1
|
Chumakova SP, Urazova OI, Shipulin VM, Andreev SL, Denisenko OA, Gladkovskaya MV, Litvinova LS, Bubenchikov MA. Role of Angiopoietic Coronary Endothelial Dysfunction in the Pathogenesis of Ischemic Cardiomyopathy. Biomedicines 2023; 11:1950. [PMID: 37509589 PMCID: PMC10377729 DOI: 10.3390/biomedicines11071950] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The angiopoietic endothelial dysfunction in ischemic cardiomyopathy (ICMP) remains unexplored. AIM The identification of the imbalance of endothelial dysfunction mediators and the number of endothelial progenitor (EPC) and desquamated (EDC) cells in patients with coronary heart disease (CHD) with and without ICMP. METHODS A total of 87 patients (47 with ICMP and 40 without ICMP) were observed. The content of EPCs (CD14+CD34+VEGFR2+) in vein blood and EDCs (CD45-CD146+) in the blood from the coronary sinus and cubital vein was determined by flow cytometry. The contents of HIF-1α and HIF-2α in vein blood as well as that of ADMA and endothelin-1 in sinus plasma and angiopoietin-2, MMP-9 and galectin-3 in both samples were assessed using ELISA, and VEGF, PDGF, SDF-1 and MCP-1 contents using immunofluorescence. RESULTS ADMA and endothelin-1 levels in the sinus blood were comparable between the patient groups; a deficiency of HIF-1α and excess of HIF-2α were detected in the vein blood of ICMP patients. The EDC content in the vein blood increased in CHD patients regardless of ICMP, and the concentrations of VEGF-A, VEGF-B, PDGF, MCP-1, angiopoietin-2, and MMP-9 were normal. In ICMP patients, vein blood was characterized by an excess of galectin-3 and sinus blood by an excess of EDCs, angiopoietin-2, MMP-9 and galectin-3. CONCLUSION ICMP is accompanied by angiopoietic endothelial dysfunction.
Collapse
Affiliation(s)
- Svetlana P Chumakova
- Pathophysiology Division, Siberian State Medical University, Tomsk 634050, Russia
- Central Research Laboratory, Siberian State Medical University, Tomsk 634050, Russia
| | - Olga I Urazova
- Pathophysiology Division, Siberian State Medical University, Tomsk 634050, Russia
- Central Research Laboratory, Siberian State Medical University, Tomsk 634050, Russia
- Department of Complex Information Security of Computer Systems, Tomsk State University of Control Systems and Radioelectronics, Tomsk 634050, Russia
| | - Vladimir M Shipulin
- Cardiovascular Surgery Unit, Cardiology Research Institute, Tomsk National Medical Research Center, Russian Academy of Sciences, Tomsk 634050, Russia
| | - Sergey L Andreev
- Cardiovascular Surgery Unit, Cardiology Research Institute, Tomsk National Medical Research Center, Russian Academy of Sciences, Tomsk 634050, Russia
| | - Olga A Denisenko
- Pathophysiology Division, Siberian State Medical University, Tomsk 634050, Russia
| | | | - Larisa S Litvinova
- Immunology and Cell Biotechnology Center, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia
| | - Mikhail A Bubenchikov
- Department of Theoretical Mechanics, National Research Tomsk State University, Tomsk 634050, Russia
| |
Collapse
|
2
|
Sui M, Li T, Lu H, Li Y, Huang J, Zhang P, Wang S, Zeng L. SOCS3 inhibits the mesenchymal stromal cell secretory factor SDF-1-mediated improvement of islet function in non-obese diabetic mice. Stem Cell Res Ther 2023; 14:172. [PMID: 37400916 DOI: 10.1186/s13287-023-03347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/18/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Islet transplantation is used therapeutically in a minority of patients with type 1 diabetes (T1D). However, successful outcomes are hampered by early islet β-cell loss caused by immune rejection and autoimmunity. Recent studies have demonstrated that mesenchymal stromal cells can enhance islet function both in vitro and in vivo by secreting ligands that activate islet G-protein coupled receptors (GPCRs). Stromal cell-derived factor 1 (SDF-1) is an MSC-secreted GPCR ligand, whereas the suppressor of cytokine signaling 3 (SOCS3) is a negative regulator of STAT3-activating cytokines. Here, we determined whether improvement in islet function mediated by exogenous SDF-1 is impaired by SOCS3 in experimental models of T1D. METHODS Isolated islets were cultured for 48 h with SDF-1. Cytokine-induced apoptosis was measured immediately. Islets from Socs3-/- mice were pre-cultured with exogenous SDF-1 and transplanted underneath the kidney capsule of C57BL/6 mice