1
|
Su YC, Chang Y, Lee WC, Wang JH, Narita T, Takeno H, Syu JY, Jou IM, Hsieh WC. Study of chondrogenesis of umbilical cord mesenchymal stem cells in curdlan- poly(vinyl alcohol) composite hydrogels and its mechanical properties of freezing-thawing treatments. Int J Biol Macromol 2024; 265:130792. [PMID: 38479670 DOI: 10.1016/j.ijbiomac.2024.130792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024]
Abstract
The curdlan gel is a natural material produced by bacteria. It utilizes chemical cross-linking reactions to form a 3D porous composite hydrogel, increasing its porosity and water content, and improving its mechanical properties. It can be used in tissue repair and regenerative medicine. Curdlan-Poly(vinyl alcohol) (PVA) composite hydrogel can rapidly swell within 1 min due to its porous structure. Compression tests confirmed that it still maintains its original mechanical strength, even after five repeated freeze-thaw (FT) processes, making it suitable for long-term cryopreservation. The purpose of this study is to transplant umbilical cord mesenchymal stem cells (UC-MSCs) on Curdlan-PVA composite hydrogel and observe the chondrocytes on the material. The results of using 4',6-diamidino-2-phenylindole (DAPI), hematoxylin and eosin (H&E), calcein-acetoxymethyl ester (calcein AM), and Collagen type II-Fluorescein isothiocyanate (FITC) staining, confirmed that UC-MSCs can attach and differentiate into chondrocytes on 3D Curdlan-PVA composite hydrogel.
Collapse
Affiliation(s)
- Yu-Chieh Su
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yu Chang
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Wei-Chang Lee
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Jhih-Han Wang
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung 824005, Taiwan
| | - Takumi Narita
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Gunma 376-8515, Japan
| | - Hiroyuki Takeno
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Gunma 376-8515, Japan; Gunma University Center for Food Science and Wellness (GUCFW), Gunma 376-8515, Japan
| | - Jie-Yu Syu
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung 824005, Taiwan
| | - I-Ming Jou
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Wen-Chuan Hsieh
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung 824005, Taiwan.
| |
Collapse
|
2
|
Chen X, Liang XM, Zheng J, Dong YH. Stromal cell-derived factor-1α regulates chondrogenic differentiation via activation of the Wnt/β-catenin pathway in mesenchymal stem cells. World J Stem Cells 2023; 15:490-501. [PMID: 37342217 PMCID: PMC10277961 DOI: 10.4252/wjsc.v15.i5.490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/21/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been applied to treat degenerative articular diseases, and stromal cell-derived factor-1α (SDF-1α) may enhance their therapeutic efficacy. However, the regulatory effects of SDF-1α on cartilage differentiation remain largely unknown. Identifying the specific regulatory effects of SDF-1α on MSCs will provide a useful target for the treatment of degenerative articular diseases.
AIM To explore the role and mechanism of SDF-1α in cartilage differentiation of MSCs and primary chondrocytes.
METHODS The expression level of C-X-C chemokine receptor 4 (CXCR4) in MSCs was assessed by immunofluorescence. MSCs treated with SDF-1α were stained for alkaline phosphatase (ALP) and with Alcian blue to observe differentiation. Western blot analysis was used to examine the expression of SRY-box transcription factor 9, aggrecan, collagen II, runt-related transcription factor 2, collagen X, and matrix metalloproteinase (MMP)13 in untreated MSCs, of aggrecan, collagen II, collagen X, and MMP13 in SDF-1α-treated primary chondrocytes, of glycogen synthase kinase 3β (GSK3β) p-GSK3β and β-catenin expression in SDF-1α-treated MSCs, and of aggrecan, collagen X, and MMP13 in SDF-1α-treated MSCs in the presence or absence of ICG-001 (SDF-1α inhibitor).
RESULTS Immunofluorescence showed CXCR4 expression in the membranes of MSCs. ALP stain was intensified in MSCs treated with SDF-1α for 14 d. The SDF-1α treatment promoted expression of collagen X and MMP13 during cartilage differentiation, whereas it had no effect on the expression of collagen II or aggrecan nor on the formation of cartilage matrix in MSCs. Further, those SDF-1α-mediated effects on MSCs were validated in primary chondrocytes. SDF-1α promoted the expression of p-GSK3β and β-catenin in MSCs. And, finally, inhibition of this pathway by ICG-001 (5 µmol/L) neutralized the SDF-1α-mediated up-regulation of collagen X and MMP13 expression in MSCs.
CONCLUSION SDF-1α may promote hypertrophic cartilage differentiation in MSCs by activating the Wnt/β-catenin pathway. These findings provide further evidence for the use of MSCs and SDF-1α in the treatment of cartilage degeneration and osteoarthritis.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Xia-Ming Liang
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Yong-Hui Dong
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
3
|
Garrison AT, Bignold RE, Wu X, Johnson JR. Pericytes: The lung-forgotten cell type. Front Physiol 2023; 14:1150028. [PMID: 37035669 PMCID: PMC10076600 DOI: 10.3389/fphys.2023.1150028] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Pericytes are a heterogeneous population of mesenchymal cells located on the abluminal surface of microvessels, where they provide structural and biochemical support. Pericytes have been implicated in numerous lung diseases including pulmonary arterial hypertension (PAH) and allergic asthma due to their ability to differentiate into scar-forming myofibroblasts, leading to collagen deposition and matrix remodelling and thus driving tissue fibrosis. Pericyte-extracellular matrix interactions as well as other biochemical cues play crucial roles in these processes. In this review, we give an overview of lung pericytes, the key pro-fibrotic mediators they interact with, and detail recent advances in preclinical studies on how pericytes are disrupted and contribute to lung diseases including PAH, allergic asthma, and chronic obstructive pulmonary disease (COPD). Several recent studies using mouse models of PAH have demonstrated that pericytes contribute to these pathological events; efforts are currently underway to mitigate pericyte dysfunction in PAH by targeting the TGF-β, CXCR7, and CXCR4 signalling pathways. In allergic asthma, the dissociation of pericytes from the endothelium of blood vessels and their migration towards inflamed areas of the airway contribute to the characteristic airway remodelling observed in allergic asthma. Although several factors have been suggested to influence this migration such as TGF-β, IL-4, IL-13, and periostin, recent evidence points to the CXCL12/CXCR4 pathway as a potential therapeutic target. Pericytes might also play an essential role in lung dysfunction in response to ageing, as they are responsive to environmental risk factors such as cigarette smoke and air pollutants, which are the main drivers of COPD. However, there is currently no direct evidence delineating the contribution of pericytes to COPD pathology. Although there is a lack of human clinical data, the recent available evidence derived from in vitro and animal-based models shows that pericytes play important roles in the initiation and maintenance of chronic lung diseases and are amenable to pharmacological interventions. Therefore, further studies in this field are required to elucidate if targeting pericytes can treat lung diseases.
Collapse
Affiliation(s)
- Annelise T. Garrison
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Rebecca E. Bignold
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Xinhui Wu
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Jill R. Johnson
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
4
|
Li Y, Yang S, Liu Y, Qin L, Yang S. IFT20 governs mesenchymal stem cell fate through positively regulating TGF-β-Smad2/3-Glut1 signaling mediated glucose metabolism. Redox Biol 2022; 54:102373. [PMID: 35751983 PMCID: PMC9243161 DOI: 10.1016/j.redox.2022.102373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Aberrant lineage allocation of mesenchymal stem cells (MSCs) could cause bone marrow osteoblast-adipocyte imbalance, and glucose as an important nutrient is required for the maintenance of the MSCs' fate and function. Intraflagellar transport 20 (IFT20) is one of the IFT complex B protein which regulates osteoblast differentiation, and bone formation, but how IFT20 regulates MSCs' fate remains undefined. Here, we demonstrated that IFT20 controls MSC lineage allocation through regulating glucose metabolism during skeletal development. IFT20 deficiency in the early stage of MSCs caused significantly shortened limbs, decreased bone mass and significant increase in marrow fat. However, deletion of IFT20 in the later stage of MSCs and osteocytes just slightly decreased bone mass and bone growth and increased marrow fat. Additionally, we found that loss of IFT20 in MSCs promotes adipocyte formation, which enhances RANKL expression and bone resorption. Conversely, ablation of IFT20 in adipocytes reversed these phenotypes. Mechanistically, loss of IFT20 in MSCs significantly decreased glucose tolerance and suppressed glucose uptake and lactate and ATP production. Moreover, loss of IFT20 significantly decreased the activity of TGF-β-Smad2/3 signaling and reduced the binding activity of Smad2/3 to Glut1 promoter to downregulate Glut1 expression. These findings indicate that IFT20 plays essential roles for preventing MSC lineage allocation into adipocytes through TGF-β-Smad2/3-Glut1 axis.
Collapse
Affiliation(s)
- Yang Li
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shuting Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yang Liu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Bignold R, Shammout B, Rowley JE, Repici M, Simms J, Johnson JR. Chemokine CXCL12 drives pericyte accumulation and airway remodeling in allergic airway disease. Respir Res 2022; 23:183. [PMID: 35831901 PMCID: PMC9277926 DOI: 10.1186/s12931-022-02108-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022] Open
Abstract
Background Airway remodeling is a significant contributor to impaired lung function in chronic allergic airway disease. Currently, no therapy exists that is capable of targeting these structural changes and the consequent loss of function. In the context of chronic allergic inflammation, pericytes have been shown to uncouple from the pulmonary microvasculature, migrate to areas of inflammation, and significantly contribute to airway wall remodeling and lung dysfunction. This study aimed to elucidate the mechanism by which pulmonary pericytes accumulate in the airway wall in a model of chronic allergic airway inflammation. Methods Mice were subjected to a protocol of chronic airway inflammation driven by the common environmental aeroallergen house dust mite. Phenotypic changes to lung pericytes were assessed by flow cytometry and immunostaining, and the functional capacity of these cells was evaluated using in vitro migration assays. The molecular mechanisms driving these processes were targeted pharmacologically in vivo and in vitro. Results Pericytes demonstrated increased CXCR4 expression in response to chronic allergic inflammation and migrated more readily to its cognate chemokine, CXCL12. This increase in migratory capacity was accompanied by pericyte accumulation in the airway wall, increased smooth muscle thickness, and symptoms of respiratory distress. Pericyte uncoupling from pulmonary vessels and subsequent migration to the airway wall were abrogated following topical treatment with the CXCL12 neutraligand LIT-927. Conclusion These results provide new insight into the role of the CXCL12/CXCR4 signaling axis in promoting pulmonary pericyte accumulation and airway remodeling and validate a novel target to address tissue remodeling associated with chronic inflammation.
