1
|
Li Z, Qi J, Fu S, Luan J, Wang Q. Effects of nanographene oxide on adipose-derived stem cell cryopreservation. Cell Tissue Bank 2024; 25:805-830. [PMID: 38844606 DOI: 10.1007/s10561-024-10140-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/17/2024] [Indexed: 09/06/2024]
Abstract
Cryoinjury mitigation is key in cell cryopreservation. Here, we aimed to assess the effectiveness of nanographene oxide (nano-GO) for improving cryoprotectant agents (CPAs) in human adipose stem cell (hADSC) cryopreservation. For in vitro experiments, nano-GO (5 μg/mL) was added to the CPAs in the control, and passage (P) 2 hADSCs were collected and cryopreserved for around two weeks. We compared cytotoxicity, cell viability, immunophenotypes, proliferation, cell apoptosis, and tri-lineage differentiation. In vivo, studies used lipoaspirate to create non-enriched or hADSC-enriched fat tissues by combining it with PBS or hADSCs cryopreserved with the aforementioned CPAs. Each nude mouse received a 0.3 mL subcutaneous injection of the graft. At 12 weeks, the grafts were harvested. Histology, adipocyte-associated genes and protein, vascular density and angiogenic cytokines, macrophage infiltration, and inflammatory cytokines were analyzed. Nano-GO CPA contributed to increased cell viability, improved cell recovery, and lowered levels of early apoptosis. Nano GO at concentrations of 0.01-100 μg/mL caused no cytotoxicity to hADSCs. The absence of nano GOs in the intracellular compartments of the cells was confirmed by transmission electron microscopy. The fat grafts from the CPA-GO group showed more viable adipocytes and significantly increased angiogenesis compared to the PBS and CPA-C groups. Adding hADSCs from the CPA-GO group to the graft reduced macrophage infiltration and MCP-1 expression. Nano-GO plays an anti-apoptotic role in the cryopreservation of hADSCs, which could improve the survival of transplanted fat tissues, possibly via improved angiogenesis and lower inflammatory response in the transplanted adipose tissue.
Collapse
Affiliation(s)
- Zifei Li
- Facial and Cervical Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan, Beijing, 100144, People's Republic of China
| | - Jun Qi
- Breast Plastic and Reconstructive Surgery Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China
| | - Su Fu
- Breast Plastic and Reconstructive Surgery Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China
| | - Jie Luan
- Breast Plastic and Reconstructive Surgery Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China.
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China.
| |
Collapse
|
2
|
Mohammed RN, Aziz Sadat SA, Hassan SMA, Mohammed HF, Ramzi DO. Combinatorial Influence of Bone Marrow Aspirate Concentrate (BMAC) and Platelet-Rich Plasma (PRP) Treatment on Cutaneous Wound Healing in BALB/c Mice. J Burn Care Res 2024; 45:59-69. [PMID: 37262317 PMCID: PMC11023107 DOI: 10.1093/jbcr/irad080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Indexed: 06/03/2023]
Abstract
Bone marrow, a soft spongy tissue, is containing mesenchymal stem cells, that are well-recognized according to their self-renewability and stemness. Therefore, we hypothesized that bone marrow aspirate concentrate (BMAC) could have a pivotal influence on the process of wound healing in particular when it is combined with platelet-rich plasma (PRP). Thirty-six albino mice (BALB/c) were used in the study and they were grouped as negative-control, PRP treated, BMAC treated and BMAC plus PRP treated. An incisional wound (1 cm2) was made at the back of mouse and their wounds were treated according to their treatment plan and group allocations. Later, the skin at the treated wound sites was collected on days 7, 14, and 21 for histopathological investigation. The results showed that there was a statistically significant difference in BMAC+PRP-treated wounds over the rest of the treated groups in the acceleration of wound healing throughout the experiment by increasing the rate of wound contraction, re-epithelization process, and granulation tissue intensity with fluctuated infiltration in the number of the neutrophils, macrophages, and lymphocytes, also restoration of the epidermal and dermal thickness with less scarring and hair follicle regeneration vs to the negative-control, PRP and BMAC only treated groups. Our findings indicated that BMAC containing mesenchymal stem cells is an efficient approach, which can be used to enhance a smooth and physiopathological healing process, especially when it is used in combination with PRP.
Collapse
Affiliation(s)
- Rebar N Mohammed
- Medical Laboratory Analysis Department, College of Health Sciences, Cihan University of Sulaimaniya, Kurdistan Region, Iraq
- Department of Microbiology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Sadat Abdulla Aziz Sadat
- Department of Microbiology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Snur M A Hassan
- Department of Anatomy and Pathology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Hawraz Farhad Mohammed
- Department of Microbiology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Derin Omer Ramzi
- Department of Basic sciences, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| |
Collapse
|
3
|
Xu W, Yang Y, Li N, Hua J. Interaction between Mesenchymal Stem Cells and Immune Cells during Bone Injury Repair. Int J Mol Sci 2023; 24:14484. [PMID: 37833933 PMCID: PMC10572976 DOI: 10.3390/ijms241914484] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Fractures are the most common large organ trauma in humans. The initial inflammatory response promotes bone healing during the initial post-fracture phase, but chronic and persistent inflammation due to infection or other factors does not contribute to the healing process. The precise mechanisms by which immune cells and their cytokines are regulated in bone healing remain unclear. The use of mesenchymal stem cells (MSCs) for cellular therapy of bone injuries is a novel clinical treatment approach. Bone progenitor MSCs not only differentiate into bone, but also interact with the immune system to promote the healing process. We review in vitro and in vivo studies on the role of the immune system and bone marrow MSCs in bone healing and their interactions. A deeper understanding of this paradigm may provide clues to potential therapeutic targets in the healing process, thereby improving the reliability and safety of clinical applications of MSCs to promote bone healing.
Collapse
Affiliation(s)
| | | | - Na Li
- Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China; (W.X.); (Y.Y.)
| | - Jinlian Hua
- Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China; (W.X.); (Y.Y.)
| |
Collapse
|
4
|
Huang J, Wang YN, Zhou Y. Constitutive aryl hydrocarbon receptor facilitates the regenerative potential of mouse bone marrow mesenchymal stromal cells. World J Stem Cells 2023; 15:807-820. [PMID: 37700822 PMCID: PMC10494570 DOI: 10.4252/wjsc.v15.i8.807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Bone marrow mesenchymal stromal cells (BMSCs) are the commonly used seed cells in tissue engineering. Aryl hydrocarbon receptor (AhR) is a transcription factor involved in various cellular processes. However, the function of constitutive AhR in BMSCs remains unclear. AIM To investigate the role of AhR in the osteogenic and macrophage-modulating potential of mouse BMSCs (mBMSCs) and the underlying mechanism. METHODS Immunochemistry and immunofluorescent staining were used to observe the expression of AhR in mouse bone marrow tissue and mBMSCs. The overexpression or knockdown of AhR was achieved by lentivirus-mediated plasmid. The osteogenic potential was observed by alkaline phosphatase and alizarin red staining. The mRNA and protein levels of osteogenic markers were detected by quantitative polymerase chain reaction (qPCR) and western blot. After coculture with different mBMSCs, the cluster of differentiation (CD) 86 and CD206 expressions levels in RAW 264.7 cells were analyzed by flow cytometry. To explore the underlying molecular mechanism, the interaction of AhR with signal transducer and activator of transcription 3 (STAT3) was observed by co-immunoprecipitation and phosphorylation of STAT3 was detected by western blot. RESULTS AhR expressions in mouse bone marrow tissue and isolated mBMSCs were detected. AhR overexpression enhanced the osteogenic potential of mBMSCs while AhR knockdown suppressed it. The ratio of CD86+ RAW 264.7 cells cocultured with AhR-overexpressed mBMSCs was reduced and that of CD206+ cells was increased. AhR directly interacted with STAT3. AhR overexpression increased the phosphorylation of STAT3. After inhibition of STAT3 via stattic, the promotive effects of AhR overexpression on the osteogenic differentiation and macrophage-modulating were partially counteracted. CONCLUSION AhR plays a beneficial role in the regenerative potential of mBMSCs partially by increasing phosphorylation of STAT3.
Collapse
Affiliation(s)
- Jing Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, Hubei Province, China
| | - Yi-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, Hubei Province, China
| | - Yi Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, Hubei Province, China.
| |
Collapse
|
5
|
Preconditioned Mesenchymal Stromal Cell-Derived Extracellular Vesicles (EVs) Counteract Inflammaging. Cells 2022; 11:cells11223695. [PMID: 36429124 PMCID: PMC9688039 DOI: 10.3390/cells11223695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Inflammaging is one of the evolutionarily conserved mechanisms underlying aging and is defined as the long-term consequence of the chronic stimulation of the innate immune system. As macrophages are intimately involved in initiating and regulating the inflammatory process, their dysregulation plays major roles in inflammaging. The paracrine factors, and in particular extracellular vesicles (EVs), released by mesenchymal stromal cells (MSCs) retain immunoregulatory effects on innate and adaptive immune responses. In this paper, we demonstrate that EVs derived from MSCs preconditioned with hypoxia inflammatory cytokines exerted an anti-inflammatory role in the context of inflammaging. In this study, macrophages isolated from aged mice presented elevated pro-inflammatory factor levels already in basal conditions compared to the young counterpart, and this pre-activation status increased when cells were challenged with IFN-γ. EVs were able to attenuate the age-associated inflammation, inducing a decrease in the expression of TNF-α, iNOS, and the NADase CD38. Moreover, we demonstrate that EVs counteracted the mitochondrial dysfunction that affected the macrophages, reducing lipid peroxidation and hindering the age-associated impairment of mitochondrial complex I activity, oxygen consumption, and ATP synthesis. These results indicate that preconditioned MSC-derived EVs might be exploited as new anti-aging therapies in a variety of age-related diseases.
Collapse
|
6
|
Jung N, Park S, Kong T, Park H, Seo WM, Lee S, Kang KS. LC-MS/MS-based serum proteomics reveals a distinctive signature in a rheumatoid arthritis mouse model after treatment with mesenchymal stem cells. PLoS One 2022; 17:e0277218. [PMID: 36331907 PMCID: PMC9635733 DOI: 10.1371/journal.pone.0277218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known to be able to modulate immune responses, possess tissue-protective properties, and exhibit healing capacities with therapeutic potential for various diseases. The ability of MSCs to secrete various cytokines and growth factors provides new insights into autoimmune-diseases such as rheumatoid arthritis (RA). RA is a systemic autoimmune disease that affects the lining of synovial joints, causing stiffness, pain, inflammation, and joint erosion. In recent years, MSCs-based therapies have been widely proposed as promising therapies in the treatment of RA. However, the mechanism involved in disease-specific therapeutic effects of MSCs on RA remains unclear. To clarify the mechanism involved in effects of MSCs on RA, proteomic profiling was performed using an RA mouse model before and after treatment with MSCs. In this study, treatment efficacy of human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) was confirmed using a type II collagen-induced arthritis (CIA) mouse model. Results of measuring incidence rates of arthritis and clinical arthritis index (CAI) revealed that mice administrated with hUCB-MSCs had a significant reduction in arthritis severity. Proteins that might affect disease progression and therapeutic efficacy of hUCB-MSC were identified through LC-MS/MS analysis using serum samples. In addition, L-1000 analysis was performed for hUCB-MSC culture medium. To analysis data obtained from LC–MS/MS and L-1000, tools such as ExDEGA, MEV, and DAVID GO were used. Results showed that various factors secreted from hUCB-MSCs might play roles in therapeutic effects of MSCs on RA, with platelet activation possibly playing a pivotal role. Results of this study also suggest that SERPINE1 and THBS1 among substances secreted by hUCB-MSC might be key factors that can inhibit platelet activation. This paper is expected to improve our understanding of mechanisms involved in treatment effects of stem cells on rheumatoid arthritis.