with streptozotocin-induced diabetes. Blood glucose levels were monitored for 28 days. AMD3100, an antagonist of the SDF-1 ligand CXCR4, was administered subcutaneously to islet transplanted mice to inhibit CXCR4 before and after transplantation. RESULTS SDF-1 protected islet cells from cytokine-induced apoptosis in vitro. SOCS3-knockout (KO) islets pretreated with SDF-1 were effective in reducing blood glucose in non-obese diabetic mice in vivo. We found that SDF-1 elicits localized immunosuppression in transplanted SOCS3-KO islets. Immunomodulation was observed when SOCS-KO islets were preconditioned with SDF-1. Gene expression and flow cytometric analyses revealed significantly decreased immune cell infiltration, inflammatory cytokines, and concomitant increases in FOXP3+ regulatory T cells, alternatively activated M2 macrophages, and dendritic cell phenotypes. Administration of AMD3100 impaired the SDF-1-mediated improvement in SOCS3-KO islet function and local immune suppression. CONCLUSION SDF-1 improves the function of islet grafts in autoimmune diabetes through regulation by CXCR4; however, the presence of SOCS3 reverses the protective effect of SDF-1 on islet grafts. These data reveal a molecular pathway that can elicit localized immunosuppression and delay graft destruction in transplanted islets.
Collapse
Affiliation(s)
- Mingxing Sui
- Department of Organ Transplantation, Shanghai Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Tuo Li
- Department of Endocrinology, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Hanlan Lu
- Department of Organ Transplantation, Shanghai Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yanhua Li
- Department of Organ Transplantation, Shanghai Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Juan Huang
- Department of Organ Transplantation, Shanghai Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Pei Zhang
- Department of Organ Transplantation, Shanghai Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China.
| | - Li Zeng
- Department of Organ Transplantation, Shanghai Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
3
|
CRISPR/Cas9-engineered mesenchymal stromal/stem cells and their extracellular vesicles: A new approach to overcoming cell therapy limitations. Biomed Pharmacother 2022; 156:113943. [DOI: 10.1016/j.biopha.2022.113943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
|
4
|
Mao Y, Chen Y, Li W, Wang Y, Qiu J, Fu Y, Guan J, Zhou P. Physiology-Inspired Multilayer Nanofibrous Membranes Modulating Endogenous Stem Cell Recruitment and Osteo-Differentiation for Staged Bone Regeneration. Adv Healthc Mater 2022; 11:e2201457. [PMID: 36027596 DOI: 10.1002/adhm.202201457] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/23/2022] [Indexed: 01/28/2023]
Abstract
Bone regeneration involves a cascade of sophisticated, multiple-staged cellular and molecular events, where early-phase stem cell recruitment mediated by chemokines and late-phase osteo-differentiation induced by pro-osteogenic factors play the crucial roles. Herein, enlightened by a bone physiological and regenerative mechanism, the multilayer nanofibrous membranes (PLLA@SDF-1α@MT01) consisting of PLLA/MT01 micro-sol electrospun nanofibers as intima and PLLA/PEG/SDF-1α electrospun nanofibers as adventitia are fabricated through micro-sol electrospinning and manual multi-layer stacking technologies. In vitro releasing profiles show that PLLA@SDF-1α@MT01 represents the rapid release of stromal cell-derived SDF-1α (SDF-1α) in the outer layers, while with long-term sustained release of MT01 in the inner layer. Owing to interconnected porosity like the natural bone extracellular matrix and improved hydrophilia, PLLA@SDF-1α@MT01 manifests good biocompatibility both in vitro and in vivo. Furthermore, PLLA@SDF-1α@MT01 can promote bone marrow mesenchymal stem cells (BMSCs) migration by amplifying the SDF-1α/CXCR4 axis and stimulating BMSCs osteo-differentiation via activating the MAPK pathway in vitro. PLLA@SDF-1α@MT01, with a programmed dual-delivery system, exhibits the synergetic promotion of bone regeneration and vascularization by emulating key characteristics of the staged bone repair in vivo. Overall, PLLA@SDF-1α@MT01 that mimics the endogenous cascades of bone regeneration can enrich the physiology-mimetic staged regenerative strategy and represent a promising tissue-engineered scaffold for the bone defect.