Collapse
Affiliation(s)
- Rebecca Bignold
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Bushra Shammout
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Jessica E Rowley
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Mariaelena Repici
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - John Simms
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Jill R Johnson
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
6
|
Torrecillas-Baena B, Gálvez-Moreno MÁ, Quesada-Gómez JM, Dorado G, Casado-Díaz A. Influence of Dipeptidyl Peptidase-4 (DPP4) on Mesenchymal Stem-Cell (MSC) Biology: Implications for Regenerative Medicine - Review. Stem Cell Rev Rep 2021; 18:56-76. [PMID: 34677817 DOI: 10.1007/s12015-021-10285-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Dipeptidyl peptidase IV (DPP4) is a ubiquitous protease that can be found in membrane-anchored or soluble form. Incretins are one of the main DPP4 substrates. These hormones regulate glucose levels, by stimulating insulin secretion and decreasing glucagon production. Because DPP4 levels are high in diabetes, DPP4 inhibitor (DPP4i) drugs derived from gliptin are widespread used as hypoglycemic agents for its treatment. However, as DPP4 recognizes other substrates such as chemokines, growth factors and neuropeptides, pleiotropic effects have been observed in patients treated with DPP4i. Several of these substrates are part of the stem-cell niche. Thus, they may affect different physiological aspects of mesenchymal stem-cells (MSC). They include viability, differentiation, mobilization and immune response. MSC are involved in tissue homeostasis and regeneration under both physiological and pathological conditions. Therefore, such cells and their secretomes have a high clinical potential in regenerative medicine. In this context, DPP4 activity may modulate different aspects of MSC regenerative capacity. Therefore, the aim of this review is to analyze the effect of different DPP4 substrates on MSC. Likewise, how the regulation of DPP4 activity by DPP4i can be applied in regenerative medicine. That includes treatment of cardiovascular and bone pathologies, cutaneous ulcers, organ transplantation and pancreatic beta-cell regeneration, among others. Thus, DPP4i has an important clinical potential as a complement to therapeutic strategies in regenerative medicine. They involve enhancing the differentiation, immunomodulation and mobilization capacity of MSC for regenerative purposes.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, CIBERFES, 14071, Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain.
| |
Collapse
|
7
|
Bone marrow CD73 + mesenchymal stem cells display increased stemness in vitro and promote fracture healing in vivo. Bone Rep 2021; 15:101133. [PMID: 34632004 PMCID: PMC8493579 DOI: 10.1016/j.bonr.2021.101133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent and considered to be of great potential for regenerative medicine. We could show recently (Breitbach, Kimura et al. 2018) that a subpopulation of MSCs as well as sinusoidal endothelial cells (sECs) in the bone marrow (BM) of CD73-EGFP reporter mice could be labeled in vivo. We took advantage of this model to explore the plasticity and osteogenic potential of CD73-EGFP+ MSCs in vitro and their role in the regenerative response upon bone lesion in vivo. Herein we show that isolated CD73-EGFP+ MSCs displayed more pronounced stemness and stronger in vitro differentiation capacity into the osteogenic lineage compared to CD73-EGFP− MSCs. In a bone fracture model, endogenous BM-resident CD73-EGFP+ MSCs were found to migrate to the fracture site and differentiate into cartilage and bone cells. Our analysis also showed that CD73-EGFP+ sECs contributed to the neovascularization of the fracture site. In addition, grafting of CD73-EGFP+ MSCs into acute bone lesions revealed their capacity to differentiate into chondrocytes and osteocytes in vivo and their contribution to callus formation in the regeneration process of fracture healing. Thus, CD73+ MSCs display enhanced stemness and osteogenic differentiation potential in vitro and in vivo illustrating a prominent role of the CD73+ MSC subpopulation to promote fracture repair. CD73+ mesenchymal stem cells (MSCs) display pronounced proliferation potential. CD73+ MSCs show high in vitro osteogenic differentiation capacity. Endogenous bone marrow-resident CD73+ MSCs contribute to fracture healing. Grafted CD73+ MSCs promote bone formation upon fracture repair.
Collapse
|
8
|
Gan QF, Choy KW, Foo CN, Leong PP, Cheong SK. Incorporating insulin growth Factor‐1 into regenerative and personalised medicine for musculoskeletal disorders: A systematic review. J Tissue Eng Regen Med 2021; 15:419-441. [DOI: 10.1002/term.3192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/17/2021] [Indexed: 01/05/2025]
Affiliation(s)
- Quan Fu Gan
- Pre‐Clinical Sciences Department Faculty of Medicine and Health Sciences UTAR Sg Long Campus Selangor Malaysia
| | - Ker Woon Choy
- Department of Anatomy Faculty of Medicine Universiti Teknologi MARA Sungai Buloh Selangor Malaysia
| | - Chai Nien Foo
- Population Medicine Department Faculty of Medicine and Health Sciences UTAR Sg Long Campus Selangor Malaysia
| | - Pooi Pooi Leong
- Pre‐Clinical Sciences Department Faculty of Medicine and Health Sciences UTAR Sg Long Campus Selangor Malaysia
| | - Soon Keng Cheong
- Medicine Department Faculty of Medicine and Health Sciences UTAR Sg Long Campus Selangor Malaysia
| |
Collapse
|
9
|
Girousse A, Mathieu M, Sastourné-Arrey Q, Monferran S, Casteilla L, Sengenès C. Endogenous Mobilization of Mesenchymal Stromal Cells: A Pathway for Interorgan Communication? Front Cell Dev Biol 2021; 8:598520. [PMID: 33490065 PMCID: PMC7820193 DOI: 10.3389/fcell.2020.598520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
To coordinate specialized organs, inter-tissue communication appeared during evolution. Consequently, individual organs communicate their states via a vast interorgan communication network (ICN) made up of peptides, proteins, and metabolites that act between organs to coordinate cellular processes under homeostasis and stress. However, the nature of the interorgan signaling could be even more complex and involve mobilization mechanisms of unconventional cells that are still poorly described. Mesenchymal stem/stromal cells (MSCs) virtually reside in all tissues, though the biggest reservoir discovered so far is adipose tissue where they are named adipose stromal cells (ASCs). MSCs are thought to participate in tissue maintenance and repair since the administration of exogenous MSCs is well known to exert beneficial effects under several pathological conditions. However, the role of endogenous MSCs is barely understood. Though largely debated, the presence of circulating endogenous MSCs has been reported in multiple pathophysiological conditions, but the significance of such cell circulation is not known and therapeutically untapped. In this review, we discuss current knowledge on the circulation of native MSCs, and we highlight recent findings describing MSCs as putative key components of the ICN.
Collapse
Affiliation(s)
- Amandine Girousse
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Maxime Mathieu
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Quentin Sastourné-Arrey
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sylvie Monferran
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Coralie Sengenès
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
10
|
Kurniawan A, Kodrat E, Gani YI. Effectiveness of granulocyte colony stimulating factor to enhance healing on delayed union fracture model Sprague-Dawley rat. Ann Med Surg (Lond) 2021; 61:54-60. [PMID: 33384875 PMCID: PMC7770509 DOI: 10.1016/j.amsu.2020.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Delayed union is a problem that can occur after fracture healing. Many studies were conducted based on the diamond concept approach to solve the problem of delayed union. Granulocyte-colony stimulating factor (G-CSF) is one of the various substances known to have a positive role in healing skeletal tissue or adjuvant regeneration. This study was conducted to see the effect of G-CSF in affecting delayed union fracture healing. MATERIALS AND METHOD The experimental study was conducted by randomized posttest only control group design on 24 experimental animals Sprague-Dawley white rats that had experienced delayed union models. The study compared the treatment group injected with subcutaneous G-CSF with a control group and was divided into four groups (n = 6). Harvest and follow-up histomorphometry and immunohistochemistry were performed in the second week and in the fourth week the histomorphometry analysis consisted of the percentage of immature bone area, cartilage, and fibrous area. The semiquantitative evaluation of immunohistochemistry with the expression of BMP-2 through the immunoreactive score (IRS). RESULT In the evaluation of histomorphometry and immunohistochemical parameters, there were significantly more woven bone area (p = 0,015), less fibrosis area (p = 0,002) and higher BMP 2 expression (p = 0,004) in treatment group week four compared to control. . CONCLUSION G-CSF was shown to increase the speed of healing in Sprague-Dawley rats on delayed union models evaluated from histomorphometry and immunohistochemical aspects.