Collapse
Affiliation(s)
- Namhee Jung
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
| | - Soyoung Park
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
| | - TaeHo Kong
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
| | - Hwanhee Park
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
| | - Woo Min Seo
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
- * E-mail: (SL); (KSK)
| | - Kyung-Sun Kang
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Geumcheon-gu, Seoul, South Korea
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
- * E-mail: (SL); (KSK)
| |
Collapse
|
7
|
Wang YH, Zhao CZ, Wang RY, Du QX, Liu JY, Pan J. The crosstalk between macrophages and bone marrow mesenchymal stem cells in bone healing. Stem Cell Res Ther 2022; 13:511. [PMID: 36333820 PMCID: PMC9636722 DOI: 10.1186/s13287-022-03199-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Bone injury plagues millions of patients worldwide every year, and it demands a heavy portion of expense from the public medical insurance system. At present, orthopedists think that autologous bone transplantation is the gold standard for treating large-scale bone defects. However, this method has significant limitations, which means that parts of patients cannot obtain a satisfactory prognosis. Therefore, a basic study on new therapeutic methods is urgently needed. The in-depth research on crosstalk between macrophages (Mϕs) and bone marrow mesenchymal stem cells (BMSCs) suggests that there is a close relationship between inflammation and regeneration. The in-depth understanding of the crosstalk between Mϕs and BMSCs is helpful to amplify the efficacy of stem cell-based treatment for bone injury. Only in the suitable inflammatory microenvironment can the damaged tissues containing stem cells obtain satisfactory healing outcomes. The excessive tissue inflammation and lack of stem cells make the transplantation of biomaterials necessary. We can expect that the crosstalk between Mϕs and BMSCs and biomaterials will become the mainstream to explore new methods for bone injury in the future. This review mainly summarizes the research on the crosstalk between Mϕs and BMSCs and also briefly describes the effects of biomaterials and aging on cell transplantation therapy.
Collapse
Affiliation(s)
- Yu-Hao Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Cheng-Zhi Zhao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Ren-Yi Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Qian-Xin Du
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Ji-Yuan Liu
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China
| | - Jian Pan
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| |
Collapse
|
8
|
Rana N, Suliman S, Mohamed-Ahmed S, Gavasso S, Gjertsen BT, Mustafa K. Systemic and local innate immune responses to surgical co-transplantation of mesenchymal stromal cells and biphasic calcium phosphate for bone regeneration. Acta Biomater 2022; 141:440-453. [PMID: 34968726 DOI: 10.1016/j.actbio.2021.12.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/03/2021] [Accepted: 12/22/2021] [Indexed: 12/23/2022]
Abstract
Bone regeneration from mesenchymal stromal cells (MSC) is attributed to comprehensive immune modulation mediated by the MSC. However, the temporal and spatial regulation of these immune responses has not yet been described. The aim of the present study was to assess the local and systemic innate immune responses to implantation of biphasic calcium phosphate biomaterial (BCP) alone, or with bone marrow derived MSC (BCP+MSC), in critical-sized calvarial bone defects of Lewis rats. Four weeks after implantation, flow cytometry analysis of innate immune cells revealed increased numbers of circulating classical monocyte-macrophages (MM) and decreased non-classical MM in the BCP+MSC group. At week 8, this differential systemic MM response was associated with an increased presence of local tissue anti-inflammatory macrophages expressing CD68 and CD163 markers (M2-like). In the BCP group without MSC, NK cells increased at weeks 1 and 4, and neutrophils increased in circulation at weeks 2 and 8. At week 8, the increase in number of neutrophils in circulation was associated with decreased local tissue neutrophils, in the BCP+MSC group. Gene expression analysis of tissue biopsies from defects implanted with BCP+MSC, in comparison to BCP alone, revealed upregulated expression of early osteogenesis genes along with macrophage differentiation-related genes at weeks 1 and 8 and neutrophil chemotaxis-related genes at week 1. This study is the first to demonstrate that surgical implantation of BCP or BCP+MSC grafts differentially regulate both systemic and local tissue innate immune responses which enhance bone formation. The results provide new insights into immune mechanisms underlying MSC-mediated bone regeneration. STATEMENT OF SIGNIFICANCE: The suitability of biphasic calcium phosphate and mesenchymal stromal cell construct (BCP+MSC) transplantation is evident from their progress in clinical trials for treating challenging maxillofacial bone defects. But less is known about the overall immune response generated by this surgical process and how it later impacts the bone formation. To this end, it is crucial to understand for both clinicians and researchers, the systemic immune response to transplanting MSC in patients for ensuring both the safety and efficacy of cell therapies. In this study, we used rat calvarial bone defect model and showed that both systemic and local innate immunes responses (monocyte-macrophages and neutrophils) are favorably directed towards enhanced bone formation in BCP+MSC implanted defects, as compared to BCP alone.
Collapse
Affiliation(s)
- Neha Rana
- Centre of Translational Oral Research (TOR) - Tissue Engineering Research Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Salwa Suliman
- Centre of Translational Oral Research (TOR) - Tissue Engineering Research Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Samih Mohamed-Ahmed
- Centre of Translational Oral Research (TOR) - Tissue Engineering Research Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Sonia Gavasso
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Norway
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Kamal Mustafa
- Centre of Translational Oral Research (TOR) - Tissue Engineering Research Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway.
| |
Collapse
|
9
|
The Crosstalk between Mesenchymal Stem Cells and Macrophages in Bone Regeneration: A Systematic Review. Stem Cells Int 2021; 2021:8835156. [PMID: 34221025 PMCID: PMC8219422 DOI: 10.1155/2021/8835156] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/28/2021] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
Bone regeneration is a complex and well-coordinated process that involves crosstalk between immune cells and resident cells in the injury site. Transplantation of mesenchymal stem cells (MSCs) is a promising strategy to enhance bone regeneration. Growing evidence suggests that macrophages have a significant impact on osteogenesis during bone regeneration. However, the precise mechanisms by which macrophage subtypes influence bone regeneration and how MSCs communicate with macrophages have not yet been fully elucidated. In this systematic literature review, we gathered evidence regarding the crosstalk between MSCs and macrophages during bone regeneration. According to the PRISMA protocol, we extracted literature from PubMed and Embase databases by using "mesenchymal stem cells" and "macrophages" and "bone regeneration" as keywords. Thirty-three studies were selected for this review. MSCs isolated from both bone marrow and adipose tissue and both primary macrophages and macrophage cell lines were used in the selected studies. In conclusion, anti-inflammatory macrophages (M2) have significantly more potential to strengthen bone regeneration compared with naïve (M0) and classically activated macrophages (M1). Transplantation of MSCs induced M1-to-M2 transition and transformed the skeletal microenvironment to facilitate bone regeneration in bone fracture and bone defect models. This review highlights the complexity between MSCs and macrophages, providing more insight into the polarized macrophage behavior in this evolving field of osteoimmunology. The results may serve as a useful reference for definite success in MSC-based therapy based on the critical interaction with macrophages.
Collapse
|
10
|
Yin X, Yang C, Wang Z, Zhang Y, Li Y, Weng J, Feng B. Alginate/chitosan modified immunomodulatory titanium implants for promoting osteogenesis in vitro and in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112087. [PMID: 33947577 DOI: 10.1016/j.msec.2021.112087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 01/08/2023]
Abstract
The essentiality of macrophages for biomaterial-mediated osteogenesis has been increasingly recognized. However, it is still unclear what is the specific role and molecular mechanisms of macrophages and material properties in the regulation of osteogenesis. As an interdisciplinary field exploring the cross-talk between immune and skeletal systems, osteoimmunology has shifted the perspective of bone substitute materials from immunosuppressive materials to immunomodulatory materials. To fabricate an immunomodulatory Ti implant, alginate/chitosan multilayer films were fabricated on the surface of titania nanotubes (TNTs) to control the release of an anti-inflammatory cytokine interleukin (IL)-4 according to our previous work. The osteogenic effects and regulation mechanisms of the immunomodulatory Ti implants were investigated in vitro in different BMSCs culture modes. Alginate/chitosan multilayer-coated samples (with or without IL-4 loading) showed better direct osteogenic ability than TNTs by promoting biomineralization and up-regulating osteogenic gene expression (BMP1α, ALP, OPN, OCN) of BMSCs. Notably, material-induced macrophage polarization, M1 and M2, enhanced early and mid-stage osteogenesis of BMSCs via distinct pathways: M1 activated both BMP6/SMADs and Wnt10b/β-catenin pathways; while M2 activated TGF-β/SMADs pathway. Material surface properties dominated in regulating late osteogenesis probably due to the surface chemical composition (alginate, chitosan and Ca2+, etc.). Due to synergistic effects of material-induced inflammatory microenvironment and material surface properties, IL-4-loaded samples exhibited superior osteogenic capability through co-activation of three signaling pathways. The in vivo studies in rat bone defect model revealed that IL-4-loaded immunomodulatory implants successfully achieved macrophage phenotypic transition from pro-inflammatory M1 to anti-inflammatory M2 and subsequently improved new bone formation.
Collapse
Affiliation(s)
- Xianzhen Yin
- Key Laboratory of Advanced Technology for Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Congling Yang
- Key Laboratory of Advanced Technology for Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; College of Chemistry and Materials Science, Sichuan Normal University, Chengdu 610068, China
| | - Ziquan Wang
- Key Laboratory of Advanced Technology for Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yan Zhang
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Yiting Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jie Weng
- Key Laboratory of Advanced Technology for Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Feng
- Key Laboratory of Advanced Technology for Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
11
|
Ismail T, Lunger A, Haumer A, Todorov A, Menzi N, Schweizer T, Bieback K, Bürgin J, Schaefer DJ, Martin I, Scherberich A. Platelet-rich plasma and stromal vascular fraction cells for the engineering of axially vascularized osteogenic grafts. J Tissue Eng Regen Med 2020; 14:1908-1917. [PMID: 33049123 DOI: 10.1002/term.3141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022]
Abstract
Avascular necrosis of bone (AVN) leads to sclerosis and collapse of bone and joints. We have previously shown that axially vascularized osteogenic constructs, engineered by combining human stromal vascular fraction (SVF) cells and a ceramic scaffold, can revitalize necrotic bone of clinically relevant size in a rat model of AVN. For a clinical translation, the fetal bovine serum (FBS) used to generate such grafts should be substituted by a nonxenogeneic culture supplement. Human thrombin-activated platelet-rich plasma (tPRP) was evaluated in this context. SVF cells were cultured inside porous hydroxyapatite scaffolds with a perfusion-based bioreactor system for 5 days. The culture medium was supplemented with either 10% FBS or 10% tPRP. The resulting constructs were inserted into devitalized bovine bone cylinders to mimic the treatment of a necrotic bone. A ligated vascular bundle was inserted into the constructs upon subcutaneous implantation in the groin of nude rats. After 1 and 8 weeks, constructs were harvested, and vascularization, host cell recruitment, and bone formation were analyzed. After 1 week in vivo, constructs were densely vascularized, with no difference between tPRP- and FBS-based ones. After 8 weeks, bone formation and vascularization was found in both tPRP- and FBS-precultured constructs. However, the amount of bone and the vessel density were respectively 2.2- and 1.8-fold higher in the tPRP group. Interestingly, the density of M2, proregenerative macrophages was also significantly higher (6.9-fold) following graft preparation with tPRP than with FBS. Our findings indicate that tPRP is a suitable substitute for FBS to generate vascularized, osteogenic grafts from SVF cells and could thus be implemented in protocols for clinical translation of this strategy towards the treatment of bone loss and AVN.