Collapse
Affiliation(s)
- Yingji Mao
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China.,Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| | - Yu Chen
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China.,Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
| | - Weifeng Li
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China
| | - Ying Wang
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China.,Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
| | - Jingjing Qiu
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| | - Yingxiao Fu
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China
| | - Jianzhong Guan
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China.,Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| | - Pinghui Zhou
- Department of Orthopedics, The First Affiliated Hospital, School of Life Science, Bengbu Medical College, Bengbu, 233030, China.,Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| |
Collapse
|
5
|
Shi H, Zhao Z, Jiang W, Zhu P, Zhou N, Huang X. A Review Into the Insights of the Role of Endothelial Progenitor Cells on Bone Biology. Front Cell Dev Biol 2022; 10:878697. [PMID: 35686054 PMCID: PMC9173585 DOI: 10.3389/fcell.2022.878697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
In addition to its important transport functions, the skeletal system is involved in complex biological activities for the regulation of blood vessels. Endothelial progenitor cells (EPCs), as stem cells of endothelial cells (ECs), possess an effective proliferative capacity and a powerful angiogenic capacity prior to their differentiation. They demonstrate synergistic effects to promote bone regeneration and vascularization more effectively by co-culturing with multiple cells. EPCs demonstrate a significant therapeutic potential for the treatment of various bone diseases by secreting a combination of growth factors, regulating cellular functions, and promoting bone regeneration. In this review, we retrospect the definition and properties of EPCs, their interaction with mesenchymal stem cells, ECs, smooth muscle cells, and immune cells in bone regeneration, vascularization, and immunity, summarizing their mechanism of action and contribution to bone biology. Additionally, we generalized their role and potential mechanisms in the treatment of various bone diseases, possibly indicating their clinical application.
Collapse
Affiliation(s)
- Henglei Shi
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Zhenchen Zhao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Weidong Jiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Peiqi Zhu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Nuo Zhou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Xuanping Huang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| |
Collapse
|
6
|
Yari A, Heidari F, Veijouye SJ, Nobakht M. Hair follicle stem cells promote cutaneous wound healing through the SDF-1α/CXCR4 axis: an animal model. J Wound Care 2021; 29:526-536. [PMID: 32924817 DOI: 10.12968/jowc.2020.29.9.526] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE An appropriate source of adult stem cells for therapeutic use is stem cells deriving from the hair follicle bulge. Following injury, ischaemic tissues produce a variety of cytokines and growth factors that are essential for tissue repair. This study sought to investigate the temporal effects of hair follicle bulge stem cells (HFSCs) on cutaneous wound healing in rats using the SDF-1α/CXCR4 axis. METHOD HFSCs obtained from rat vibrissa, labeled with DiI and then special markers, were detected using flow cytometry. The animals were divided into five groups: control (non-treated, n=18), sham (PBS, n=18), AMD (treated with AMD3100, n=18), HFSC + AMD (treated with HFSCs + AMD3100, n=18) and HFSC (treated with HFSCs, n=18). A full-thickness excisional wound model was created and DiI-labeled HFSCs were injected around the wound bed. Wound healing was recorded with digital photographs. The animals were sacrificed 3, 7 and 14 days after the surgery and were used for histological (H&E, Masson's trichrome staining) and molecular (ELISA and q-PCR) assays. RESULTS The flow cytometry results demonstrated that HFSCs were CD34-positive, nestin-positive, but Kr15-negative. The morphological analysis of the HFSC-treated wounds showed accelerated wound closure. The histological analysis of the photomicrographs exhibited more re-epithelialisation and dermal structural regeneration in the HFSC-treated wounds compared with the control group. In the HFSC + AMD group, the histological parameters improved on the same days, but showed a significant decrease compared with the HFSC group in all the days assayed. In the AMD group, there was a significant reduction in the noted parameters. qRT-PCR and ELISA showed a high expression level of SDF-1α, CXCR4 and VEGFR-2 in the HFSC-treated wounded skin tissue, but the expression of CXCR4 and VEGFR-2 showed a significant reduction in the HFSC + AMD group compared with the HFSC group. CONCLUSIONS Based on the findings of this study, HFSC transplantation affects wound closure parameters and the expression of SDF-1α and CXCR4. As the SDF-1α expression level increases in the injured area, the HFSCs contribute to wound repair through the SDF-1α/CXCR4 axis. This result is extremely valuable because it raises the possibility of wounds healed by isolating autologous HFSCs from the patient.