Collapse
Affiliation(s)
- Aryadi Kurniawan
- Paediatric Orthopaedic Division, Department of Orthopaedics and Traumatology, Faculty of Medicine Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Evelina Kodrat
- Musculoskletal Pathology Division, Departement of Anatomic Phatology, Faculty of Medicine Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Yogi Ismail Gani
- Orthopaedics and Traumatology, Faculty of Medicine, Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
11
|
Möhlhenrich SC, Kniha K, Magnuska Z, Gremse F, Peters F, Danesh G, Hölzle F, Modabber A. Ischial tuberosity: new donor site for bone grafts in animal cleft research. Sci Rep 2020; 10:20699. [PMID: 33244089 PMCID: PMC7691372 DOI: 10.1038/s41598-020-77862-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/17/2020] [Indexed: 12/30/2022] Open
Abstract
In the context of cleft repair in animal research in rat models, different areas can be used for bone grafting. The aim of the present study was to present the tuberosity of the ischium as a new donor site and to evaluate its quality in relation to an artificial alveolar cleft. Four weeks after creating experimental alveolar clefts in seven Wistar rats, the repair was performed in the now twelve-week-old male animals using bone blocks grafted from the ischial tuberosity. Two days before surgery and two as well as twenty-eight days after surgery, microCT scans were performed, and the grafted bone blocks were analyzed regarding height, width, thickness, and volume. Additionally, bone mineral density (BMD) and bone volume fraction (BV/TV) were measured in the repaired cleft. The mean bone volume of the graft was about 19.77 ± 7.77mm3. Immediately after jaw reconstruction the BMD and BV/TV were about 0.54 ± 0.05 g/cm3 and 54.9 ± 5.07% for the transplant and about 1.13 ± 0.08 g/cm3 and 94.5 ± 3.70%, respectively, for the surrounding bone. Four weeks later the BMD and BV/TV were about 0.57 ± 0.13 g/cm3 and 56.60 ± 13.70% for the transplant and about 11.17 ± 0.07 g/cm3 and 97.50 ± 2.15%, respectively, for the surrounding bone. A hip fracture was found in four of the animals after surgery. The ischial tuberosity offers large bone blocks, which are sufficient for cleft repair in the rat model. However, the bone quality regarding BMD and BV/TV is less compared with the surrounding bone of the alveolar cleft, even after a period of 4 weeks, despite recognizable renovation processes.
Collapse
Affiliation(s)
- Stephan Christian Möhlhenrich
- Department of Orthodontics, University of Witten/Herdecke, Alfred-Herrhausen Str. 45, 58455, Witten, Germany. .,Department of Oral and Maxillofacial Surgery, University Hospital of Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Kristian Kniha
- Department of Oral and Maxillofacial Surgery, University Hospital of Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Zuzanna Magnuska
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Felix Gremse
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Florian Peters
- Department of Oral and Maxillofacial Surgery, University Hospital of Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Gholamreza Danesh
- Department of Orthodontics, University of Witten/Herdecke, Alfred-Herrhausen Str. 45, 58455, Witten, Germany
| | - Frank Hölzle
- Department of Oral and Maxillofacial Surgery, University Hospital of Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Ali Modabber
- Department of Oral and Maxillofacial Surgery, University Hospital of Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| |
Collapse
|
12
|
Shi Z, Xu Y, Mulatibieke R, Zhong Q, Pan X, Chen Y, Lian Q, Luo X, Shi Z, Zhu Q. Nano-Silicate-Reinforced and SDF-1α-Loaded Gelatin-Methacryloyl Hydrogel for Bone Tissue Engineering. Int J Nanomedicine 2020; 15:9337-9353. [PMID: 33262591 PMCID: PMC7699450 DOI: 10.2147/ijn.s270681] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Autologous bone grafts are the gold standard for treating bone defects. However, limited bone supply and morbidity at the donor site restrict its extensive use. Therefore, developing bone graft materials as an alternative to autologous grafts has gained considerable attention. Injectable hydrogels endowed with osteogenic potential have the ability to fill irregular bone defects using minimally invasive procedures and have thus been attracting researchers' attention. However, from a clinical perspective, most fabrication methods employed for the current injectable osteogenic hydrogels are difficult and inconvenient. In the current study, we fabricated an injectable osteogenic hydrogel using a simple and convenient strategy. MATERIALS AND METHODS Gelatin-methacryloyl (GelMA) pre-polymer was synthetized. Nano silicate (SN) and stromal cell-derived factor-1 alpha (SDF-1α) were introduced into the pre-polymer to achieve injectability, controlled release property, excellent osteogenic ability, and efficient stem cell homing. RESULTS The GelMA-SN-SDF-1α demonstrated excellent injectability via a 17-G needle at room temperature. The loaded SDF-1α exhibited a long-term controlled release pattern and efficiently stimulated MSC migration and homing. The GelMA-SN-SDF-1α hydrogel amplified cell spreading, migration, osteogenic-related biomarker expression, and matrix mineralization. The GelMA-SN-SDF-1α hydrogel filled critical-sized calvaria defects in rats and demonstrated excellent bone regeneration ability, as assessed using micro-CT scanning and histomorphometric staining. CONCLUSION The GelMA-SN-SDF-1α hydrogel provides a simple and convenient strategy for the fabrication of injectable osteogenic graft materials.
Collapse
Affiliation(s)
- Zhe Shi
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yichuan Xu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Ruzha Mulatibieke
- Department of Plastic Surgery, The Third Affiliated Hospital of Sun Yet-Sen University, Guangzhou, People’s Republic of China
| | - Qiang Zhong
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xin Pan
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yuhang Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Qiang Lian
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xin Luo
- Rehabilitation Medical School, Guangzhou International Economics College, Guangzhou, People’s Republic of China
| | - Zhanjun Shi
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Qingan Zhu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
13
|
Ye D, Chen C, Wang Q, Zhang Q, Li S, Liu H. Short-wave enhances mesenchymal stem cell recruitment in fracture healing by increasing HIF-1 in callus. Stem Cell Res Ther 2020; 11:382. [PMID: 32894200 PMCID: PMC7487968 DOI: 10.1186/s13287-020-01888-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 01/14/2023] Open
Abstract
Background As a type of high-frequency electrotherapy, a short-wave can promote the fracture healing process; yet, its underlying therapeutic mechanisms remain unclear. Purpose To observe the effect of Short-Wave therapy on mesenchymal stem cell (MSC) homing and relative mechanisms associated with fracture healing. Materials and methods For in vivo study, the effect of Short-Wave therapy to fracture healing was examined in a stabilized femur fracture model of 40 SD rats. Radiography was used to analyze the morphology and microarchitecture of the callus. Additionally, fluorescence assays were used to analyze the GFP-labeled MSC homing after treatment in 20 nude mice with a femoral fracture. For in vitro study, osteoblast from newborn rats simulated fracture site was first irradiated by the Short-Wave; siRNA targeting HIF-1 was used to investigate the role of HIF-1. Osteoblast culture medium was then collected as chemotaxis content of MSC, and the migration of MSC from rats was evaluated using wound healing assay and trans-well chamber test. The expression of HIF-1 and its related factors were quantified by q RT-PCR, ELISA, and Western blot. Results Our in vivo experiment indicated that Short-Wave therapy could promote MSC migration, increase local and serum HIF-1 and SDF-1 levels, induce changes in callus formation, and improve callus microarchitecture and mechanical properties, thus speeding up the healing process of the fracture site. Moreover, the in vitro results further indicated that Short-Wave therapy upregulated HIF-1 and SDF-1 expression in osteoblast and its cultured medium, as well as the expression of CXCR-4, β-catenin, F-actin, and phosphorylation levels of FAK in MSC. On the other hand, the inhibition of HIF-1α was significantly restrained by the inhibition of HIF-1α in osteoblast, and it partially inhibited the migration of MSC. Conclusions These results suggested that Short-Wave therapy could increase HIF-1 in callus, which is one of the crucial mechanisms of chemotaxis MSC homing in fracture healing.
Collapse
Affiliation(s)
- Dongmei Ye
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.
| | - Chen Chen
- Department of Anatomy, Medical College of Dalian University, Dalian, China
| | - Qiwen Wang
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.,Department of Rehabilitation, The people's Hospital of Longhua District, Shenzhen, China
| | - Qi Zhang
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Sha Li
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Hongwei Liu
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| |
Collapse
|
14
|
Desai S, Jayasuriya CT. Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair. Bioengineering (Basel) 2020; 7:E86. [PMID: 32759659 PMCID: PMC7552784 DOI: 10.3390/bioengineering7030086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Harnessing adult mesenchymal stem/progenitor cells to stimulate skeletal tissue repair is a strategy that is being actively investigated. While scientists continue to develop creative and thoughtful ways to utilize these cells for tissue repair, the vast majority of these methodologies can ultimately be categorized into two main approaches: (1) Facilitating the recruitment of endogenous host cells to the injury site; and (2) physically administering into the injury site cells themselves, exogenously, either by autologous or allogeneic implantation. The aim of this paper is to comprehensively review recent key literature on the use of these two approaches in stimulating healing and repair of different skeletal tissues. As expected, each of the two strategies have their own advantages and limitations (which we describe), especially when considering the diverse microenvironments of different skeletal tissues like bone, tendon/ligament, and cartilage/fibrocartilage. This paper also discusses stem/progenitor cells commonly used for repairing different skeletal tissues, and it lists ongoing clinical trials that have risen from the implementation of these cells and strategies. Lastly, we discuss our own thoughts on where the field is headed in the near future.