Collapse
Affiliation(s)
- Tarek Ismail
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexander Lunger
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Alexander Haumer
- Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Atanas Todorov
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nadia Menzi
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Thierry Schweizer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Karen Bieback
- Medical Faculty, University of Mannheim/Experimental Cell Therapy, University of Heidelberg, Heidelberg, Germany
| | - Joel Bürgin
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Tissue Engineering Laboratory, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
12
|
Safarova (Yantsen) Y, Olzhayev F, Umbayev B, Tsoy A, Hortelano G, Tokay T, Murata H, Russell A, Askarova S. Mesenchymal Stem Cells Coated with Synthetic Bone-Targeting Polymers Enhance Osteoporotic Bone Fracture Regeneration. Bioengineering (Basel) 2020; 7:bioengineering7040125. [PMID: 33053753 PMCID: PMC7711537 DOI: 10.3390/bioengineering7040125] [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: 08/21/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 11/16/2022] Open
Abstract
Osteoporosis is a progressive skeletal disease characterized by reduced bone density leading to bone fragility and an elevated risk of bone fractures. In osteoporotic conditions, decrease in bone density happens due to the augmented osteoclastic activity and the reduced number of osteoblast progenitor cells (mesenchymal stem cells, MSCs). We investigated a new method of cell therapy with membrane-engineered MSCs to restore the osteoblast progenitor pool and to inhibit osteoclastic activity in the fractured osteoporotic bones. The primary active sites of the polymer are the N-hydroxysuccinimide and bisphosphonate groups that allow the polymer to covalently bind to the MSCs' plasma membrane, target hydroxyapatite molecules on the bone surface and inhibit osteolysis. The therapeutic utility of the membrane-engineered MSCs was investigated in female rats with induced estrogen-dependent osteoporosis and ulnar fractures. The analysis of the bone density dynamics showed a 27.4% and 21.5% increase in bone density at 4 and 24 weeks after the osteotomy of the ulna in animals that received four transplantations of polymer-modified MSCs. The results of the intravital observations were confirmed by the post-mortem analysis of histological slices of the fracture zones. Therefore, this combined approach that involves polymer and cell transplantation shows promise and warrants further bio-safety and clinical exploration.
Collapse
Affiliation(s)
- Yuliya Safarova (Yantsen)
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.S.(Y.)); (F.O.); (B.U.); (A.T.)
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
| | - Farkhad Olzhayev
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.S.(Y.)); (F.O.); (B.U.); (A.T.)
| | - Bauyrzhan Umbayev
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.S.(Y.)); (F.O.); (B.U.); (A.T.)
| | - Andrey Tsoy
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.S.(Y.)); (F.O.); (B.U.); (A.T.)
| | - Gonzalo Hortelano
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (G.H.); (T.T.)
| | - Tursonjan Tokay
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (G.H.); (T.T.)
| | - Hironobu Murata
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (H.M.); (A.R.)
| | - Alan Russell
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (H.M.); (A.R.)
| | - Sholpan Askarova
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.S.(Y.)); (F.O.); (B.U.); (A.T.)
- Correspondence: ; Tel.: +7-7172-706514
| |
Collapse
|
13
|
Xu T, Luo Y, Wang J, Zhang N, Gu C, Li L, Qian D, Cai W, Fan J, Yin G. Exosomal miRNA-128-3p from mesenchymal stem cells of aged rats regulates osteogenesis and bone fracture healing by targeting Smad5. J Nanobiotechnology 2020; 18:47. [PMID: 32178675 PMCID: PMC7077029 DOI: 10.1186/s12951-020-00601-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) has been considered an effective therapeutic treatment for a variety of diseases including bone fracture. However, there are associated complications along with MSCs transplantation. There is evidence to show that exosomes (Exos) derived from MSCs exert a similar paracrine function. In addition, repair capabilities of MSCs decline with age. Hence, this study aims to confirm whether the Exos protective function on osteogenic differentiation and fracture healing from aged MSCs was attenuated. This information was used in order to investigate the underlying mechanism. MSCs were successfully isolated and identified from young and aged rats, and Exos were then obtained. Aged-Exos exhibited significantly attenuated effects on MSCs osteogenic differentiation in vitro and facture healing in vivo. Using miRNA array analysis, it was shown that miR-128-3p was markedly upregulated in Aged-Exos. In vitro experiments confirmed that Smad5 is a direct downstream target of miR-128-3p, and was inhibited by overexpressed miR-128-3p. A series gain- and loss- function experiment indicated that miR-128-3P serves a suppressor role in the process of fracture healing. Furthermore, effects caused by miR-128-3P mimic/inhibitor were reversed by the application of Smad5/siSmad5. Taken together, these results suggest that the therapeutic effects of MSCs-derived Exos may vary according to differential expression of miRNAs. Exosomal miR-128-3P antagomir may act as a promising therapeutic strategy for bone fracture healing, especially for the elderly.
Collapse
Affiliation(s)
- Tao Xu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yongjun Luo
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiaxing Wang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ning Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Changjiang Gu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Linwei Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Dingfei Qian
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Weihua Cai
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Jin Fan
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Guoyong Yin
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
14
|
Yun BG, Lee SH, Jeon JH, Kim SW, Jung CK, Park G, Kim SY, Jeon S, Lee MS, Park SH, Jang J, Yang HS, Cho DW, Lim JY, Kim SW. Accelerated Bone Regeneration via Three-Dimensional Cell-Printed Constructs Containing Human Nasal Turbinate-Derived Stem Cells as a Clinically Applicable Therapy. ACS Biomater Sci Eng 2019; 5:6171-6185. [DOI: 10.1021/acsbiomaterials.9b01356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Byeong Gon Yun
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Se-Hwan Lee
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
| | - Jung Ho Jeon
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seok-Won Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Gyeongsin Park
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Su Young Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sora Jeon
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Min Suk Lee
- Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 31116, Republic of Korea
| | - Sun Hwa Park
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Jinah Jang
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
| | - Hee Seok Yang
- Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungnam 31116, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
| | - Jung Yeon Lim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sung Won Kim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
15
|
Bone Marrow-Derived CD44 + Cells Migrate to Tissue-Engineered Constructs via SDF-1/CXCR4-JNK Pathway and Aid Bone Repair. Stem Cells Int 2019; 2019:1513526. [PMID: 31428156 PMCID: PMC6681616 DOI: 10.1155/2019/1513526] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/05/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
Background and Aims Host-derived cells play crucial roles in the regeneration process of tissue-engineered constructs (TECs) during the treatment of large segmental bone defects (LSBDs). However, their identity, source, and cell recruitment mechanisms remain elusive. Methods A complex model was created using mice by combining methods of GFP+ bone marrow transplantation (GFP-BMT), parabiosis (GFP+-BMT and wild-type mice), and femoral LSBD, followed by implantation of TECs or DBM scaffolds. Postoperatively, the migration of host BM cells was detected by animal imaging and immunofluorescent staining. Bone repair was evaluated by micro-CT. Signaling pathway repressors including AMD3100 and SP600125 associated with the migration of BM CD44+ cells were further investigated. In vitro, transwell migration and western-blotting assays were performed to verify the related signaling pathway. In vivo, the importance of the SDF-1/CXCR4-JNK pathway was validated by ELISA, fluorescence-activated cell sorting (FACS), immunofluorescent staining, and RT-PCR. Results First, we found that host cells recruited to facilitate TEC-mediated bone repair were derived from bone marrow and most of them express CD44, indicating the significance of CD44 in the migration of bone marrow cells towards donor MSCs. Then, the predominant roles of SDF-1/CXCR4 and downstream JNK in the migration of BM CD44+ cells towards TECs were demonstrated. Conclusion Together, we demonstrated that during bone repair promoted by TECs, BM-derived CD44+ cells were essential and their migration towards TECs could be regulated by the SDF-1/CXCR4-JNK signaling pathway.
Collapse
|
16
|
Hsieh MK, Wu CJ, Su XC, Chen YC, Tsai TT, Niu CC, Lai PL, Wu SC. Bone regeneration in Ds-Red pig calvarial defect using allogenic transplantation of EGFP-pMSCs - A comparison of host cells and seeding cells in the scaffold. PLoS One 2019; 14:e0215499. [PMID: 31318872 PMCID: PMC6638893 DOI: 10.1371/journal.pone.0215499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
Background Cells, scaffolds, and factors are the triad of regenerative engineering; however, it is difficult to distinguish whether cells in the regenerative construct are from the seeded cells or host cells via the host blood supply. We performed a novel in vivo study to transplant enhanced green fluorescent pig mesenchymal stem cells (EGFP-pMSCs) into calvarial defect of DsRed pigs. The cell distribution and proportion were distinguished by the different fluorescent colors through the whole regenerative period. Method/Results Eight adult domestic Ds-Red pigs were treated with five modalities: empty defects without scaffold (group 1); defects filled only with scaffold (group 2); defects filled with osteoinduction medium-loaded scaffold (group 3); defects filled with 5 x 103 cells/scaffold (group 4); and defects filled with 5 x 104 cells/scaffold (group 5). The in vitro cell distribution, morphology, osteogenic differentiation, and fluorescence images of groups 4 and 5 were analyzed. Two animals were sacrificed at 1, 2, 3, and 4 weeks after transplantation. The in vivo fluorescence imaging and quantification data showed that EGFP-pMSCs were represented in the scaffolds in groups 4 and 5 throughout the whole regenerative period. A higher seeded cell density resulted in more sustained seeded cells in bone regeneration compared to a lower seeded cell density. Host cells were recruited by seeded cells if enough space was available in the scaffold. Host cells in groups 1 to 3 did not change from the 1st week to 4th week, which indicates that the scaffold without seeded cells cannot recruit host cells even when enough space is available for cell ingrowth. The histological and immunohistochemical data showed that more cells were involved in osteogenesis in scaffolds with seeded cells. Conclusion Our in vivo results showed that more seeded cells recruit more host cells and that both cell types participate in osteogenesis. These results suggest that scaffolds without seeded cells may not be effective in bone transplantation.