Collapse
Affiliation(s)
- Abazar Yari
- Department of Anatomy, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Dietary Supplements and Probiotics Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Fatemeh Heidari
- Cellular and Molecular Research Center, Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sanaz Joulai Veijouye
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maliheh Nobakht
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Infectious Diseases, Iran.,Physiology Research Center, Iran, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Babazadeh S, Nassiri SM, Siavashi V, Sahlabadi M, Hajinasrollah M, Zamani-Ahmadmahmudi M. Macrophage polarization by MSC-derived CXCL12 determines tumor growth. Cell Mol Biol Lett 2021; 26:30. [PMID: 34174813 PMCID: PMC8236206 DOI: 10.1186/s11658-021-00273-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Phenotypic and functional heterogeneity of macrophages is known to be the main reason for their ability to regulate inflammation and promote tumorigenesis. Mesenchymal stem cells (MSCs) are one of the principal cells commonly found in the tumor stromal niche, with capability of macrophage phenotypic switching. The objective of this study was to evaluate the role of C-X-C motif chemokine ligand 12 (CXCL12) produced by marrow-derived MSCs in the phenotypic and functional pattern of bone marrow-derived macrophages (BMDMs). METHODS First, the CRISPR/Cas9 system was used for the CXCL12 gene knock-out in MSCs. Then, coculture systems were used to investigate the role of MSCsCXCL12-/- and MSCsCXCL12+/+ in determination of macrophage phenotype. To further analyze the role of the MSC-derived CXCL12 niche, cocultures of 4T1 mammary tumor cells and macrophages primed with MSCsCXCL12-/- or MSCsCXCL12+/+ as well as in-vivo limiting dilution assays were performed. RESULTS Our results revealed that the expression of IL-4, IL-10, TGF-β and CD206 as M2 markers was significantly increased in macrophages co-cultured with MSCsCXCL12+/+ , whereas the expression of IL-6, TNF-α and iNOS was conversely decreased. The number and size of multicellular tumor spheroids were remarkably higher when 4T1 cells were cocultured with MSCCXCL12+/+-induced M2 macrophages. We also found that the occurrence of tumors was significantly higher in coinjection of 4T1 cells with MSCCXCL12+/+-primed macrophages. Tumor initiating cells were significantly decreased after coinjection of 4T1 cells with macrophages pretreated with MSCsCXCL12-/-. CONCLUSIONS In conclusion, our findings shed new light on the role of MSC-derived CXCL12 in macrophage phenotypic switching to M2, affecting their function in tumorigenesis.
Collapse
Affiliation(s)
- Shabnam Babazadeh
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Vahid Siavashi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohadeseh Sahlabadi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamad Zamani-Ahmadmahmudi
- Department of Clinical Science, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
8
|
Hakki SS, Bozkurt BS, Hakki EE, Karaoz E, Unlu A, Kayis SA. SDF-1 modulates periodontal ligament-Mesenchymal Stem Cells (pdl-MSCs). J Periodontal Res 2021; 56:774-781. [PMID: 33733508 DOI: 10.1111/jre.12876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND/OBJECTIVES In this in vitro study, the effects of Stromal cell-derived factor-1 (SDF-1) was evaluated on the periodontal ligament-Mesenchymal Stem Cells (pdl-MSCs) functions. MATERIAL AND METHODS Real-time cell analyzer-single plate (RTCA-SP) was employed for proliferation, and RTCA-dual purpose (DP) was utilized for pdl-MSCs migration potential treated with different SDF-1 concentrations (0, 0.1, 1, 10, 100, 200, and 400 ng/ml). Based on the dose-response findings, 10 ng/ml SDF-1 was used for further mRNA experiments. RNAs isolated at 6 and 24 h were checked using quantitative RT-PCR for mineralized tissue-associated genes including type I collagen (COL I), osteocalcin (OCN), osteopontin (OPN), and runt-related transcription factor 2 (Runx2). cRNA was synthesized for 6 h, and whole-genome array analysis was performed for over 47.000 probes. Data were subjected to quantile normalization before analysis. RESULTS Increased proliferation and migration were observed in pdl-MSCs treated with 0.1, 1, and 10 ng/ml SDF-1. Increased COL I was observed at both time points: 6 and 24 h. While there was no significant change for OCN, OPN, and Runx2 at 6 h, SDF-1 up-regulated OCN and OPN, but down-regulated Runx2 mRNA expressions at 24 h. IL-8 and ESM1 genes were differentially expressed over twofold when the pdl-MSCs were exposed to SDF-1 at whole-genome array analysis. IL-8 induction was confirmed with RT-PCR. CONCLUSION Findings of this study displayed that SDF-1 modulated pdl-MSCs which were important for periodontal regeneration, inducing migration and proliferation, and regulating extracellular matrix synthesis in favor of the formation of new attachment.