Collapse
Affiliation(s)
| | - Chathuraka T. Jayasuriya
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and the Rhode Island Hospital, Providence, RI 02903, USA;
| |
Collapse
|
15
|
Liu Y, Xu L, Hu L, Chen D, Yu L, Li X, Chen H, Zhu J, Chen C, Luo Y, Wang B, Li G. Stearic acid methyl ester promotes migration of mesenchymal stem cells and accelerates cartilage defect repair. J Orthop Translat 2020; 22:81-91. [PMID: 32440503 PMCID: PMC7231966 DOI: 10.1016/j.jot.2019.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/22/2019] [Accepted: 09/25/2019] [Indexed: 01/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) can be easily expanded without losing the ability of multilineage differentiation, including oesteogenic, chondrogenic and adipogenic differentiation. These characters make MSCs a promising cell resource for cartilage defect repair. MSCs could be recruited by inflammatory stimulation, then home to the injury tissues. However, its capacity of homing is extremely limited. Thus, it has become extremely necessary to develop an agent or a method, which can be used to enhance the efficiency of MSCs homing. This study investigates the effect of stearic acid methyl ester (SAME) on MSCs mobilisation and cartilage regeneration. Methods MSCs were isolated from femurs of Sprague-Dawley (SD) rats. MTT assay was used to detect effect of SAME on viability of MSCs. Transwell assay and wound healing assay were used to detect effect of SAME on migration of MSCs. RNA-seq, quantitative real-time PCR and western blot were performed to analyze the expression of RNAs and proteins. Colony forming assay and flow cytometry were used to evaluate the effect of SAME on MSCs mobilisation in vivo. A rat cartilage defect model was created to evaluate the effect of SAME on cartilage regeneration. Results We found that SAME could promote the migration of MSCs. Interestingly, we found SAME significantly increased the expression levels of Vav1 in MSCs. On the other hand, the enhanced migration ability of MSCs induced by SAME was retarded by Vav1 small interfering RNA (siRNA) and Rho-associated protein kinase 2 (ROCK2) inhibitor. In addition, we also checked the effect of SAME on mobilisation of MSCs in vivo. The results showed that SAME increased the number of MSCs in peripheral blood and enhanced the capacity of colony formation. Finally, using a cartilage defect model in rats, we found SAME could improve cartilage repair. Conclusion Our study demonstrates that SAME can enhance MSCs migration ability mainly through the Vav1/ROCK2 signaling pathway, which could contribute to the accelerated cartilage regeneration. The translational potential of this article These findings provide evidence that SAME could be used as a therapeutic reagent for MSCs mobilisation and cartilage regeneration.
Collapse
Affiliation(s)
- Yamei Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liuchao Hu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Dongfeng Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lijuan Yu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongtai Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Junlang Zhu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Chen Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yiwen Luo
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Bin Wang
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| |
Collapse
|
16
|
Wang Y, Yao F, Wang L, Li Z, Ren Z, Li D, Zhang M, Han L, Wang SQ, Zhou B, Wang L. Single-cell analysis of murine fibroblasts identifies neonatal to adult switching that regulates cardiomyocyte maturation. Nat Commun 2020; 11:2585. [PMID: 32444791 PMCID: PMC7244751 DOI: 10.1038/s41467-020-16204-w] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiac maturation lays the foundation for postnatal heart development and disease, yet little is known about the contributions of the microenvironment to cardiomyocyte maturation. By integrating single-cell RNA-sequencing data of mouse hearts at multiple postnatal stages, we construct cellular interactomes and regulatory signaling networks. Here we report switching of fibroblast subtypes from a neonatal to adult state and this drives cardiomyocyte maturation. Molecular and functional maturation of neonatal mouse cardiomyocytes and human embryonic stem cell-derived cardiomyocytes are considerably enhanced upon co-culture with corresponding adult cardiac fibroblasts. Further, single-cell analysis of in vivo and in vitro cardiomyocyte maturation trajectories identify highly conserved signaling pathways, pharmacological targeting of which substantially delays cardiomyocyte maturation in postnatal hearts, and markedly enhances cardiomyocyte proliferation and improves cardiac function in infarcted hearts. Together, we identify cardiac fibroblasts as a key constituent in the microenvironment promoting cardiomyocyte maturation, providing insights into how the manipulation of cardiomyocyte maturity may impact on disease development and regeneration.
Collapse
Affiliation(s)
- Yin Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Lipeng Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Zheng Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zongna Ren
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Dandan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Mingzhi Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA
| | - Shi-Qiang Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
17
|
Peeples ES, Dafferner A, Jiang J, Lyden E, Punsoni M, Agrawal DK. Combined Treatment with Insulin-Like Growth Factor 1 and AMD3100 Improves Motor Outcome in a Murine Model of Neonatal Hypoxic-Ischemic Encephalopathy. Dev Neurosci 2020; 41:255-262. [PMID: 32053821 DOI: 10.1159/000505264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/05/2019] [Indexed: 11/19/2022] Open
Abstract
Stem cell transplantation is a promising intervention for neonatal hypoxic-ischemic encephalopathy (HIE); however, universal feasibility and safety have not been thoroughly evaluated. AMD3100 and insulin-like growth factor 1 (IGF1) mobilize progenitor cells into peripheral circulation. The objective of this study was to assess the short-term efficacy of inducing endogenous stem cell mobilization after injury in a model of neonatal HIE. Postnatal day 9 CD1 pups received sham surgery or unilateral carotid artery ligation and 30 min of hypoxia followed by saline, AMD3100, IGF1, or both agents. Intraperitoneal injections of 5-ethynyl-2'-deoxy-uridine (EdU) and 5-bromo-2'-deoxyuridine were used to -label replicating progenitor cells. At P14, animals underwent rotarod testing, and the brains were sectioned for area measurements and immunofluorescence staining. Comparisons were made using one-way analysis of variance. Spearman's rho was calculated to assess correlation between rotarod results and markers of brain injury. Pups treated with both agents had improved rotarod performance (p = 0.02) and increased EdU+ progenitor cells in the subgranular zone (SGZ) compared to injured controls (p = 0.10). An increase in active cells within the SGZ was correlated with improved rotarod performance (r = 0.84, p = 0.04). There were no differences in overall injury score or in brain area or number of activated cells in the subventricular zone between the treatment groups. Combined treatment with AMD3100 and IGF1 shows promise for decreasing brain injury and improving motor function in pups after HIE which correlated with changes in the number of active progenitor cells in the SGZ.
Collapse
Affiliation(s)
- Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA,
| | - Alicia Dafferner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jiang Jiang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Elizabeth Lyden
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Michael Punsoni
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Science, Pomona, California, USA
| |
Collapse
|
18
|
Liu C, Zhu J, Hai B, Zhang W, Wang H, Leng H, Xu Y, Song C. Single Intraosseous Injection of Simvastatin Promotes Endothelial Progenitor Cell Mobilization, Neovascularization, and Wound Healing in Diabetic Rats. Plast Reconstr Surg 2020; 145:433-443. [DOI: 10.1097/prs.0000000000006502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
19
|
Patry C, Doniga T, Lenz F, Viergutz T, Weiss C, Tönshoff B, Kalenka A, Yard B, Krebs J, Schaible T, Beck G, Rafat N. Increased mobilization of mesenchymal stem cells in patients with acute respiratory distress syndrome undergoing extracorporeal membrane oxygenation. PLoS One 2020; 15:e0227460. [PMID: 31986159 PMCID: PMC6984734 DOI: 10.1371/journal.pone.0227460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 12/18/2019] [Indexed: 01/31/2023] Open
Abstract
Background The acute respiratory distress syndrome (ARDS) is characterized by pulmonary epithelial and endothelial barrier dysfunction and injury. In severe forms of ARDS, extracorporeal membrane oxygenation (ECMO) is often the last option for life support. Endothelial progenitor (EPC) and mesenchymal stem cells (MSC) can regenerate damaged endothelium and thereby improve pulmonary endothelial dysfunction. However, we still lack sufficient knowledge about how ECMO might affect EPC- and MSC-mediated regenerative pathways in ARDS. Therefore, we investigated if ECMO impacts EPC and MSC numbers in ARDS patients. Methods Peripheral blood mononuclear cells from ARDS patients undergoing ECMO (n = 16) and without ECMO support (n = 12) and from healthy volunteers (n = 16) were isolated. The number and presence of circulating EPC and MSC was detected by flow cytometry. Serum concentrations of vascular endothelial growth factor (VEGF) and angiopoietin 2 (Ang2) were determined. Results In the ECMO group, MSC subpopulations were higher by 71% compared to the non-ECMO group. Numbers of circulating EPC were not significantly altered. During ECMO, VEGF and Ang2 serum levels remained unchanged compared to the non-ECMO group (p = 0.16), but Ang2 serum levels in non-survivors of ARDS were significantly increased by 100% (p = 0.02) compared to survivors. Conclusions ECMO support in ARDS is specifically associated with an increased number of circulating MSC, most likely due to enhanced mobilization, but not with a higher numbers of EPC or serum concentrations of VEGF and Ang2.
Collapse
Affiliation(s)
- Christian Patry
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Thalia Doniga
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Franziska Lenz
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Tim Viergutz
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Armin Kalenka
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Hospital Bergstraße, Heppenheim, Germany
| | - Benito Yard
- Department of Medicine V, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Jörg Krebs
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Grietje Beck
- Department of Anaesthesiology and Intensive Care Medicine, Dr. Horst-Schmidt Clinic, Wiesbaden, Germany
| | - Neysan Rafat
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Pharmaceutical Sciences, Bahá'í Institute of Higher Education (BIHE), Teheran, Iran
- * E-mail:
| |
Collapse
|
20
|
Klüter T, Hassan R, Rasch A, Naujokat H, Wang F, Behrendt P, Lippross S, Gerdesmeyer L, Eglin D, Seekamp A, Fuchs S. An Ex Vivo Bone Defect Model to Evaluate Bone Substitutes and Associated Bone Regeneration Processes. Tissue Eng Part C Methods 2020; 26:56-65. [DOI: 10.1089/ten.tec.2019.0274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Tim Klüter
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Rywan Hassan
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Alexander Rasch
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Hendrik Naujokat
- Department of Oral and Maxillofacial Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Fanlu Wang
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Peter Behrendt
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sebastian Lippross
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ludger Gerdesmeyer
- Department of Trauma and Orthopedic Surgery, Section for Oncological and Rheumatological Orthopedics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland
| | - Andreas Seekamp
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sabine Fuchs
- Department of Trauma and Orthopedic Surgery, Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
21
|
Allogeneic Versus Autologous Injectable Mesenchymal Stem Cells for Knee Osteoarthritis: Review and Current Status. Tech Orthop 2019. [DOI: 10.1097/bto.0000000000000357] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
22
|
Herrmann M, Jakob F. Bone Marrow Niches for Skeletal Progenitor Cells and their Inhabitants in Health and Disease. Curr Stem Cell Res Ther 2019; 14:305-319. [PMID: 30674266 DOI: 10.2174/1574888x14666190123161447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/04/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
The bone marrow hosts skeletal progenitor cells which have most widely been referred to as Mesenchymal Stem or Stromal Cells (MSCs), a heterogeneous population of adult stem cells possessing the potential for self-renewal and multilineage differentiation. A consensus agreement on minimal criteria has been suggested to define MSCs in vitro, including adhesion to plastic, expression of typical surface markers and the ability to differentiate towards the adipogenic, osteogenic and chondrogenic lineages but they are critically discussed since the differentiation capability of cells could not always be confirmed by stringent assays in vivo. However, these in vitro characteristics have led to the notion that progenitor cell populations, similar to MSCs in bone marrow, reside in various tissues. MSCs are in the focus of numerous (pre)clinical studies on tissue regeneration and repair. Recent advances in terms of genetic animal models enabled a couple of studies targeting skeletal progenitor cells in vivo. Accordingly, different skeletal progenitor cell populations could be identified by the expression of surface markers including nestin and leptin receptor. While there are still issues with the identity of, and the overlap between different cell populations, these studies suggested that specific microenvironments, referred to as niches, host and maintain skeletal progenitor cells in the bone marrow. Dynamic mutual interactions through biological and physical cues between niche constituting cells and niche inhabitants control dormancy, symmetric and asymmetric cell division and lineage commitment. Niche constituting cells, inhabitant cells and their extracellular matrix are subject to influences of aging and disease e.g. via cellular modulators. Protective niches can be hijacked and abused by metastasizing tumor cells, and may even be adapted via mutual education. Here, we summarize the current knowledge on bone marrow skeletal progenitor cell niches in physiology and pathophysiology. We discuss the plasticity and dynamics of bone marrow niches as well as future perspectives of targeting niches for therapeutic strategies.