Collapse
Affiliation(s)
- Ming-Kai Hsieh
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Jung Wu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Xuan-Chun Su
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Chen Chen
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Chien Niu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (PLL); (SCW)
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
- * E-mail: (PLL); (SCW)
| |
Collapse
|
17
|
Li T, Ma H, Ma H, Ma Z, Qiang L, Yang Z, Yang X, Zhou X, Dai K, Wang J. Mussel-Inspired Nanostructures Potentiate the Immunomodulatory Properties and Angiogenesis of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:17134-17146. [PMID: 31008578 DOI: 10.1021/acsami.8b22017] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The therapeutic effects of mesenchymal stem cells (MSCs)-material constructs mainly come from the secretion of trophic factors from MSCs, especially the immunomodulatory and angiogenic cytokines. Recent findings indicate the significance of topographical cues from these materials in modulating paracrine functions of MSCs. Here, we developed functionalized three-dimensional-printed bioceramic (BC) scaffolds with a mussel-inspired surface coating in order to regulate the paracrine function of adipose-derived MSCs (Ad-MSCs). We found that Ad-MSCs cultured on polydopamine-modified BC scaffolds (DOPA-BC) significantly produced more immunomodulatory and pro-angiogenic factors when compared with those cultured on BC scaffolds or microplates. Functional assays, such as endothelial progenitor cells migration, tube formation, and macrophage polarization, were performed to confirm the enhanced paracrine functions of the secreted trophic factors from Ad-MSCs cultured on DOPA-BC scaffolds. Further investigation identified that both focal adhesion kinase- and extracellular signal-related kinase signaling were the required mechano-transduction pathways through which the mussel-inspired surface stimulated the paracrine effect of Ad-MSCs. In a diabetic skin-defect-healing model in rats, conditioned medium received from the Ad-MSCs cultured on DOPA-BC sped wound closure, enhanced vascularization, and promoted macrophage switching from a proinflammatory M1 to a pro-healing and anti-inflammatory M2 phenotype in the wound bed. These results demonstrate that a bio-inspired coating with polydopamine represents an effective method to enhance the paracrine function of MSCs. Our findings illustrate a novel strategy to accelerate tissue regeneration by guiding the paracrine-signaling network.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Hongshi Ma
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Hongzhi Ma
- Department of Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine , Central South University , Changsha 410006 , Hunan , China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Lei Qiang
- Southwest Jiaotong University College of Medicine , No. 111, North Section, 2nd Ring Road , Chengdu 610031 , Sichuan , China
| | - Zezheng Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Xiaoxiao Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Xiaojun Zhou
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , P. R. China
| |
Collapse
|
18
|
Platelet Lysate Inhibits NF-κB Activation and Induces Proliferation and an Alert State in Quiescent Human Umbilical Vein Endothelial Cells Retaining Their Differentiation Capability. Cells 2019; 8:cells8040331. [PMID: 30970613 PMCID: PMC6523925 DOI: 10.3390/cells8040331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/24/2022] Open
Abstract
Injured blood vessel repair and blood circulation re-establishment are crucial events for tissue repair. We investigated in primary cultures of human umbilical vein endothelial cells (HUVEC), the effects of platelet lysate (PL), a cocktail of factors released by activated platelets following blood vessel disruption and involved in the wound-healing process triggering. PL exerted a protective effect on HUVEC in an inflammatory milieu by inhibiting IL-1α-activated NF-κB pathway and by inducing the secretion of PGE2, a pro-resolving molecule in the wound microenvironment. Moreover, PL enhanced HUVEC proliferation, without affecting their capability of forming tube-like structures on matrigel, and activated resting quiescent cells to re-enter cell cycle. In agreement with these findings, proliferation-related pathways Akt and ERK1/2 were activated. The expression of the cell-cycle activator Cyclin D1 was also enhanced, as well as the expression of the High Mobility Group Box-1 (HMGB1), a protein of the alarmin group involved in tissue homeostasis, repair, and remodeling. These in vitro data suggest a possible in vivo contribution of PL to new vessel formation after a wound by activation of cells resident in vessel walls. Our biochemical study provides a rationale for the clinical use of PL in the treatment of wound healing-related pathologies.
Collapse
|
19
|
Tang H, Husch JFA, Zhang Y, Jansen JA, Yang F, van den Beucken JJJP. Coculture with monocytes/macrophages modulates osteogenic differentiation of adipose-derived mesenchymal stromal cells on poly(lactic-co-glycolic) acid/polycaprolactone scaffolds. J Tissue Eng Regen Med 2019; 13:785-798. [PMID: 30771241 PMCID: PMC6594112 DOI: 10.1002/term.2826] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/17/2018] [Accepted: 02/13/2019] [Indexed: 12/18/2022]
Abstract
The effects of immune cells, in particular macrophages, on the behaviour of mesenchymal stromal cells (MSCs) have recently gained much attention for MSCs‐based tissue‐engineered constructs. This study aimed to evaluate the effect of monocytes/macrophages on the osteogenic differentiation of adipose‐derived mesenchymal stromal cells (ADMSCs) in three‐dimensional (3D) cocultures. For this, we cocultured THP‐1 monocytes, M1 macrophages, or M2 macrophages with ADMSCs on 3D poly(lactic‐co‐glycolic) acid (PLGA)/polycaprolactone (PCL) scaffolds using osteogenic medium for up to 42 days. We found that osteogenic differentiation of ADMSCs was inhibited by monocytes and both macrophage subtypes in 3D scaffolds. Furthermore, coculture of monocytes/macrophages with ADMSCs resulted in downregulated secretion of oncostatin M (OSM) and bone morphogenetic protein 2 (BMP‐2) and inhibited expression of osteogenic markers alkaline phosphatase (ALP), bone sialoprotein (BSP), and runt‐related transcription factor 2 (RUNX2). Compared with both macrophage subtypes, monocytes inhibited osteogenic differentiation of ADMSCs more significantly. These data suggest that the mutual interactions between monocytes/macrophages and ADMSCs negatively affect MSC osteogenic differentiation and thus possibly bone healing capacity, which highlights the importance of the micro‐environment in influencing cell‐based constructs to treat bone defects and the potential to improve their performance by resolving the inflammation ahead of treatment.
Collapse
Affiliation(s)
- Hongbo Tang
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Johanna F A Husch
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Yang Zhang
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - John A Jansen
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Fang Yang
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Jeroen J J P van den Beucken
- Department of Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Romaldini A, Mastrogiacomo M, Cancedda R, Descalzi F. Platelet Lysate Activates Human Subcutaneous Adipose Tissue Cells by Promoting Cell Proliferation and Their Paracrine Activity Toward Epidermal Keratinocytes. Front Bioeng Biotechnol 2018; 6:203. [PMID: 30622945 PMCID: PMC6308153 DOI: 10.3389/fbioe.2018.00203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023] Open
Abstract
Skin chronic wounds are non-healing ulcerative defects, which arise in association with a morbidity state, such as diabetes and vascular insufficiency or as the consequence of systemic factors including advanced age. Platelet Rich Plasma, a platelet-rich blood fraction, can significantly improve the healing of human skin chronic ulcers. Given that the subcutaneous adipose tissue is located beneath the skin and plays a role in the skin homeostasis, in this study, we investigated the in vitro response of human subcutaneous adipose tissue cells to platelet content in a model mimicking in vitro the in situ milieu of a deep skin injury. Considering that, at the wound site, plasma turn to serum, platelets are activated and inflammation occurs, human adipose-derived stromal cells (hASC) were cultured with Human Serum (HS) supplemented or not with Platelet Lysate (PL) and/or IL-1α. We observed that HS sustained hASC proliferation more efficiently than FBS and induced a spontaneous adipogenic differentiation in the cells. PL added to HS enhanced hASC proliferation, regardless the presence of IL-1α. In the presence of PL, hASC progressively lessened the adipogenic phenotype, possibly because the proliferation of less committed cells was induced. However, these cells resumed adipogenesis in permissive conditions. Accordingly, PL induced in quiescent cells activation of the proliferation-related pathways ERK, Akt, and STAT-3 and expression of Cyclin D1. Moreover, PL induced an early and transient increase of the pro-inflammatory response triggered by IL-1α, by inducing COX-2 expression and secretion of a large amount of PGE2, IL-6, and IL-8. Media conditioned by PL-stimulated hASC exerted a chemotactic activity on human keratinocytes and favored the healing of an in vitro scratch wound. In order to bridge the gap between in vitro results and possible in vivo events, the stimuli were also tested in ex vivo cultures of in toto human adipose tissue biopsies (hAT). PL induced cell proliferation in hAT and outgrowth of committed progenitor cells able to differentiate in permissive conditions. In conclusion, we report that the adipose tissue responds to the wound microenvironment by activating the proliferation of adipose tissue progenitor cells and promoting the release of factors favoring wound healing.
Collapse
Affiliation(s)
- Alessio Romaldini
- Department of Experimental Medicine (DIMES) and Department of Internal Medicine (DIMI), University of Genoa; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maddalena Mastrogiacomo
- Department of Experimental Medicine (DIMES) and Department of Internal Medicine (DIMI), University of Genoa; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Ranieri Cancedda
- Department of Experimental Medicine (DIMES) and Department of Internal Medicine (DIMI), University of Genoa; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fiorella Descalzi
- Department of Experimental Medicine (DIMES) and Department of Internal Medicine (DIMI), University of Genoa; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
21
|
Zhang Y, Grosfeld EC, Camargo WA, Tang H, Magri AMP, van den Beucken JJJP. Efficacy of intraoperatively prepared cell-based constructs for bone regeneration. Stem Cell Res Ther 2018; 9:283. [PMID: 30359312 PMCID: PMC6203202 DOI: 10.1186/s13287-018-1026-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/16/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Conventional cell-based bone regeneration suffers from the major disadvantage of limited cell supply, time-consuming in vitro expansion cultures, and limited patient-friendliness related to cell isolation and multiple visits to the clinic. Here, we utilized an alternative concept using "easy access cells" that can be obtained in an intraoperative manner to prepare cell-based constructs. METHODS We used stromal vascular fraction (SVF) from human adipose tissue and human monocytes for intraoperative preparation of bone constructs. Conventional constructs grafted with expanded human adipose tissue mesenchymal stem cells (ADMSCs) derived from the same donor were set as positive controls. Additionally, we combined both cell types either or not with monocytes. The cellular interaction of human SVF and ADMSCs with human monocytes was evaluated in vitro. The feasibility and bone-regenerative capacity of intraoperative constructs were determined histologically and histomorphometrically in a rat femoral condyle bone defect model. RESULTS SVF displayed equal in vitro osteogenic differentiation compared to donor-matched expanded ADMSCs, which for both was significantly enhanced upon co-culture with monocytes. Moreover, SVF and ADMSCs displayed different immunoregulatory effects on monocytes/macrophages. Upon implantation in rat femoral bone defects, SVF constructs demonstrated superior bone formation compared to ADMSC constructs and cell-free controls; no effects of monocyte addition were observed. CONCLUSION In conclusion, we here demonstrate the feasibility of intraoperative SVF construct preparation and superior bone-regenerative capacity thereof compared to donor-matched ADMSC constructs. The superiority of SVF constructs was found to be linked to the distinct differences between immunoregulatory effects of SVF and ADMSCs.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biomaterials, PO Box 9101, 6500HB Radboudumc, Nijmegen, the Netherlands
| | - Eline C Grosfeld
- Department of Biomaterials, PO Box 9101, 6500HB Radboudumc, Nijmegen, the Netherlands
| | - Winston A Camargo
- Department of Biomaterials, PO Box 9101, 6500HB Radboudumc, Nijmegen, the Netherlands
| | - Hongbo Tang
- Department of Biomaterials, PO Box 9101, 6500HB Radboudumc, Nijmegen, the Netherlands.,Department of Plastic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Angela M P Magri
- Department of Biomaterials, PO Box 9101, 6500HB Radboudumc, Nijmegen, the Netherlands.,Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | |
Collapse
|
22
|
Lo Sicco C, Reverberi D, Pascucci L, Tasso R. A Method for Isolating and Characterizing Mesenchymal Stromal Cell-derived Extracellular Vesicles. ACTA ACUST UNITED AC 2018; 46:e55. [DOI: 10.1002/cpsc.55] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Claudia Lo Sicco
- Department of Experimental Medicine, University of Genova; Genova Italy
| | - Daniele Reverberi
- U.O. Molecular Pathology, Ospedale Policlinico San Martino, IRCCS per l'Oncologia; Genova Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia; Perugia Italy
| | - Roberta Tasso
- U.O. Regenerative Medicine, Ospedale Policlinico San Martino, IRCCS per l'Oncologia; Genova Italy
| |
Collapse
|
23
|
Li T, Liu ZL, Xiao M, Yang ZZ, Peng MZ, Li CD, Zhou XJ, Wang JW. Impact of bone marrow mesenchymal stem cell immunomodulation on the osteogenic effects of laponite. Stem Cell Res Ther 2018; 9:100. [PMID: 29642953 PMCID: PMC5896058 DOI: 10.1186/s13287-018-0818-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND With the development of osteoimmunology and bone tissue engineering (BTE), it has been recognized that the immunomodulatory properties of bone biomaterials have considerable impact in determining their fate after implantation. In this regard, the polarization of macrophages secondary to biomaterials is postulated to play a crucial role in modulating their osteogenesis; thus, strategies that may facilitate this process engender increasing levels of attention. Whereas a variety of reports highlight the immunomodulation of bone marrow mesenchymal stem cells (BMSCs) in cell therapy or their osteogenesis in BTE, few have focused on the effect of BMSCs in promoting osteogenesis in BTE through regulating the phenotype of macrophages. Accordingly, there is an urgent need to clarify the immunomodulatory properties of agents such as laponite (Lap), which is comprised of bioactive silicate nanoplatelets with excellent osteogenesis-inducing potential, to enhance their use in BTE. METHODS In the present study, we analyzed the osteoimmunomodulatory properties of Lap alone, as well as following the introduction of BMSCs into Lap, to determine whether BMSCs could modulate its immunomodulatory properties and promote osteogenesis. RESULTS It was found that the BMSCs reversed the polarization of murine-derived macrophage RAW 264.7 cells from M1 as induced by pure Lap to M2 and promoted osteogenesis. In vivo study confirmed that BMSCs combined with Lap initiated a less severe immune response and had an improved effect on bone regeneration compared with Lap alone, which corresponded with the in vitro evaluation. CONCLUSION These results suggest that BMSCs could ameliorate the inflammation induced by Lap and enhance its bone formation. The immunomodulatory characteristics of BMSCs suggest that these might be tailored as a new strategy to promote the osteogenic capacity of biomaterials.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Zhong Long Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Ming Xiao
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Ze Zheng Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Ming Zheng Peng
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Cui Di Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xiao Jun Zhou
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Jin Wu Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| |
Collapse
|
24
|
Haumer A, Bourgine PE, Occhetta P, Born G, Tasso R, Martin I. Delivery of cellular factors to regulate bone healing. Adv Drug Deliv Rev 2018; 129:285-294. [PMID: 29357301 DOI: 10.1016/j.addr.2018.01.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/08/2018] [Accepted: 01/13/2018] [Indexed: 02/06/2023]
Abstract
Bone tissue has a strong intrinsic regenerative capacity, thanks to a delicate and complex interplay of cellular and molecular processes, which tightly involve the immune system. Pathological settings of anatomical, biomechanical or inflammatory nature may lead to impaired bone healing. Innovative strategies to enhance bone repair, including the delivery of osteoprogenitor cells or of potent cytokines/morphogens, indicate the potential of 'orthobiologics', but are not fully satisfactory. Here, we review different approaches based on the delivery of regenerative cues produced by cells but in cell-free, possibly off-the-shelf configurations. Such strategies exploit the paracrine effect of the secretome of mesenchymal stem/stromal cells, presented in soluble form, shuttled through extracellular vesicles, or embedded within the network of extracellular matrix molecules. In addition to osteoinductive molecules, attention is given to factors targeting the resident immune cells, to reshape inflammatory and immunity processes from scarring to regenerative patterns.