Collapse
Affiliation(s)
- Sema S Hakki
- Faculty of Dentistry, Department of Periodontology, Selcuk University, Konya, Turkey.,Research Center of Dental Faculty, Selcuk University, Konya, Turkey
| | - Buket S Bozkurt
- Research Center of Dental Faculty, Selcuk University, Konya, Turkey.,Faculty of Dentistry, Research Lab of Faculty of Dentistry, Hacettepe University, Ankara, Turkey
| | - Erdogan E Hakki
- Faculty of Agriculture, Department of Soil Science and Plant Nutrition, Molecular Genetics & Biotechnology Laboratories Konya, Selcuk University, Konya, Turkey
| | - Erdal Karaoz
- Faculty of Medicine, Department of Histology & Embryology, Istinye University, İstanbul, Turkey.,Center for Stem Cell and Tissue Engineering Research & Practice, Istinye University, İstanbul, Turkey.,Center for Regenerative Medicine and Stem Cell Manufacturing (LivMedCell), Liv Hospital, İstanbul, Turkey
| | - Ali Unlu
- Faculty of Medicine, Department of Biochemistry, Selcuk University, Konya, Turkey
| | - Seyit Ali Kayis
- Faculty of Medicine, Department of Biostatistics, Bolu Abant İzzet Baysal University, Bolu, Turkey
| |
Collapse
|
9
|
Conditioned medium-preconditioned EPCs enhanced the ability in oligovascular repair in cerebral ischemia neonatal rats. Stem Cell Res Ther 2021; 12:118. [PMID: 33579354 PMCID: PMC7881622 DOI: 10.1186/s13287-021-02157-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/11/2021] [Indexed: 11/30/2022] Open
Abstract
Background Oligovascular niche mediates interactions between cerebral endothelial cells and oligodendrocyte precursor cells (OPCs). Disruption of OPC-endothelium trophic coupling may aggravate the progress of cerebral white matter injury (WMI) because endothelial cells could not provide sufficient support under diseased conditions. Endothelial progenitor cells (EPCs) have been reported to ameliorate WMI in the adult brain by boosting oligovascular remodeling. It is necessary to clarify the role of the conditioned medium from hypoxic endothelial cells preconditioned EPCs (EC-pEPCs) in WMI since EPCs usually were recruited and play important roles under blood-brain barrier disruption. Here, we investigated the effects of EC-pEPCs on oligovascular remodeling in a neonatal rat model of WMI. Methods In vitro, OPC apoptosis induced by the conditioned medium from oxygen-glucose deprivation-injured brain microvascular endothelial cells (OGD-EC-CM) was analyzed by TUNEL and FACS. The effects of EPCs on EC damage and the expression of cytomokine C-X-C motif ligand 12 (CXCL12) were examined by western blot and FACS. The effect of the CM from EC-pEPCs against OPC apoptosis was also verified by western blot and silencing RNA. In vivo, P3 rat pups were subjected to right common carotid artery ligation and hypoxia and treated with EPCs or EC-pEPCs at P7, and then angiogenesis and myelination together with cognitive outcome were evaluated at the 6th week. Results In vitro, EPCs enhanced endothelial function and decreased OPC apoptosis. Meanwhile, it was confirmed that OGD-EC-CM induced an increase of CXCL12 in EPCs, and CXCL12-CXCR4 axis is a key signaling since CXCR4 knockdown alleviated the anti-apoptosis effect of EPCs on OPCs. In vivo, the number of EPCs and CXCL12 protein level markedly increased in the WMI rats. Compared to the EPCs, EC-pEPCs significantly decreased OPC apoptosis, increased vascular density and myelination in the corpus callosum, and improved learning and memory deficits in the neonatal rat WMI model. Conclusions EC-pEPCs more effectively promote oligovascular remodeling and myelination via CXCL12-CXCR4 axis in the neonatal rat WMI model. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02157-4.