Collapse
Affiliation(s)
- Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Wuerzburg, Wuerzburg, Germany.,Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
23
|
Meeson R, Sanghani-Keri A, Coathup M, Blunn G. CXCR4 Antagonism to Treat Delayed Fracture Healing. Tissue Eng Part A 2019; 25:1242-1250. [PMID: 30612520 PMCID: PMC6864747 DOI: 10.1089/ten.tea.2018.0265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A significant number of fractures develop nonunion. Stem cell homing is regulated through stromal cell-derived factor 1 (SDF1) and its receptor CXCR4. Stem/progenitor cell populations can be endogenously mobilized by administering growth factors with a pharmacological antagonist of CXCR4, AMD3100, which may be a means to improve fracture healing. A 1.5 mm femoral osteotomy in Wistar rats was stabilized with an external fixator. Rats were pretreated with phosphate buffered saline [PBS(P)], vascular endothelial growth factor [VEGF(V)], insulin-like growth factor-1 [IGF1(I)], or granulocyte colony stimulating factor [GCSF(G)] before AMD3100. A control group (C) did not receive growth factors or AMD3100. Bone formation after 5 weeks was analyzed. Group P had a significant increase in total bone volume (BV) (p = 0.01) and group I in percentage bone in the fracture gap (p = 0.035). Group G showed a decrease in BV. All treated groups had an increase in trabecular thickness. Histology showed decreased cartilage tissue associated with increased bone in groups with improved healing, and increased fibrous tissue in poorly performing groups. Antagonism of SDF1-CXCR4 axis can boost impaired fracture healing. AMD3100 given alone was the most effective means to boost healing, whereas pretreatment with GCSF reduced healing. AMD3100 is likely mobilizing stem cells into the blood stream that home to the fracture site enhancing healing.
Collapse
Affiliation(s)
- Richard Meeson
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,Department of Clinical Services and Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Anita Sanghani-Keri
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom
| | - Melanie Coathup
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,University of Central Florida, Orlando, Florida
| | - Gordon Blunn
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
24
|
Meeson R, Sanghani‐Keri A, Coathup M, Blunn G. VEGF with AMD3100 endogenously mobilizes mesenchymal stem cells and improves fracture healing. J Orthop Res 2019; 37:1294-1302. [PMID: 30345545 PMCID: PMC6563072 DOI: 10.1002/jor.24164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/11/2018] [Indexed: 02/04/2023]
Abstract
A significant number of fractures develop non-union. Mesenchymal stem cell (MSC) therapy may be beneficial, however, this requires cell acquisition, culture and delivery. Endogenous mobilization of stem cells offers a non-invasive alternative. The hypothesis was administration of VEGF and the CXCR4 antagonist AMD3100 would increase the circulating pool of available MSCs and improve fracture healing. Ex-breeder female wistar rats received VEGF followed by AMD3100, or sham PBS. Blood prepared for culture and colonies were counted. P3 cells were analyzed by flow cytometry, bi-differentiation. The effect of mobilization on fracture healing was evaluated with 1.5 mm femoral osteotomy stabilized with an external fixator in 12-14 week old female Wistars. The mobilized group had significantly greater number of cfus/ml compared to controls, p = 0.029. The isolated cells expressed 1.8% CD34, 35% CD45, 61% CD29, 78% CD90, and differentiated into osteoblasts but not into adipocytes. The fracture gap in animals treated with VEGF and AMD3100 showed increased bone volume; 5.22 ± 1.7 µm3 and trabecular thickness 0.05 ± 0.01 µm compared with control animals (4.3 ± 3.1 µm3 , 0.04 ± 0.01 µm, respectively). Radiographic scores quantifying fracture healing (RUST) showed that the animals in the mobilization group had a higher healing score compared to controls (9.6 vs. 7.7). Histologically, mobilization resulted in significantly lower group variability in bone formation (p = 0.032) and greater amounts of bone and less fibrous tissue than the control group. Clinical significance: This pre-clinical study demonstrates a beneficial effect of endogenous MSC mobilization on fracture healing, which may have translation potential to prevent or treat clinical fractures at risk of delayed or non-union fractures. © 2018 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 37:1294-1302, 2019.
Collapse
Affiliation(s)
- Richard Meeson
- Division of SurgeryUniversity College LondonStanmoreUnited Kingdom,Royal Veterinary CollegeHertfordshireUnited Kingdom
| | | | - Melanie Coathup
- Division of SurgeryUniversity College LondonStanmoreUnited Kingdom,University of Central FloridaFlorida
| | - Gordon Blunn
- Division of SurgeryUniversity College LondonStanmoreUnited Kingdom,University of PortsmouthPortsmouthUnited Kingdom
| |
Collapse
|
25
|
Liu S, Liu Y, Jiang L, Li Z, Lee S, Liu C, Wang J, Zhang J. Recombinant human BMP-2 accelerates the migration of bone marrow mesenchymal stem cells via the CDC42/PAK1/LIMK1 pathway in vitro and in vivo. Biomater Sci 2019; 7:362-372. [PMID: 30484785 DOI: 10.1039/c8bm00846a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Biomaterials are widely used for bone regeneration and fracture repair. The migration of bone marrow mesenchymal stem cells (BMSCs) into bone defect sites or material implantation sites, and their differentiation into osteoblasts, is central to the fracture healing process, and the directional migration of BMSCs depends on cytokines or chemokines at the defect site. BMP-2 can stimulate the migration of a variety of cells, but it remains unclear whether BMSC migration can be induced. To provide evidence for BMP-2-induced BMSC migration, we tested the cytoskeletal changes and migration ability of BMSCs after treatment with recombinant human BMP-2 (rhBMP-2). We also explored the recruitment of BMSCs from the circulatory system using a collagen sponge incorporating rhBMP-2 that was implanted in vivo. Furthermore, to understand the mechanism underlying this migration, we investigated the effect of rhBMP-2 on migration-related signal pathways. Here, we found that, rhBMP-2 treatment significantly increased the migration of BMSCs in vitro via activation of the CDC42/PAK1/LIMK1 pathway, and that this migration could be blocked by silencing CDC42. In vivo, collagen sponge material loaded with rhBMP-2 could recruit BMSCs injected into the circulatory system. Moreover, inhibition using the small interfering RNA for CDC42 led to a significant decrease in the number of BMSCs within the material. In conclusion, our data prove that rhBMP-2 can accelerate BMSC migration via the CDC42/PAK1/LIMK1 pathway both in vivo and in vitro, and therefore provides a foundation for further understanding and application of rhBMP-2-incorporated materials by enhancing BMSC recruitment.
Collapse
Affiliation(s)
- Shuhao Liu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030 People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Longhini ALF, Salazar TE, Vieira C, Trinh T, Duan Y, Pay LM, Li Calzi S, Losh M, Johnston NA, Xie H, Kim M, Hunt RJ, Yoder MC, Santoro D, McCarrel TM, Grant MB. Peripheral blood-derived mesenchymal stem cells demonstrate immunomodulatory potential for therapeutic use in horses. PLoS One 2019; 14:e0212642. [PMID: 30870461 PMCID: PMC6417789 DOI: 10.1371/journal.pone.0212642] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Previously, we showed that mesenchymal stem cells (MSC) can be mobilized into peripheral blood using electroacupuncture (EA) at acupoints, LI-4, LI-11, GV-14, and GV-20. The purpose of this study was to determine whether EA-mobilized MSC could be harvested and expanded in vitro to be used as an autologous cell therapy in horses. Peripheral blood mononuclear cells (PBMC) isolated from young and aged lame horses (n = 29) showed a marked enrichment for MSCs. MSC were expanded in vitro (n = 25) and administered intravenously at a dose of 50 x 106 (n = 24). Treatment resulted in significant improvement in lameness as assessed by the American Association of Equine Practitioners (AAEP) lameness scale (n = 23). MSCs exhibited immunomodulatory function by inhibition of lymphocyte proliferation and induction of IL-10. Intradermal testing showed no immediate or delayed immune reactions to MSC (1 x 106 to 1 x 104). In this study, we demonstrated an efficient, safe and reproducible method to mobilize and expand, in vitro, MSCs in sufficiently high concentrations for therapeutic administration. We confirm the immunomodulatory function of these cells in vitro. This non-pharmacological and non-surgical strategy for stem cell harvest has a broad range of biomedical applications and represents an improved clinically translatable and economical cell source for humans.