Collapse
Affiliation(s)
- Alexander Haumer
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Paul Emile Bourgine
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Paola Occhetta
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Gordian Born
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| | - Roberta Tasso
- Ospedale Policlinico San Martino-IST, IRCCS per l'Oncologia, Genova, Italy
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Switzerland.
| |
Collapse
|
25
|
Huang J, Zhou Y, Wang Y, Cai X, Wang Y. The mutual effects between macrophages and cartilage templates in the process of subcutaneous endochondral bone formation. RSC Adv 2018; 8:23679-23687. [PMID: 35540265 PMCID: PMC9081772 DOI: 10.1039/c8ra04463e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/21/2018] [Indexed: 11/30/2022] Open
Abstract
The interplay between implants and the recipient immune environment is key to the long-term effectiveness of bone tissue engineering. In this study, we aimed to investigate the mutual effects between macrophages and cartilage templates in the process of subcutaneous osteogenesis. Primary mice bone marrow derived mesenchymal stem cells (BMSCs) were seeded into gelatin sponge and chondrogenically cultured for 4 weeks in vitro to form cartilage templates. The constructs were then implanted subcutaneously in monocyte-depleted mice or normal C57BL/6 mice. Implants harvested at two months showed inferior osteogenic quality in monocyte-depleted mice compared with that of normal mice. In normal mice, the cartilage templates recruited a high ratio of alternatively activated macrophages (CAM or M2) to classically activated macrophages (AAM or M1), compared with empty sponge. In vitro co-culture assay of macrophages with cartilage templates also showed that the cartilage templates polarized macrophages to the M2 phenotype and that these effects were even stronger than those of primary BMSCs. In turn, the co-culture of polarized macrophages with cartilage templates showed that compared to M0 or M2, M1 significantly increased the expressions of osteogenic and angiogenic markers of cartilage templates. These data suggested that macrophages seem to be indispensable in the osteogenesis of cartilage templates and that cartilage templates have a favorable immunomodulatory ability to polarize macrophages to the M2 phenotype. M1 was the contributing phenotype of macrophages that promoted the osteogenesis and angiogenesis of cartilage templates. Macrophages and cartilage templates cooperate to achieve endochondral bone formation. The interplay between implants and the recipient immune environment is key to the long-term effectiveness of bone tissue engineering.![]()
Collapse
Affiliation(s)
- Jing Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST)
- Key Laboratory of Oral Biomedicine Ministry of Education
- School and Hospital of Stomatology
- Wuhan University
- Wuhan 430079
| | - Yi Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST)
- Key Laboratory of Oral Biomedicine Ministry of Education
- School and Hospital of Stomatology
- Wuhan University
- Wuhan 430079
| | - Yan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST)
- Key Laboratory of Oral Biomedicine Ministry of Education
- School and Hospital of Stomatology
- Wuhan University
- Wuhan 430079
| | - Xinjie Cai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST)
- Key Laboratory of Oral Biomedicine Ministry of Education
- School and Hospital of Stomatology
- Wuhan University
- Wuhan 430079
| | - Yining Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST)
- Key Laboratory of Oral Biomedicine Ministry of Education
- School and Hospital of Stomatology
- Wuhan University
- Wuhan 430079
| |
Collapse
|
26
|
Martín-Saavedra F, Crespo L, Escudero-Duch C, Saldaña L, Gómez-Barrena E, Vilaboa N. Substrate Microarchitecture Shapes the Paracrine Crosstalk of Stem Cells with Endothelial Cells and Osteoblasts. Sci Rep 2017; 7:15182. [PMID: 29123118 PMCID: PMC5680323 DOI: 10.1038/s41598-017-15036-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/19/2017] [Indexed: 01/08/2023] Open
Abstract
We examined the hypothesis that substrate microarchitecture regulates the crosstalk between human mesenchymal stem cells (hMSC) and cell types involved in bone regeneration. Compared with polyester flat substrates having uniformly distributed homogenous pores (2D), three-dimensional polystyrene substrates with randomly oriented and interconnected pores of heterogeneous size (3D) stimulated the stromal secretion of IGF-1 while lessened the production of VEGFR-1, MCP-1 and IL-6. The medium conditioned by hMSC cultured in 3D substrates stimulated tube formation by human endothelial cells (hEC) to a higher extent than medium from 2D cultures. 3D co-cultures of hMSC and hEC contained higher secreted levels of IGF-1, EGF and FGF-2 than 2D co-cultures, resulting in increased hEC proliferation and migration. Substrate microarchitecture influenced the secretion of factors related to bone remodeling as the ratio RANKL to OPG, and the levels of M-CSF and IL-6 were higher in 3D co-cultures of hMSC and human osteoblasts (hOB) than in 2D co-cultures. Cytokine microenvironment in 3D co-cultures stimulated osteoblast matrix reorganization while demoted the late steps of osteoblastic maturation. Altogether, data in this study may unveil a new role of scaffold microarchitecture during bone regeneration, as modulator of the paracrine relationships that hMSC establish with hEC and hOB.
Collapse
Affiliation(s)
- Francisco Martín-Saavedra
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Lara Crespo
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Clara Escudero-Duch
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Laura Saldaña
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Enrique Gómez-Barrena
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Departamento de Cirugía, Universidad Autónoma de Madrid, Calle del Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - Nuria Vilaboa
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain. .,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
27
|
Ismail T, Osinga R, Todorov A, Haumer A, Tchang LA, Epple C, Allafi N, Menzi N, Largo RD, Kaempfen A, Martin I, Schaefer DJ, Scherberich A. Engineered, axially-vascularized osteogenic grafts from human adipose-derived cells to treat avascular necrosis of bone in a rat model. Acta Biomater 2017; 63:236-245. [PMID: 28893630 DOI: 10.1016/j.actbio.2017.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/04/2017] [Accepted: 09/05/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Avascular necrosis of bone (AVN) leads to sclerosis and collapse of bone and joints. The standard of care, vascularized bone grafts, is limited by donor site morbidity and restricted availability. The aim of this study was to generate and test engineered, axially vascularized SVF cells-based bone substitutes in a rat model of AVN. METHODS SVF cells were isolated from lipoaspirates and cultured onto porous hydroxyapatite scaffolds within a perfusion-based bioreactor system for 5days. The resulting constructs were inserted into devitalized bone cylinders mimicking AVN-affected bone. A ligated vascular bundle was inserted upon subcutaneous implantation of constructs in nude rats. After 1 and 8weeks in vivo, bone formation and vascularization were analyzed. RESULTS Newly-formed bone was found in 80% of SVF-seeded scaffolds after 8weeks but not in unseeded controls. Human ALU+cells in the bone structures evidenced a direct contribution of SVF cells to bone formation. A higher density of regenerative, M2 macrophages was observed in SVF-seeded constructs. In both experimental groups, devitalized bone was revitalized by vascularized tissue after 8 weeks. CONCLUSION SVF cells-based osteogenic constructs revitalized fully necrotic bone in a challenging AVN rat model of clinically-relevant size. SVF cells contributed to accelerated initial vascularization, to bone formation and to recruitment of pro-regenerative endogenous cells. STATEMENT OF SIGNIFICANCE Avascular necrosis (AVN) of bone often requires surgical treatment with autologous bone grafts, which is surgically demanding and restricted by significant donor site morbidity and limited availability. This paper describes a de novo engineered axially-vascularized bone graft substitute and tests the potential to revitalize dead bone and provide efficient new bone formation in a rat model. The engineering of an osteogenic/vasculogenic construct of clinically-relevant size with stromal vascular fraction of human adipose, combined to an arteriovenous bundle is described. This construct revitalized and generated new bone tissue. This successful approach proposes a novel paradigm in the treatment of AVN, in which an engineered, vascularized osteogenic graft would be used as a germ to revitalize large volumes of necrotic bone.