Collapse
|
10
|
Cun Y, Diao B, Zhang Z, Wang G, Yu J, Ma L, Rao Z. Role of the stromal cell derived factor-1 in the biological functions of endothelial progenitor cells and its underlying mechanisms. Exp Ther Med 2020; 21:39. [PMID: 33273969 PMCID: PMC7706408 DOI: 10.3892/etm.2020.9471] [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: 01/17/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Stromal cell derived factor-1 (SDF-1) is a chemokine that plays a critical role in the homing of stem and progenitor cells, including endothelial progenitor cells (EPCs). However, little research has been undertaken to evaluate the roles of SDF-1 in the biological functions of EPCs and related signaling pathways. The present study aimed to investigate the biological functions of EPCs in response to SDF-1, as well as the underlying mechanisms. The effects of SDF-1 treatment on EPC proliferation, migration and tube formation were assessed by performing MTS, Transwell and in vitro tube formation assays, respectively. The phosphorylation status of Akt and ERK was evaluated by western blotting. The present results indicated that SDF-1 treatment enhanced EPC proliferation, migration and tube formation compared with the control group. Furthermore, SDF-1-induced EPC proliferation was significantly reduced following treatment with a C-X-C Motif Chemokine Receptor 4 antagonist (AMD3100), a PI3K inhibitor (LY294002) and the mitogen-activated protein kinase kinase inhibitor (MEK; PD98059). SDF-1-induced migration and angiogenesis were significantly suppressed by the PI3K inhibitor, but not the MEK inhibitor. Moreover, SDF-1 significantly increased the protein expression levels of phosphorylated (p)-Akt and p-ERK; however, SDF-1-induced effects on protein expression were suppressed by AMD3100, LY294002 and PD98059. Thus, SDF-1-induced EPC proliferation was mediated by activation of the Akt and ERK signaling pathways, whereas SDF-1-mediated EPC migration and tube formation only involved activation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Yanping Cun
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Bo Diao
- Department of Clinical Experiment, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Zhimin Zhang
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Gang Wang
- Department of Clinical Experiment, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Jing Yu
- Department of Clinical Experiment, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Lianting Ma
- Department of Neurosurgery, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Zhiguo Rao
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
11
|
Therapeutic Potential of Endothelial Colony-Forming Cells in Ischemic Disease: Strategies to Improve their Regenerative Efficacy. Int J Mol Sci 2020; 21:ijms21197406. [PMID: 33036489 PMCID: PMC7582994 DOI: 10.3390/ijms21197406] [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: 09/22/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) comprises a range of major clinical cardiac and circulatory diseases, which produce immense health and economic burdens worldwide. Currently, vascular regenerative surgery represents the most employed therapeutic option to treat ischemic disorders, even though not all the patients are amenable to surgical revascularization. Therefore, more efficient therapeutic approaches are urgently required to promote neovascularization. Therapeutic angiogenesis represents an emerging strategy that aims at reconstructing the damaged vascular network by stimulating local angiogenesis and/or promoting de novo blood vessel formation according to a process known as vasculogenesis. In turn, circulating endothelial colony-forming cells (ECFCs) represent truly endothelial precursors, which display high clonogenic potential and have the documented ability to originate de novo blood vessels in vivo. Therefore, ECFCs are regarded as the most promising cellular candidate to promote therapeutic angiogenesis in patients suffering from CVD. The current briefly summarizes the available information about the origin and characterization of ECFCs and then widely illustrates the preclinical studies that assessed their regenerative efficacy in a variety of ischemic disorders, including acute myocardial infarction, peripheral artery disease, ischemic brain disease, and retinopathy. Then, we describe the most common pharmacological, genetic, and epigenetic strategies employed to enhance the vasoreparative potential of autologous ECFCs by manipulating crucial pro-angiogenic signaling pathways, e.g., extracellular-signal regulated kinase/Akt, phosphoinositide 3-kinase, and Ca2+ signaling. We conclude by discussing the possibility of targeting circulating ECFCs to rescue their dysfunctional phenotype and promote neovascularization in the presence of CVD.