Collapse
Affiliation(s)
- Ana Leda F. Longhini
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Tatiana E. Salazar
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Cristiano Vieira
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Thao Trinh
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Yaqian Duan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Louise M. Pay
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Megan Losh
- Office of Vice President for Research, Indiana University, School of Medicine IACUC, Indianapolis, Indiana, United States of America
| | - Nancy A. Johnston
- Laboratory Animal Resource Center, Indiana University, Indianapolis, Indiana, United States of America
| | - Huisheng Xie
- College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Minsu Kim
- Department of Clinical Science, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Robert J. Hunt
- Hagyard Equine Medical Institute, Lexington, Kentucky, United States of America
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Domenico Santoro
- College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Taralyn M. McCarrel
- College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
27
|
He C, Li D, Gao J, Li J, Liu Z, Xu W. Inhibition of CXCR4 inhibits the proliferation and osteogenic potential of fibroblasts from ankylosing spondylitis via the Wnt/β‑catenin pathway. Mol Med Rep 2019; 19:3237-3246. [PMID: 30816502 DOI: 10.3892/mmr.2019.9980] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 10/15/2018] [Indexed: 11/06/2022] Open
Abstract
Ankylosing spondylitis (AS) is an autoimmune condition characterized by chronic inflammation and abnormal ossification as the primary features of the disease. The aim of the present study was to investigate the role of C‑X‑C chemokine receptor type 4 (CXCR4) in ossification from patients with AS. CXCR4 expression was assessed by western blot analysis and immunohistochemistry analysis of tissues obtained from patients with AS and controls. Fibroblasts were isolated, cultured and incubated with AMD 3100 and stromal cell‑derived factor‑1 to inhibit and promote CXCR4 levels, respectively. CXCR4 was upregulated in hip synovial tissues from patients with AS compared with that observed in controls. AS fibroblasts exhibited increased proliferation and growth rates. Inhibition of CXCR4 increased the phosphorylation of β‑catenin and downregulated the expression of β‑catenin, v‑myc avian myelocytomatosis viral oncogene homolog, cyclin D1 and osteocalcin. Alizarin red staining demonstrated a decrease in biomineralization activity following the inhibition of CXCR4. These data support the hypothesis that inhibiting CXCR4 in patients with AS may suppress the ossification of fibroblasts.
Collapse
Affiliation(s)
- Chongru He
- Department of Orthopedics, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, P.R. China
| | - Dahe Li
- Department of Orthopedics, The 960th Hospital of People's Liberation Army, Tai'an, Shandong 271000, P.R. China
| | - Jinwei Gao
- Department of Orthopedics, Jiangyan Traditional Chinese Medicine Hospital, Taizhou, Jiangsu 225500, P.R. China
| | - Jia Li
- Department of Orthopedics, Jiangyan Traditional Chinese Medicine Hospital, Taizhou, Jiangsu 225500, P.R. China
| | - Zhongtang Liu
- Department of Orthopedics, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, P.R. China
| | - Weidong Xu
- Department of Orthopedics, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
28
|
Li L, Lu H, Zhao Y, Luo J, Yang L, Liu W, He Q. Functionalized cell-free scaffolds for bone defect repair inspired by self-healing of bone fractures: A review and new perspectives. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:1241-1251. [PMID: 30813005 DOI: 10.1016/j.msec.2019.01.075] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/15/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022]
Abstract
Studies have demonstrated that scaffolds, a component of bone tissue engineering, play an indispensable role in bone repair. However, these scaffolds involving ex-vivo cultivated cells seeded have disadvantages in clinical practice, such as limited autologous cells, time-consuming cell expansion procedures, low survival rate and immune-rejection issues. To overcome these disadvantages, recent focus has been placed on the design of functionalized cell-free scaffolds, instead of cell-seeded scaffolds, that can reduplicate the natural self-healing events of bone fractures, such as inflammation, cell recruitment, vascularization, and osteogenic differentiation. New approaches and applications in tissue engineering and regenerative medicine continue to drive the development of functionalized cell-free scaffolds for bone repair. In this review, the self-healing processes were highlighted, and approaches for the functionalization were summarized. Also, ongoing efforts and breakthroughs in the field of functionalization for bone defect repair were discussed. Finally, a brief summery and new perspectives for functionalization strategies were presented to provide guidelines for further efforts in the design of bioinspired cell-free scaffolds.
Collapse
Affiliation(s)
- Li Li
- Institute for Clean Energy & Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing 400715, PR China; Orthopedic Department, Southwest Hospital, Army Medical University, Chongqing 400038, PR China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Orthopedic Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, PR China
| | - Hongwei Lu
- Orthopedic Department, Southwest Hospital, Army Medical University, Chongqing 400038, PR China
| | - Yulan Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Jiangming Luo
- Center of Joint Surgery, Southwest Hospital, Army Medical University, Chongqing 400038, PR China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Qingyi He
- Institute for Clean Energy & Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing 400715, PR China; Orthopedic Department, Southwest Hospital, Army Medical University, Chongqing 400038, PR China; Orthopedic Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
29
|
Asiedu KO, Ferdousi M, Ton PT, Adler SS, Choyke PL, Sato N. Bone marrow cell homing to sites of acute tibial fracture: 89Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model. EJNMMI Res 2018; 8:109. [PMID: 30547233 PMCID: PMC6292830 DOI: 10.1186/s13550-018-0463-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Background Bone fracture healing is dependent upon the rapid migration and engraftment of bone marrow (BM) progenitor and stem cells to the site of injury. Stromal cell-derived factor-1 plays a crucial role in recruiting BM cells expressing its receptor CXCR4. Recently, a CXCR4 antagonist, plerixafor, has been used to mobilize BM cells into the blood in efforts to enhance cell migration to sites of injury presumably improving healing. In this study, we employed zirconium-89 (89Zr)-oxine-labeled BM cells imaged with positron emission tomography (PET)/computed tomography (CT) to visualize and quantitate BM cell trafficking following acute bone injury and to investigate the effect of plerixafor on BM cell homing. Unilateral 1-mm incisions were created in the distal tibia of mice either on the same day (d0) or 24 h (d1) after 89Zr-oxine-labeled BM cell transfer (n = 4–6, 2–2.3 × 107 cells at 9.65–15.7 kBq/106 cells). Serial microPET/CT imaging was performed and migration of 89Zr-labeled cells to the bone injury was quantified. The effects of three daily doses of plerixafor on cell trafficking were evaluated beginning on the day of fracture generation (n = 4–6). The labeled cells localizing to the fracture were analyzed by flow cytometry and immunohistochemistry. Results In d0- and d1-fracture groups, 0.7% and 1.7% of administered BM cells accumulated within the fracture, respectively. Plerixafor treatment reduced BM cell migration to the fracture by approximately one-third (p < 0.05 for both fracture groups). Flow cytometry analysis of donor cells collected from the injured site revealed a predominance of CD45+ stem/progenitor cell populations and subsequent histological analysis demonstrated the presence of donor cells engrafted within sites of fracture repair. Conclusion 89Zr-oxine labeling enabled visualization and quantitation of BM cell recruitment to acute fractures and further demonstrated that plerixafor plays an inhibitory role in this recruitment. Electronic supplementary material The online version of this article (10.1186/s13550-018-0463-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kingsley O Asiedu
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Munira Ferdousi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Phuongnga T Ton
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Stephen S Adler
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, 21702, USA
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Noriko Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA.
| |
Collapse
|
30
|
Kelly RR, McDonald LT, Pellegrini VD, Cray JJ, Larue AC. Identification of circulating murine CD34 +OCN + cells. Cytotherapy 2018; 20:1371-1380. [PMID: 30340982 DOI: 10.1016/j.jcyt.2018.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND AIMS Previous studies identified a circulating human osteoblastic population that expressed osteocalcin (OCN), increased following fracture and pubertal growth, and formed mineralized colonies in vitro and bone in vivo. A subpopulation expressed CD34, a hematopoietic/endothelial marker. These findings led to our hypothesis that hematopoietic-derived CD34+OCN+ cells exist in the circulation of mice and are modulated after fracture. METHODS Flow cytometry was used to identify CD34+OCN+ cells in male B6.SJL-PtprcaPepcb/BoyJ and Vav-Cre/mTmG (VavR) mice. Non-stabilized tibial fractures were created by three-point bend. Fractures were longitudinally imaged by micro-computed tomography, and immunofluorescent staining was used to evaluate CD34+OCN+ cells within fracture callus. AMD3100 (10 mg/kg) was injected subcutaneously for 3 days and the CD34+OCN+ population was evaluated by flow cytometry. RESULTS Circulating CD34+OCN+ cells were identified in mice and confirmed to be of hematopoietic origin (CD45+; Vav1+) using two mouse models. Both circulating and bone marrow-derived CD34+OCN+ cells peaked three weeks post-non-stabilized tibial fracture, suggesting association with cartilage callus transition to bone and early mineralization. Co-expression of CD34 and OCN in the fracture callus at two weeks post-fracture was observed. By three weeks, there was 2.1-fold increase in number of CD34+OCN+ cells, and these were observed throughout the fracture callus. AMD3100 altered CD34+OCN+ cell levels in peripheral blood and bone marrow. DISCUSSION Together, these data demonstrate a murine CD34+OCN+ circulating population that may be directly involved in fracture repair. Future studies will molecularly characterize CD34+OCN+ cells, determine mechanisms regulating their contribution, and examine if their number correlates with improved fracture healing outcomes.
