhance the SCs regenerative potential have been applied and/or suggested, position papers and recommendations have been published. But what have we learned so far and how can we properly use the knowledge gained? This review will analytically discuss each of the above topics, summarizing the current state of knowledge in the field.
Collapse
|