Collapse
|
12
|
Siavashi V, Cheraghi H, Pourmohammad P, Nooshirvani P, Abdolahi S, Solghani A, Nassiri SM, Ghazanfari T. Impairment of endothelial progenitor cells function in patient with mustard gas intoxication. Inhal Toxicol 2020; 32:131-140. [PMID: 32312128 DOI: 10.1080/08958378.2020.1755396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Sulfur mustard (SM), also known as mustard gas, was first widely used in the Iraq-Iran. After SM exposure, the most prominent clinical signs are the development of extensive non-healing skin wounds and pulmonary signs, persisting over long time. Since the most frequent complications in SM-intoxicated patients are respiratory and dermatologic lesions, and with respect to the important role of endothelial progenitor cells (EPCs) in the pathophysiology of these lesion, we conducted this study to recognize the potential effects of SM on biological features of EPCs in patients exposed with this gas.Methods: In this study, 30 patients with the history of SM exposure during the Iran-Iraq war (1984-1988), 27 patients with pulmonary signs with no history of SM exposure and 20 healthy participants were included. Cell population and function of EPCs were assessed 4 years post-exposure. For this purpose, circulating EPCs (cEPCs) were harvested and cultivated, then the biological features of these cells, including migratory, proliferative, and tubulogenic activities were analyzed. We also measured serum antioxidants levels and mRNA levels of some proangiogenic factors in EPCs from SM-intoxicated patients.Results: Our results showed lesser number of cEPCs in patients exposed with SM, which was associated with decreased proliferative, migratory, and tubulogenic activity of these cells. Also, we found the lesser serum activity of SOD, GPX and MDA in the SM group than in the healthy control group.Conclusions: SM exposure resulted in decreased proliferation and migration of EPCs, which was associated with decreased tubule formation and angiogenic factors.
Collapse
Affiliation(s)
- Vahid Siavashi
- Immunoregulation Research Center, Shahed University, Tehran, Iran.,Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hadi Cheraghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Pirouz Pourmohammad
- Department of Biochemistry, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Parviz Nooshirvani
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sara Abdolahi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Amir Solghani
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
13
|
Si R, Gao C, Guo R, Lin C, Li J, Guo W. Human mesenchymal stem cells encapsulated-coacervated photoluminescent nanodots layered bioactive chitosan/collagen hydrogel matrices to indorse cardiac healing after acute myocardial infarction. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2020; 206:111789. [PMID: 32240945 DOI: 10.1016/j.jphotobiol.2020.111789] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Acute Myocardial Infarction (MI) is one of the foremost causes of human death worldwide and it leads to mass death of cardiomyocytes, interchanges of unfavorable biological environment and affecting electrical communications by fibrosis scar formations, and specifically deficiency of blood supply to heart which leads to heart damage and heart failure. Recently, numerous appropriate strategies have been applied to base on solve these problems wound be provide prominent therapeutic potential to cardiac regeneration after acute MI. In the present study, a combined biopolymeric conductive hydrogel was fabricated with conductive ultra-small graphene quantum dots as a soft injectable hydrogel for cardiac regenerations. The resultant hydrogel was combined with human Mesenchymal stem cells (hMSCs) to improved angiogenesis in cardiovascular tissues and decreasing cardiomyocyte necrosis of hydrogel treated acute-infarcted region has been greatly associated with the development of cardiac functions in MI models. The prepared graphene quantum dots and hydrogel groups was physico-chemically analyzed and confirmed the suitability of the materials for cardiac regeneration applications. The in vitro analyzes of hydrogels with hMSCs have established that enhanced cell survival rate, increased expressions of pro-inflammatory factors, pro-angiogenic factors and early cardiogenic markers. The results of in vivo myocardial observations and electrocardiography data demonstrated a favorable outcome of ejection fraction, fibrosis area, vessel density with reduced infarction size, implying that significant development of heart regenerative function after MI. This novel strategy of injectable hydrogel with hMSCs could be appropriate for the effective treatment of cardiac therapies after acute MI.
Collapse
Affiliation(s)
- Rui Si
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, People's Republic of China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, People's Republic of China
| | - Rui Guo
- Department of Physiology, Collage of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, People's Republic of China
| | - Chen Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, People's Republic of China
| | - Jiayi Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, People's Republic of China
| | - Wenyi Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, People's Republic of China..
| |
Collapse
|