Collapse
Affiliation(s)
- Ryan R Kelly
- Research Services, Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Lindsay T McDonald
- Research Services, Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Vincent D Pellegrini
- Department of Orthopedics, Medical University of South Carolina, Charleston, SC, USA
| | - James J Cray
- Division of Anatomy, The Ohio State University, Columbus, OH, USA
| | - Amanda C Larue
- Research Services, Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
31
|
Abstract
Supplemental Digital Content is available in the text Endogenously mobilized stem and progenitor cells (SPCs) or exogenously provided SPCs are thought to be beneficial for trauma therapy. However, still little is known about the synchronized dynamics of the number of SPCs in blood after severe injury and parameters like cytokine profiles that correlate with these numbers. We determined the number of hematopoietic stem cells, common myeloid progenitors, granulocyte-macrophage progenitors, and mesenchymal stem/stromal cells in peripheral blood (PB) 0 to 3, 8, 24, 48, and 120 h after polytrauma in individual patients (injury severity score ≥ 21). We found that the number of blood SPCs follows on average a synchronous, inverse bell-shaped distribution, with an increase at 0 to 3 h, followed by a strong decrease, with a nadir in SPC numbers in blood at 24 or 48 h. The change in numbers of SPCs in PB between 48 h and 120 h revealed two distinct patterns: Pattern 1 is characterized by an increase in the number of SPCs to a level higher than normal, pattern 2 is characterized by an almost absent increase in the number of SPCs compared to the nadir. Changes in the concentrations of the cytokines CK, MDC, IL-8, G-CSF Gro-α, VEGF, and MCP-1 correlated with changes in the number of SPCs in PB or were closely associated with Pattern 1 or Pattern 2. Our data provide novel rationale for investigations on the role of stem cell mobilization in polytraumatized patients and its likely positive impact on trauma outcome.
Collapse
|
32
|
Moukoko D, Pourquier D, Genovesio C, Thezenas S, Chabrand P, Roffino S, Pithioux M. Granulocyte-colony stimulating factor enhances bone fracture healing. Clin Biomech (Bristol, Avon) 2018; 58:62-68. [PMID: 30036852 DOI: 10.1016/j.clinbiomech.2018.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 04/27/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Circulating mesenchymal stem cells contribute to bone repair. Their incorporation in fracture callus is correlated to their bioavailability. In addition, Granulocyte-colony stimulating factor induces the release of vascular and mesenchymal progenitors. We hypothesized that this glycoprotein stimulates fracture healing, and analyzed the effects of its administration at low doses on bone healing. METHODS 27 adult male Sprague-Dawley rats underwent mid-femur osteotomy stabilized by centromedullar pinning. In a post (pre) operative group, rats were subcutaneously injected with 5 μg/kg per day of Granulocyte-colony stimulating factor for 5 days after (before) surgery. In a control group, rats were injected with saline solution for 5 days immediately after surgery. A radiographic consolidation score was calculated. At day 35, femurs were studied histologically and underwent biomechanical tests. FINDINGS 5 weeks after surgery, mean radiographic scores were significantly higher in the Preop group 7.75 (SD 0.42) and in the Postop group 7.67 (SD 0.52) than in the control group 6.75 (SD 0.69). Biomechanical tests showed femur stiffness to be more than three times higher in both the Preop 109.24 N/mm (SD 51.86) and Postop groups 100.05 N/mm (SD 60.24) than in control 32.01 N/mm (SD 15.78). Mean maximal failure force was twice as high in the Preop group 68.66 N (SD 27.78) as in the control group 34.21 N (SD 11.79). Histological results indicated a later consolidation process in control than in treated groups. INTERPRETATION Granulocyte-colony stimulating factor injections strongly stimulated early femur fracture healing, indicating its potential utility in human clinical situations such as programmed osteotomy and fracture.
Collapse
Affiliation(s)
- Didier Moukoko
- Département de Chirurgie Orthopédique Pédiatrique, CHU Angers, 4 rue Larrey, 49100 Angers, France
| | - Didier Pourquier
- Institut régional du Cancer de Montpellier (ICM)- Val d'Aurelle, Montpellier, France
| | - Cécile Genovesio
- Laboratoire de Biochimie, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Simon Thezenas
- Institut régional du Cancer de Montpellier (ICM)- Val d'Aurelle, Montpellier, France
| | - Patrick Chabrand
- Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France; APHM, Hôpital Sainte Marguerite, IML, Marseille, France
| | - Sandrine Roffino
- Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France; Université Côte d'Azur, Univ Nice Sophia Antipolis, France
| | - Martine Pithioux
- Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France; APHM, Hôpital Sainte Marguerite, IML, Marseille, France.
| |
Collapse
|
33
|
Leitão L, Alves CJ, Alencastre IS, Sousa DM, Neto E, Conceição F, Leitão C, Aguiar P, Almeida-Porada G, Lamghari M. Bone marrow cell response after injury and during early stage of regeneration is independent of the tissue-of-injury in 2 injury models. FASEB J 2018; 33:857-872. [PMID: 30044924 DOI: 10.1096/fj.201800610rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Selectively recruiting bone marrow (BM)-derived stem and progenitor cells to injury sites is a promising therapeutic approach. The coordinated action of soluble factors is thought to trigger the mobilization of stem cells from the BM and recruit them to lesions to contribute to tissue regeneration. Nevertheless, the temporal response profile of the major cellular players and soluble factors involved in priming the BM and recruiting BM-derived cells to promote regeneration is unknown. We show that injury alters the BM cellular composition, introducing population-specific fluctuations during tissue regeneration. We demonstrate that injury causes an immediate, transient response of mesenchymal stromal cells and endothelial cells followed by a nonoverlapping increase in hematopoietic stem and progenitor cells. Moreover, BM reaction is identical whether the injury is inflicted on skin and muscle or also involves a bone defect, but these 2 injury paradigms trigger distinct systemic cytokine responses. Together, our results indicate that the BM response to injury in the early stages of regeneration is independent of the tissue-of-injury based on the 2 models used, but the injured tissue dictates the systemic cytokine response.-Leitão, L., Alves, C. J., Alencastre, I. S., Sousa, D. M., Neto, E., Conceição, F., Leitão, C., Aguiar, P., Almeida-Porada, G., Lamghari, M. Bone marrow cell response after injury and during early stage of regeneration is independent of the tissue-of-injury in 2 injury models.
Collapse
Affiliation(s)
- Luís Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cecília J Alves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Inês S Alencastre
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Daniela M Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Francisco Conceição
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Catarina Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal; and
| | - Paulo Aguiar
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
34
|
Novel Lipid Signaling Mediators for Mesenchymal Stem Cell Mobilization during Bone Repair. Cell Mol Bioeng 2018; 11:241-253. [PMID: 29983824 DOI: 10.1007/s12195-018-0532-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Introduction Mesenchymal stem and progenitor cells (MSCs), which normally reside in the bone marrow, are critical to bone health and can be recruited to sites of traumatic bone injury, contributing to new bone formation. The ability to control the trafficking of MSCs provides therapeutic potential for improving traumatic bone healing and therapy for genetic bone diseases such as hypophosphatasia. Methods In this study, we explored the sphingosine-1-phosphate (S1P) signaling axis as a means to control the mobilization of MSCs into blood and possibly to recruit MSCs enhancing bone growth. Results Loss of S1P receptor 3 (S1PR3) leads to an increase in circulating CD45-/CD29+/CD90+/Sca1 putative mesenchymal progenitor cells, suggesting that blocking S1PR3 may stimulate MSCs to leave the bone marrow. Antagonism of S1PR3 with the small molecule VPC01091 stimulated acute migration of CD45-/CD29+/CD90+/Sca1+ MSCs into the blood as early as 1.5 hours after treatment. VPC01091 administration also increased ectopic bone formation induced by BMP-2 and significantly increased new bone formation in critically sized rat cranial defects, suggesting that mobilized MSCs may home to injuries to contribute to healing. We also explored the possibility of combining S1P manipulation of endogenous host cell occupancy with exogenous MSC transplantation for potential use in combination therapies. Importantly, reducing niche occupancy of host MSCs with VPC01091 does not impede engraftment of exogenous MSCs. Conclusions Our studies suggest that MSC mobilization through S1PR3 antagonism is a promising strategy for endogenous tissue engineering and improving MSC delivery to treat bone diseases.
Collapse
|
35
|
Liu L, Yu Q, Fu S, Wang B, Hu K, Wang L, Hu Y, Xu Y, Yu X, Huang H. CXCR4 Antagonist AMD3100 Promotes Mesenchymal Stem Cell Mobilization in Rats Preconditioned with the Hypoxia-Mimicking Agent Cobalt Chloride. Stem Cells Dev 2018; 27:466-478. [PMID: 29433375 DOI: 10.1089/scd.2017.0191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mobilization of mesenchymal stem cells (MSCs) is an attractive strategy for cell therapy. Our previous study demonstrated that MSCs can be mobilized in circulating blood by short-term hypoxia, and hypoxia-inducible factor-1α is essential for MSC mobilization. In the present study, the effect of the hypoxia-mimicking agent CoCl2 was examined on MSC mobilization. The results indicated that the frequency of circulating MSCs increased slightly by administration of CoCl2. However, the mobilization efficiency was low. Considering the critical role of stromal cell-derived factor-1α (SDF-1)/CXCR4 axis in the regulation of MSC migration, the effects of granulocyte colony-stimulating factor (G-CSF) and the CXCR4 antagonist AMD3100 were investigated on MSC mobilization. The experiments were notably demonstrated in animals preconditioned with CoCl2. The frequency of colony-forming unit fibroblast and the proportion of CD45-CD90+ cells did not significantly increase in the peripheral blood of rats treated with G-CSF and/or AMD3100 alone. The concomitant administration of G-CSF with CoCl2 could not stimulate the release of MSCs. However, AMD3100 dramatically increased MSC mobilization efficiency in rats pretreated with CoCl2. Furthermore, we identified and compared the multilineage differentiation capacities of MSCs derived from bone marrow (BM-MSCs) and mobilized peripheral blood (PB-MSCs). The results indicated that PB-MSCs exhibited higher osteogenic potential and lower adipogenic differentiation as compared with BM-MSCs. The findings may inform studies investigating mechanisms of the regulation of MSC mobilization and can aid in the development of clinically useful therapeutic agents.