Collapse
|
28
|
Lo Sicco C, Tasso R. Harnessing Endogenous Cellular Mechanisms for Bone Repair. Front Bioeng Biotechnol 2017; 5:52. [PMID: 28929099 PMCID: PMC5591576 DOI: 10.3389/fbioe.2017.00052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/08/2017] [Indexed: 12/24/2022] Open
Abstract
Although autologous tissue transplantation represents a valid approach for bone repair, it has encountered crucial barriers in therapeutic translation, not least the invasive process necessary for stem cell isolation. In recent years, the scientific community has made significant strides for identifying new treatment options, and great emphasis has been placed on the tight interaction between skeletal and immune system in modulating the outcome of bone repair. Within the context of specific injury environmental cues, the cross talk among inflammatory cells and tissue resident and/or circulating progenitor cells is crucial to finely coordinate repair and remodeling processes. The appropriate modulation of the inflammatory response can now be considered a new trend in the field of regenerative medicine, as it raises the attracting possibility to enhance endogenous progenitor cell functions, finally leading to tissue repair. Therefore, new treatment options have been developed considering the wide spectrum of bone–inflammation interplay, considering in particular the cell intrinsic cues responsible for the modulation of the injured environment. In this review, we will provide a panoramic overview focusing on novel findings developed to uphold endogenous bone repair.
Collapse
Affiliation(s)
- Claudia Lo Sicco
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Roberta Tasso
- Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico per l'Oncologia, Genoa, Italy
| |
Collapse
|
29
|
Mebarki M, Coquelin L, Layrolle P, Battaglia S, Tossou M, Hernigou P, Rouard H, Chevallier N. Enhanced human bone marrow mesenchymal stromal cell adhesion on scaffolds promotes cell survival and bone formation. Acta Biomater 2017. [PMID: 28636926 DOI: 10.1016/j.actbio.2017.06.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In order to induce an efficient bone formation with human bone marrow mesenchymal stromal cells (hBMSC) associated to a scaffold, it is crucial to determine the key points of the hBMSC action after in vivo transplantation as well as the appropriate features of a scaffold. To this aim we compared the hBMSC behavior when grafted onto two biomaterials allowing different bone potential in vivo. The cancellous devitalized Tutoplast®-processed bone (TPB) and the synthetic hydroxyapatite/β-tricalcium-phosphate (HA/βTCP) which give at 6weeks 100% and 50% of bone formation respectively. We first showed that hBMSC adhesion is two times favored on TPB in vitro and in vivo compared to HA/βTCP. Biomaterial structure analysis indicated that the better cell adhesion on TPB is associated to its higher and smooth open pore architecture as well as its content in collagen. Our 6week time course analysis, showed using qPCR that only adherent cells are able to survive in vivo giving thus an advantage in term of cell number on TPB during the first 4weeks after graft. We then showed that grafted hBMSC survival is crucial as cells participate directly to bone formation and play a paracrine action via the secretion of hIGF1 and hRANKL which are known to regulate the bone formation and resorption pathways respectively. Altogether our results point out the importance of developing a smooth and open pore scaffold to optimize hBMSC adhesion and ensure cell survival in vivo as it is a prerequisite to potentiate their direct and paracrine functions. STATEMENT OF SIGNIFICANCE Around 10% of skeletal fractures do not heal correctly causing nonunion. An approach involving mesenchymal stromal cells (MSC) associated with biomaterials emerges as an innovative strategy for bone repair. The diversity of scaffolds is a source of heterogeneity for bone formation efficiency. In order to better determine the characteristics of a powerful scaffold it is crucial to understand their relationship with cells after graft. Our results highlight that a biomaterial architecture similar to cancellous bone is important to promote MSC adhesion and ensure cell survival in vivo. Additionally, we demonstrated that the grafted MSC play a direct role coupled to a paracrine effect to enhance bone formation and that both of those roles are governed by the used scaffold.
Collapse
Affiliation(s)
- Miryam Mebarki
- IMRB U955-E10, INSERM, Creteil, France; Faculty of Medicine, Paris Est University, Creteil, France; Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Créteil, France
| | - Laura Coquelin
- IMRB U955-E10, INSERM, Creteil, France; Faculty of Medicine, Paris Est University, Creteil, France; Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Créteil, France
| | - Pierre Layrolle
- INSERM U957, Lab. Pathophysiology of Bone Resorption, Faculty of Medicine, University of Nantes, Nantes, France
| | - Séverine Battaglia
- INSERM U957, Lab. Pathophysiology of Bone Resorption, Faculty of Medicine, University of Nantes, Nantes, France
| | - Marine Tossou
- IMRB U955-E10, INSERM, Creteil, France; Faculty of Medicine, Paris Est University, Creteil, France; Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Créteil, France
| | - Philippe Hernigou
- IMRB U955-E10, INSERM, Creteil, France; Faculty of Medicine, Paris Est University, Creteil, France; Orthopaedic Surgery Department, Henri-Mondor AP-HP Hospital, Creteil, France
| | - Hélène Rouard
- IMRB U955-E10, INSERM, Creteil, France; Faculty of Medicine, Paris Est University, Creteil, France; Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Créteil, France
| | - Nathalie Chevallier
- IMRB U955-E10, INSERM, Creteil, France; Faculty of Medicine, Paris Est University, Creteil, France; Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Créteil, France.
| |
Collapse
|
30
|
Human adipose-derived mesenchymal stem cells seeded into a collagen-hydroxyapatite scaffold promote bone augmentation after implantation in the mouse. Sci Rep 2017; 7:7110. [PMID: 28769083 PMCID: PMC5541101 DOI: 10.1038/s41598-017-07672-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/03/2017] [Indexed: 12/29/2022] Open
Abstract
Traumatic injury or surgical excision of diseased bone tissue usually require the reconstruction of large bone defects unable to heal spontaneously, especially in older individuals. This is a big challenge requiring the development of biomaterials mimicking the bone structure and capable of inducing the right commitment of cells seeded within the scaffold. In particular, given their properties and large availability, the human adipose-derived stem cells are considered as the better candidate for autologous cell transplantation. In order to evaluate the regenerative potential of these cells along with an osteoinductive biomaterial, we have used collagen/hydroxyapatite scaffolds to test ectopic bone formation after subcutaneous implantation in mice. The process was analysed both in vivo, by Fluorescent Molecular Tomography (FMT), and ex vivo, to evaluate the formation of bone and vascular structures. The results have shown that the biomaterial could itself be able of promoting differentiation of host cells and bone formation, probably by means of its intrinsic chemical and structural properties, namely the microenvironment. However, when charged with human mesenchymal stem cells, the ectopic bone formation within the scaffold was increased. We believe that these results represent an important advancement in the field of bone physiology, as well as in regenerative medicine.
Collapse
|
31
|
Todeschi MR, El Backly RM, Varghese OP, Hilborn J, Cancedda R, Mastrogiacomo M. Host cell recruitment patterns by bone morphogenetic protein-2 releasing hyaluronic acid hydrogels in a mouse subcutaneous environment. Regen Med 2017; 12:525-539. [DOI: 10.2217/rme-2017-0023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aimed to identify host cell recruitment patterns in a mouse model in response to rhBMP-2 releasing hyaluronic acid hydrogels and influence of added nano-hydroxyapatite particles on rhBMP-2 release and pattern of bone formation. Materials & methods: Implanted gels were retrieved after implantation and cells were enzymatically dissociated for flow cytometric analysis. Percentages of macrophages, progenitor endothelial cells and putative mesenchymal stem cells were measured. Implants were evaluated for BMP-2 release by ELISA and by histology to monitor tissue formation. Results & conclusion: Hyaluronic acid+BMP-2 gels influenced the inflammatory response in the bone healing microenvironment. Host-derived putative mesenchymal stem cells were major contributors. Addition of hydroxyapatite nanoparticles modified the release pattern of rhBMP-2, resulting in enhanced bone formation.
Collapse
Affiliation(s)
- Maria R Todeschi
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
| | - Rania M El Backly
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
- Endodontics, Conservative Dentistry Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Oommen P Varghese
- Department of Chemistry, Angstrom Laboratory, Uppsala University, Uppsala, Sweden
| | - Jöns Hilborn
- Department of Chemistry, Angstrom Laboratory, Uppsala University, Uppsala, Sweden
| | - Ranieri Cancedda
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
| | - Maddalena Mastrogiacomo
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
| |
Collapse
|
32
|
Tang H, Zhang Y, Jansen JA, van den Beucken JJJP. Effect of monocytes/macrophages on the osteogenic differentiation of adipose-derived mesenchymal stromal cells in 3D co-culture spheroids. Tissue Cell 2017; 49:461-469. [PMID: 28684045 DOI: 10.1016/j.tice.2017.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 02/08/2023]
Abstract
This study aimed to investigate the distinctive roles of the monocytes and macrophages on osteogenic differentiation of adipose-derived mesenchymal stromal cells (ADMSCs) in 3D spheroid co-cultures. We hypothesized that monocytes or macrophages (subtypes pro-inflammatory M1 and pro-wound healing M2) would affect the osteogenic differentiation of ADMSCs in 3D spheroids and that cell-cell interactions between monocytes/macrophages and ADMSCs play an important role in the osteogenic differentiation process of ADMSCs. The obtained results indicated that the osteogenic differentiation of ADMSCs was inhibited by monocytes and both macrophage subtypes in 3D spheroids. Monocytes and M2 macrophages had a stronger inhibiting effect than M1 macrophages. Cell-cell interactions mediated by N-cadherin likely played a role in the inhibiting effect of monocytes/macrophages on the osteogenic differentiation of ADMSCs.
Collapse
Affiliation(s)
- Hongbo Tang
- Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands; Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Zhang
- Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | - John A Jansen
- Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | | |
Collapse
|
33
|
Cancedda R, Bollini S, Descalzi F, Mastrogiacomo M, Tasso R. Learning from Mother Nature: Innovative Tools to Boost Endogenous Repair of Critical or Difficult-to-Heal Large Tissue Defects. Front Bioeng Biotechnol 2017; 5:28. [PMID: 28503549 PMCID: PMC5408079 DOI: 10.3389/fbioe.2017.00028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/10/2017] [Indexed: 12/16/2022] Open
Abstract
For repair of chronic or difficult-to-heal tissue lesions and defects, major constraints exist to a broad application of cell therapy and tissue engineering approaches, i.e., transplantation of “ex vivo” expanded autologous stem/progenitor cells, alone or associated with carrier biomaterials. To enable a large number of patients to benefit, new strategies should be considered. One of the main goals of contemporary regenerative medicine is to develop new regenerative therapies, inspired from Mother Nature. In all injured tissues, when platelets are activated by tissue contact, their released factors promote innate immune cell migration to the wound site. Platelet-derived factors and factors secreted by migrating immune cells create an inflammatory microenvironment, in turn, causing the activation of angiogenesis and vasculogenesis processes. Eventually, repair or regeneration of the injured tissue occurs via paracrine signals activating, mobilizing or recruiting to the wound site cells with healing potential, such as stem cells, progenitors, or undifferentiated cells derived from the reprogramming of tissue differentiated cells. This review, largely based on our studies, discusses the identification of new tools, inspired by cellular and molecular mechanisms overseeing physiological tissue healing, that could reactivate dormant endogenous regeneration mechanisms lost during evolution and ontogenesis.
Collapse
Affiliation(s)
- Ranieri Cancedda
- Biorigen Srl, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Sveva Bollini
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Roberta Tasso
- IRCCS AOU San Martino-IST National Institute of Cancer Research, Genova, Italy
| |
Collapse
|
34
|
Zhang Y, Böse T, Unger RE, Jansen JA, Kirkpatrick CJ, van den Beucken JJJP. Macrophage type modulates osteogenic differentiation of adipose tissue MSCs. Cell Tissue Res 2017; 369:273-286. [PMID: 28361303 PMCID: PMC5552848 DOI: 10.1007/s00441-017-2598-8] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/24/2017] [Indexed: 12/21/2022]
Abstract
Since the reconstruction of large bone defects remains a challenge, knowledge about the biology of bone healing is desirable to develop novel strategies for improving the treatment of bone defects. In osteoimmunology, macrophages are the central component in the early stage of physiological response after bone injury and bone remodeling in the late stage. During this process, a switch of macrophage phenotype from pro-inflammatory (M1) to anti-inflammatory (M2) is observed. An appealing option for bone regeneration would be to exploit this regulatory role for the benefit of osteogenic differentiation of osteoprogenitor cells (e.g., mesenchymal stem cells; MSCs) and to eventually utilize this knowledge to improve the therapeutic outcome of bone regenerative treatment. In view of this, we focused on the in vitro interaction of different macrophage subtypes with adipose tissue MSCs to monitor the behavior (i.e. proliferation, differentiation and mineralization) of the latter in dedicated co-culture models. Our data show that co-culture of MSCs with M2 macrophages, but not with M1 macrophages or M0 macrophages, results in significantly increased MSC mineralization caused by soluble factors. Specifically, M2 macrophages promoted the proliferation and osteogenic differentiation of MSCs, while M0 and M1 macrophages solely stimulated the osteogenic differentiation of MSCs in the early and middle stages during co-culture. Secretion of the soluble factors oncostatin M (OSM) and bone morphogenetic protein 2 (BMP-2) by macrophages showed correlation with MSC gene expression levels for OSM-receptor and BMP-2, suggesting the involvement of both signaling pathways in the osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biomaterials (309), Radboudumc, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Thomas Böse
- REPAIR-lab, Institute of Pathology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ronald E Unger
- REPAIR-lab, Institute of Pathology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - John A Jansen
- Department of Biomaterials (309), Radboudumc, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | | | | |
Collapse
|
35
|
Lo Sicco C, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, Becherini P, Bosco MC, Varesio L, Franzin C, Pozzobon M, Cancedda R, Tasso R. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Mediators of Anti-Inflammatory Effects: Endorsement of Macrophage Polarization. Stem Cells Transl Med 2017; 6:1018-1028. [PMID: 28186708 PMCID: PMC5442783 DOI: 10.1002/sctm.16-0363] [Citation(s) in RCA: 398] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/31/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal Stem Cells (MSCs) are effective therapeutic agents enhancing the repair of injured tissues mostly through their paracrine activity. Increasing evidences show that besides the secretion of soluble molecules, the release of extracellular vesicles (EVs) represents an alternative mechanism adopted by MSCs. Since macrophages are essential contributors toward the resolution of inflammation, which has emerged as a finely orchestrated process, the aim of the present study was to carry out a detailed characterization of EVs released by human adipose derived-MSCs to investigate their involvement as modulators of MSC anti-inflammatory effects inducing macrophage polarization. The EV-isolation method was based on repeated ultracentrifugations of the medium conditioned by MSC exposed to normoxic or hypoxic conditions (EVNormo and EVHypo ). Both types of EVs were efficiently internalized by responding bone marrow-derived macrophages, eliciting their switch from a M1 to a M2 phenotype. In vivo, following cardiotoxin-induced skeletal muscle damage, EVNormo and EVHypo interacted with macrophages recruited during the initial inflammatory response. In injured and EV-treated muscles, a downregulation of IL6 and the early marker of innate and classical activation Nos2 were concurrent to a significant upregulation of Arg1 and Ym1, late markers of alternative activation, as well as an increased percentage of infiltrating CD206pos cells. These effects, accompanied by an accelerated expression of the myogenic markers Pax7, MyoD, and eMyhc, were even greater following EVHypo administration. Collectively, these data indicate that MSC-EVs possess effective anti-inflammatory properties, making them potential therapeutic agents more handy and safe than MSCs. Stem Cells Translational Medicine 2017 Stem Cells Translational Medicine 2017;6:1018-1028.
Collapse
Affiliation(s)
- Claudia Lo Sicco
- Department of Experimental Medicine, University of Genova, Genova, Italy
- U.O. Regenerative Medicine, IRCCS AOU San Martino-IST, National Cancer Research Institute, Genova, Italy
| | - Daniele Reverberi
- U.O. Molecular Pathology, IRCCS AOU San Martino-IST, National Cancer Research Institute, Genova, Italy
| | - Carolina Balbi
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Valentina Ulivi
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Elisa Principi
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Pamela Becherini
- Molecular Biology Laboratory, Istituto Giannina Gaslini, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Maria Carla Bosco
- Molecular Biology Laboratory, Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Molecular Biology Laboratory, Istituto Giannina Gaslini, Genova, Italy
| | - Chiara Franzin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Women and Children Health, University of Padova, Padova, Italy
| | - Ranieri Cancedda
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Roberta Tasso
- U.O. Regenerative Medicine, IRCCS AOU San Martino-IST, National Cancer Research Institute, Genova, Italy
| |
Collapse
|
36
|
Abstract
The contribution of inflammation to bone loss is well documented in arthritis and other diseases with an emphasis on how inflammatory cytokines promote osteoclastogenesis. Macrophages are the major producers of cytokines in inflammation, and the factors they produce depend upon their activation state or polarization. In recent years, it has become apparent that macrophages are also capable of interacting with osteoblasts and their mesenchymal precursors. This interaction provides growth and differentiation factors from one cell that act on the other and visa versa-a concept akin to the requirement for a feeder layer to grow hemopoietic cells or the coupling that occurs between osteoblasts and osteoclasts to maintain bone homeostasis. Alternatively, activated macrophages are the most likely candidates to promote bone formation and have also been implicated in the tissue repair process in other tissues. In bone, a number of factors, including oncostatin M, have been shown to promote osteoblast formation both in vitro and in vivo. This review discusses the different cell types involved, cellular mediators, and how this can be used to direct new bone anabolic approaches.
Collapse
|
37
|
Aktas E, Chamberlain CS, Saether EE, Duenwald-Kuehl SE, Kondratko-Mittnacht J, Stitgen M, Lee JS, Clements AE, Murphy WL, Vanderby R. Immune modulation with primed mesenchymal stem cells delivered via biodegradable scaffold to repair an Achilles tendon segmental defect. J Orthop Res 2017; 35:269-280. [PMID: 27061844 DOI: 10.1002/jor.23258] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/04/2016] [Indexed: 02/06/2023]
Abstract
Tendon healing is a complex coordinated series of events resulting in protracted recovery, limited regeneration, and scar formation. Mesenchymal stem cell (MSC) therapy has shown promise as a new technology to enhance soft tissue and bone healing. A challenge with MSC therapy involves the ability to consistently control the inflammatory response and subsequent healing. Previous studies suggest that preconditioning MSCs with inflammatory cytokines, such as IFN-γ, TNF-α, and IL-1β may accelerate cutaneous wound closure. The objective of this study was to therefore elucidate these effects in tendon. That is, the in vivo healing effects of TNF-α primed MSCs were studied using a rat Achilles segmental defect model. Rat Achilles tendons were subjected to a unilateral 3 mm segmental defect and repaired with either a PLG scaffold alone, MSC-seeded PLG scaffold, or TNF-α-primed MSC-seeded PLG scaffold. Achilles tendons were analyzed at 2 and 4 weeks post-injury. In vivo, MSCs, regardless of priming, increased IL-10 production and reduced the inflammatory factor, IL-1α. Primed MSCs reduced IL-12 production and the number of M1 macrophages, as well as increased the percent of M2 macrophages, and synthesis of the anti-inflammatory factor IL-4. Primed MSC treatment also increased the concentration of type I procollagen in the healing tissue and increased failure stress of the tendon 4 weeks post-injury. Taken together delivery of TNF-α primed MSCs via 3D PLG scaffold modulated macrophage polarization and cytokine production to further accentuate the more regenerative MSC-induced healing response. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:269-280, 2017.
Collapse
Affiliation(s)
- Erdem Aktas
- Department of Orthopedics, Ankara Oncology Research and Training Hospital, Ankara, Turkey
| | - Connie S Chamberlain
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705
| | - Erin E Saether
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Sarah E Duenwald-Kuehl
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | | | - Michael Stitgen
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Jae Sung Lee
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Anna E Clements
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Ray Vanderby
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| |
Collapse
|
38
|
Abstract
The concept of pericyte has been changing over years. This cell type was believed to possess only a function of trophic support to endothelial cells and to maintain vasculature stabilization. In the last years, the discovery of multipotent ability of perivascular populations led to the concept of vessel/wall niche. Likewise, several perivascular populations have been identified in animal and human bone marrow. In this review, we provide an overview on bone marrow perivascular population, their cross-talk with other niche components, relationship with bone marrow stromal stem cells, and similarities and differences with the perivascular population of the vessel/wall niche. Finally, we focus on the regenerative potential of these cells and the forthcoming challenges related to their use as cell therapy products.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Andrea Cordaro
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| |
Collapse
|
39
|
Kuznetsova D, Prodanets N, Rodimova S, Antonov E, Meleshina A, Timashev P, Zagaynova E. Study of the involvement of allogeneic MSCs in bone formation using the model of transgenic mice. Cell Adh Migr 2016; 11:233-244. [PMID: 27314915 DOI: 10.1080/19336918.2016.1202386] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are thought to be the most attractive type of cells for bone repair. However, much still remains unknown about MSCs and needs to be clarified before this treatment can be widely applied in the clinical practice. The purpose of this study was to establish the involvement of allogeneic MSCs in the bone formation in vivo, using a model of transgenic mice and genetically labeled cells. Polylactide scaffolds with hydroxyapatite obtained by surface selective laser sintering were used. The scaffolds were sterilized and individually seeded with MSCs from the bone marrow of 5-week-old GFP(+) transgenic C57/Bl6 or GFP(-)C57/Bl6 mice. 4-mm-diameter critical-size defects were created on the calvarial bone of mice using a dental bur. Immediately after the generation of the cranial bone defects, the scaffolds with or without seeded cells were implanted into the injury sites. The cranial bones were harvested at either 6 or 12 weeks after the implantation. GFP(+) transgenic mice having scaffolds with unlabeled MSCs were used for the observation of the host cell migration into the scaffold. GFP(-) mice having scaffolds with GFP(+)MSCs were used to assess the functioning of the seeded MSCs. The obtained data demonstrated that allogeneic MSCs were found on the scaffolds 6 and 12 weeks post-implantation. By week 12, a newly formed bone tissue from the seeded cells was observed, without an osteogenic pre-differentiation. The host cells did not appear, and the control scaffolds without seeded cells remained empty. Besides, a possibility of vessel formation from seeded MSCs was shown, without a preliminary cell cultivation under controlled conditions.
Collapse
Affiliation(s)
- Daria Kuznetsova
- a Institute of Biomedical Technologies, Nizhny Novgorod State Medical Academy , Nizhny Novgorod , Russia.,b Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod , Nizhny Novgorod , Russia
| | - Natalia Prodanets
- a Institute of Biomedical Technologies, Nizhny Novgorod State Medical Academy , Nizhny Novgorod , Russia
| | - Svetlana Rodimova
- b Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod , Nizhny Novgorod , Russia
| | - Evgeny Antonov
- c Institute of Laser and Information Technologies, Russian Academy of Sciences , Troitsk , Russia
| | - Aleksandra Meleshina
- a Institute of Biomedical Technologies, Nizhny Novgorod State Medical Academy , Nizhny Novgorod , Russia
| | - Peter Timashev
- d Sechenov First Moscow State Medical University, Research Institute for Uronephrology and Reproductive Health , Moscow , Russia
| | - Elena Zagaynova
- a Institute of Biomedical Technologies, Nizhny Novgorod State Medical Academy , Nizhny Novgorod , Russia
| |
Collapse
|
40
|
Ottoboni L, De Feo D, Merlini A, Martino G. Commonalities in immune modulation between mesenchymal stem cells (MSCs) and neural stem/precursor cells (NPCs). Immunol Lett 2015; 168:228-39. [DOI: 10.1016/j.imlet.2015.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
|
41
|
Lattanzi W, Parolisi R, Barba M, Bonfanti L. Osteogenic and Neurogenic Stem Cells in Their Own Place: Unraveling Differences and Similarities Between Niches. Front Cell Neurosci 2015; 9:455. [PMID: 26635534 PMCID: PMC4656862 DOI: 10.3389/fncel.2015.00455] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/06/2015] [Indexed: 02/06/2023] Open
Abstract
Although therapeutic use of stem cells (SCs) is already available in some tissues (cornea, blood, and skin), in most organs we are far from reaching the translational goal of regenerative medicine. In the nervous system, due to intrinsic features which make it refractory to regeneration/repair, it is very hard to obtain functionally integrated regenerative outcomes, even starting from its own SCs (the neural stem cells; NSCs). Besides NSCs, mesenchymal stem cells (MSCs) have also been proposed for therapeutic purposes in neurological diseases. Yet, direct (regenerative) and indirect (bystander) effects are often confused, as are MSCs and bone marrow-derived (stromal, osteogenic) stem cells (BMSCs), whose plasticity is actually overestimated (i.e., trans-differentiation along non-mesodermal lineages, including neural fates). In order to better understand failure in the "regenerative" use of SCs for neurological disorders, it could be helpful to understand how NSCs and BMSCs have adapted to their respective organ niches. In this perspective, here the adult osteogenic and neurogenic niches are considered and compared within their in vivo environment.
Collapse
Affiliation(s)
- Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy ; Latium Musculoskeletal Tissue Bank , Rome , Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi (NICO) , Orbassano , Italy ; Department of Veterinary Sciences, University of Turin , Turin , Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO) , Orbassano , Italy ; Department of Veterinary Sciences, University of Turin , Turin , Italy
| |
Collapse
|
42
|
Identification of a New Cell Population Constitutively Circulating in Healthy Conditions and Endowed with a Homing Ability Toward Injured Sites. Sci Rep 2015; 5:16574. [PMID: 26560420 PMCID: PMC4642305 DOI: 10.1038/srep16574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 10/06/2015] [Indexed: 12/23/2022] Open
Abstract
Stem and progenitor cells are the critical units for tissue maintenance, regeneration, and repair. The activation of regenerative events in response to tissue injury has been correlated with mobilization of tissue-resident progenitor cells, which is functional to the wound healing process. However, until now there has been no evidence for the presence of cells with a healing capacity circulating in healthy conditions. We identified a rare cell population present in the peripheral blood of healthy mice that actively participates in tissue repair. These Circulating cells, with a Homing ability and involved in the Healing process (CH cells), were identified by an innovative flowcytometry strategy as small cells not expressing CD45 and lineage markers. Their transcriptome profile revealed that CH cells are unique and present a high expression of key pluripotency- and epiblast-associated genes. More importantly, CH-labeled cells derived from healthy Red Fluorescent Protein (RFP)-transgenic mice and systemically injected into syngeneic fractured wild-type mice migrated and engrafted in wounded tissues, ultimately differentiating into tissue-specific cells. Accordingly, the number of CH cells in the peripheral blood rapidly decreased following femoral fracture. These findings uncover the existence of constitutively circulating cells that may represent novel, accessible, and versatile effectors of therapeutic tissue regeneration.
Collapse
|
43
|
Shao J, Zhang W, Yang T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res 2015; 48:62. [PMID: 26530042 PMCID: PMC4630918 DOI: 10.1186/s40659-015-0053-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
Bone is a unique tissue which could regenerate completely after injury rather than heal itself with a scar. Compared with other tissues the difference is that, during bone repairing and regeneration, after the inflammatory phase the mesenchymal stem cells (MSCs) are recruited to the injury site and differentiate into either chondroblasts or osteoblasts precursors, leading to bone repairing and regeneration. Besides these two precursors, the MSCs can also differentiate into adipocyte precursors, skeletal muscle precursors and some other mesodermal cells. With this multilineage potentiality, the MSCs are probably used to cure bone injury and other woundings in the near future. Here we will introduce the recent developments in understanding the mechanism of MSCs action in bone regeneration and repairing.
Collapse
Affiliation(s)
- Jin Shao
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Tieyi Yang
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
44
|
Thompson EM, Matsiko A, Farrell E, Kelly DJ, O'Brien FJ. Recapitulating endochondral ossification: a promising route toin vivobone regeneration. J Tissue Eng Regen Med 2014; 9:889-902. [DOI: 10.1002/term.1918] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/14/2014] [Accepted: 04/24/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Emmet M. Thompson
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| | - Amos Matsiko
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC; University Medical Centre Rotterdam; The Netherlands
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering; Trinity College Dublin; Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| |
Collapse
|
45
|
Ulivi V, Tasso R, Cancedda R, Descalzi F. Mesenchymal stem cell paracrine activity is modulated by platelet lysate: induction of an inflammatory response and secretion of factors maintaining macrophages in a proinflammatory phenotype. Stem Cells Dev 2014; 23:1858-69. [PMID: 24720766 DOI: 10.1089/scd.2013.0567] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Wound healing is achieved through distinct programmed phases: hemostasis, inflammation, mesenchymal cell proliferation and migration, and tissue remodeling. At the injury site, clot formation and platelet degranulation release cytokines and growth factors and actively participating in the healing process and regulating the migration of inflammatory cells, such as neutrophils, macrophages, and lymphocytes. We previously demonstrated that, in an inflammatory environment, prostaglandin E2 (PGE2) secreted by mesenchymal stem cells (MSCs) promoted the macrophage switch from a proinflammatory to a proresolving phenotype. Using an in vitro model, we here evaluated the role carried out by the two main players of the wound healing process, the platelet degranulation content mimicked by the platelet lysate (PL) and the inflammatory stimulus, on the modulation of mouse bone-marrow-derived MSC paracrine activity. We demonstrated that, in MSCs, PL induced nuclear factor kappaB (NF-κB) activation, expression of COX-2 and mPGE synthase, and PGE2 production; in an inflammatory microenvironment, PL increased the inflammatory response and promoted the secretion of the proinflammatory cytokine IL-6. We assayed on mouse primary macrophages the paracrine activity of MSCs exposed to the different microenvironments and we observed that PL-treated MSC-conditioned medium maintained macrophages in a proinflammatory state. The involved factors were granulocyte macrophage-colony stimulating factor induced by PL in MSCs and TNF-α induced by PL-MSC-conditioned medium in macrophages. Our findings indicate that PL triggers an inflammatory response in MSCs and induces the secretion of factors maintaining macrophages in a proinflammatory state thus enhancing the initial inflammatory response to the injury, a key element in the activation of wound healing.
Collapse
Affiliation(s)
- Valentina Ulivi
- 1 Department of Experimental Medicine, University of Genova , Genova, Italy
| | | | | | | |
Collapse
|
46
|
Watson L, Elliman SJ, Coleman CM. From isolation to implantation: a concise review of mesenchymal stem cell therapy in bone fracture repair. Stem Cell Res Ther 2014; 5:51. [PMID: 25099622 PMCID: PMC4055164 DOI: 10.1186/scrt439] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Compromised bone-regenerating capability following a long bone fracture is often the result of reduced host bone marrow (BM) progenitor cell numbers and efficacy. Without surgical intervention, these malunions result in mobility restrictions, deformities, and disability. The clinical application of BM-derived mesenchymal stem cells (MSCs) is a feasible, minimally invasive therapeutic option to treat non-union fractures. This review focuses on novel, newly identified cell surface markers in both the mouse and human enabling the isolation and purification of osteogenic progenitor cells as well as their direct and indirect contributions to fracture repair upon administration. Furthermore, clinical success to date is summarized with commentary on autologous versus allogeneic cell sources and the methodology of cell administration. Given our clinical success to date in combination with recent advances in the identification, isolation, and mechanism of action of MSCs, there is a significant opportunity to develop improved technologies for defining therapeutic MSCs and potential to critically inform future clinical strategies for MSC-based bone regeneration.
Collapse
|
47
|
Raeth S, Sacchetti B, Siegel G, Mau-Holzmann UA, Hansmann J, Vacun G, Hauk TG, Pfizenmaier K, Hausser A. A mouse bone marrow stromal cell line with skeletal stem cell characteristics to study osteogenesis in vitro and in vivo. Stem Cells Dev 2014; 23:1097-108. [PMID: 24405418 DOI: 10.1089/scd.2013.0367] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Bone marrow stromal cells (BMSCs) are composed of progenitor and multipotent skeletal stem cells, which are able to differentiate in vitro into osteocytes, adipocytes, and chondrocytes. Mouse BMSCs (mBMSCs) are a versatile model system to investigate factors involved in BMSC differentiation in vitro and in vivo as a variety of transgenic mouse models are available. In this study, mBMSCs were isolated and osteogenic differentiation was investigated in tissue culture and in vivo. Three out of seven independent cell isolates showed the ability to differentiate into osteocytes, adipocytes, and chondrocytes in vitro. In vitro multipotency of an established mBMSC line was maintained over 45 passages. The osteogenic differentiation of this cell line was confirmed by quantitative polymerase chain reaction (qPCR) analysis of specific markers such as osteocalcin and shown to be Runx2 dependent. Notably, the cell line, when transplanted subcutaneously into mice, possesses full skeletal stem cell characteristics in vivo in early and late passages, evident from bone tissue formation, induction of vascularization, and hematopoiesis. This cell line provides, thus, a versatile tool to unravel the molecular mechanisms governing osteogenesis in vivo thereby aiding to improve current strategies in bone regenerative therapy.
Collapse
Affiliation(s)
- Sebastian Raeth
- 1 Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nassiri SM, Rahbarghazi R. Interactions of Mesenchymal Stem Cells with Endothelial Cells. Stem Cells Dev 2014; 23:319-32. [DOI: 10.1089/scd.2013.0419] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Rahbarghazi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
49
|
The Regenerative Role of the Fetal and Adult Stem Cell Secretome. J Clin Med 2013; 2:302-27. [PMID: 26237150 PMCID: PMC4470151 DOI: 10.3390/jcm2040302] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/17/2013] [Accepted: 11/25/2013] [Indexed: 02/07/2023] Open
Abstract
For a long time, the stem cell regenerative paradigm has been based on the assumption that progenitor cells play a critical role in tissue repair by means of their plasticity and differentiation potential. However, recent works suggest that the mechanism underlying the benefits of stem cell transplantation might relate to a paracrine modulatory effect rather than the replacement of affected cells at the site of injury. Therefore, mounting evidence that stem cells may act as a reservoir of trophic signals released to modulate the surrounding tissue has led to a paradigm shift in regenerative medicine. Attention has been shifted from analysis of the stem cell genome to understanding the stem cell “secretome”, which is represented by the growth factors, cytokines and chemokines produced through paracrine secretion. Insights into paracrine-mediated repair support a new approach in regenerative medicine and the isolation and administration of specific stem cell-derived paracrine factors may represent an extremely promising strategy, introducing paracrine-based therapy as a novel and feasible clinical application. In this review, we will discuss the regenerative potential of fetal and adult stem cells, with particular attention to their secretome.
Collapse
|