Collapse
Affiliation(s)
- Lizhen Liu
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Qin Yu
- 2 College of Life Science, Zhejiang Chinese Medical University , Hangzhou, People's Republic of China
| | - Shan Fu
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Binsheng Wang
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Kaimin Hu
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Limengmeng Wang
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Yongxian Hu
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Yulin Xu
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - Xiaohong Yu
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| | - He Huang
- 1 Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou, People's Republic of China
| |
Collapse
|
36
|
Parry SM, Peeples ES. The impact of hypoxic-ischemic brain injury on stem cell mobilization, migration, adhesion, and proliferation. Neural Regen Res 2018; 13:1125-1135. [PMID: 30028311 PMCID: PMC6065219 DOI: 10.4103/1673-5374.235012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy continues to be a significant cause of death or neurodevelopmental delays despite standard use of therapeutic hypothermia. The use of stem cell transplantation has recently emerged as a promising supplemental therapy to further improve the outcomes of infants with hypoxic-ischemic encephalopathy. After the injury, the brain releases several chemical mediators, many of which communicate directly with stem cells to encourage mobilization, migration, cell adhesion and differentiation. This manuscript reviews the biomarkers that are released from the injured brain and their interactions with stem cells, providing insight regarding how their upregulation could improve stem cell therapy by maximizing cell delivery to the injured tissue.
Collapse
Affiliation(s)
- Stephanie M Parry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
37
|
Poudel SB, Bhattarai G, Kim JH, Kook SH, Seo YK, Jeon YM, Lee JC. Local delivery of recombinant human FGF7 enhances bone formation in rat mandible defects. J Bone Miner Metab 2017; 35:485-496. [PMID: 27766421 DOI: 10.1007/s00774-016-0784-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
Fibroblast growth factor 7 (FGF7) plays an important role in regulating the proliferation, migration, and differentiation of cells. However, the role of FGF7 in bone formation is not yet fully understood. We examined the effect of FGF7 on bone formation using a rat model of mandible defects. Rats underwent mandible defect surgery and then either scaffold treatment alone (control group) or FGF7-impregnated scaffold treatment (FGF7 group). Micro-CT and histological analyses revealed that the FGF7 group exhibited greater bone formation than did the control group 10 weeks after surgery. With the exception of total porosity (%), all bone parameters had higher values in the FGF7 group than in the control group at each follow-up after surgery. The FGF7 group showed greater expression of osteogenic markers, such as runt-related transcription factor 2, osterix, osteocalcin, bone morphogenetic protein 2, osteopontin, and type I collagen in newly formed bone than did the control group. The delivery of FGF7 also increased the messenger RNA expression of stromal-cell-derived factor 1 (SDF-1) and CXCR4 in newly formed bone in the FGF7 group compared with the control group. Further, addition of exogenous FGF7 induced migration of rat bone marrow stromal cells and increased the expression of SDF-1 and CXCR4 in the cells. Furthermore, the addition of FGF7 augmented mineralization in the cells with increased expression of osteogenic markers, and this augmentation was significantly suppressed by an inhibitor specific for c-Jun N-terminal kinase (SP600125) or extracellular-signal-regulated kinase (PD98059). Collectively, these results suggest that local delivery of FGF7 increases bone formation in a mandible defect with enhanced osteogenesis and chemoattraction.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Chonbuk National University, Jeonju, 54896, South Korea
| | - Govinda Bhattarai
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Chonbuk National University, Jeonju, 54896, South Korea
| | - Jae-Hwan Kim
- Chonnam National University Dental Hospital, Kwangju, 61186, South Korea
| | - Sung-Ho Kook
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Chonbuk National University, Jeonju, 54896, South Korea
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, 54896, South Korea
| | - Young-Kwon Seo
- Research Institute of Biotechnology, Dongguk University, Seoul, 04620, South Korea
| | - Young-Mi Jeon
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Chonbuk National University, Jeonju, 54896, South Korea.
- Biomedical Research Institute of Chonbuk National University Hospital, Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, 54896, South Korea.
- Research Institute of Clinical Medicine of Chonbuk National University, School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea.
| | - Jeong-Chae Lee
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Chonbuk National University, Jeonju, 54896, South Korea.
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, 54896, South Korea.
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, 561-756, South Korea.
| |
Collapse
|
38
|
Blázquez-Prunera A, Almeida CR, Barbosa MA. Human Bone Marrow Mesenchymal Stem/Stromal Cells Preserve Their Immunomodulatory and Chemotactic Properties When Expanded in a Human Plasma Derived Xeno-Free Medium. Stem Cells Int 2017; 2017:2185351. [PMID: 28588620 PMCID: PMC5446864 DOI: 10.1155/2017/2185351] [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: 11/29/2016] [Revised: 02/08/2017] [Accepted: 02/26/2017] [Indexed: 01/14/2023] Open
Abstract
Due to their immunomodulatory and chemotactic properties, hMSC are being explored to treat immune-related diseases. For their use in human therapies, it is necessary to culture hMSC in xeno-free conditions. In this study, the impact that a xeno-free medium based on a human plasma derivate has on these properties was analysed. Bone marrow-derived hMSC preserved their immunosuppressive and immunostimulatory properties, as observed with in vitro assays with hMSC cocultured with mixed leukocyte reactions or with mitogen-stimulated leukocytes. Moreover, hMSC expanded in xeno-free medium were recruited by macrophages in both migration and invasion assays, which indicates that the cells maintained their chemotactic properties. These data suggest that xeno-free expanded hMSC preserved their immunomodulatory and chemotactic properties, indicating that the described xeno-free medium composition is a potential candidate to culture and expand hMSC for human cell therapies.
Collapse
Affiliation(s)
- A. Blázquez-Prunera
- Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - C. R. Almeida
- Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Department of Medical Sciences and Institute for Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| | - M. A. Barbosa
- Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
39
|
Salazar TE, Richardson MR, Beli E, Ripsch MS, George J, Kim Y, Duan Y, Moldovan L, Yan Y, Bhatwadekar A, Jadhav V, Smith JA, McGorray S, Bertone AL, Traktuev DO, March KL, Colon-Perez LM, Avin K, Sims E, Mund JA, Case J, Deng S, Kim MS, McDavitt B, Boulton ME, Thinschmidt J, Calzi SL, Fitz SD, Fuchs RK, Warden SJ, McKinley T, Shekhar A, Febo M, Johnson PL, Chang LJ, Gao Z, Kolonin MG, Lai S, Ma J, Dong X, White FA, Xie H, Yoder MC, Grant MB. Electroacupuncture Promotes Central Nervous System-Dependent Release of Mesenchymal Stem Cells. Stem Cells 2017; 35:1303-1315. [PMID: 28299842 PMCID: PMC5530374 DOI: 10.1002/stem.2613] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/16/2017] [Accepted: 02/11/2017] [Indexed: 12/12/2022]
Abstract
Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.
Collapse
Affiliation(s)
- Tatiana E. Salazar
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Matthew R. Richardson
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eleni Beli
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S. Ripsch
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - John George
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Youngsook Kim
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - Yaqian Duan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leni Moldovan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuanqing Yan
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
| | - Ashay Bhatwadekar
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vaishnavi Jadhav
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jared A. Smith
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - Susan McGorray
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA
| | - Alicia L. Bertone
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Dmitri O. Traktuev
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Vascular Biology and Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Keith L. March
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Vascular Biology and Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Luis M. Colon-Perez
- Department of Psychiatry, University of Florida, McKnight Brain Institute, Gainesville, FL 32610, USA
| | - Keith Avin
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Emily Sims
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Julie A. Mund
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Jamie Case
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
- Scripps Clinic Medical Group, Scripps Center for Organ and Cell Transplantation, La Jolla, CA 92037, USA
- Department of Pediatrics, Indiana University, Indianapolis, IN 46202USA
| | - Shaolin Deng
- Mainland Acupuncture, Gainesville, FL 32653, USA
| | - Min Su Kim
- College of Veterinary Medicine, Chon Buk National University, Jeonju, South Korea
| | | | - Michael E. Boulton
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeffrey Thinschmidt
- Department of Pharmacology, University of Florida, Gainesville, FL 32610, USA
| | - Sergio Li Calzi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephanie D. Fitz
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Robyn K. Fuchs
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Stuart J. Warden
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Todd McKinley
- Department of Orthopedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anantha Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, McKnight Brain Institute, Gainesville, FL 32610, USA
| | - Phillip L. Johnson
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lung Ji Chang
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL 32611, USA
| | - Zhanguo Gao
- Center for Metabolic and Degenerative Diseases, Harry E. Bovay Institute of Molecular Medicine University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mikhail G. Kolonin
- Center for Metabolic and Degenerative Diseases, Harry E. Bovay Institute of Molecular Medicine University of Texas Health Science Center, Houston, TX 77030, USA
| | - Song Lai
- Department of Radiation Oncology, University of Florida School of Medicine, Gainesville, FL 32610, USA
| | - Jinfeng Ma
- Department of Radiation Oncology, University of Florida School of Medicine, Gainesville, FL 32610, USA
| | - Xinzhong Dong
- Department of Neuroscience, Center of Sensory Biology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fletcher A. White
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - Huisheng Xie
- College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Mervin C. Yoder
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Maria B. Grant
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
40
|
Zorzopulos J, Opal SM, Hernando-Insúa A, Rodriguez JM, Elías F, Fló J, López RA, Chasseing NA, Lux-Lantos VA, Coronel MF, Franco R, Montaner AD, Horn DL. Immunomodulatory oligonucleotide IMT504: Effects on mesenchymal stem cells as a first-in-class immunoprotective/immunoregenerative therapy. World J Stem Cells 2017; 9:45-67. [PMID: 28396715 PMCID: PMC5368622 DOI: 10.4252/wjsc.v9.i3.45] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/12/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023] Open
Abstract
The immune responses of humans and animals to insults (i.e., infections, traumas, tumoral transformation and radiation) are based on an intricate network of cells and chemical messengers. Abnormally high inflammation immediately after insult or abnormally prolonged pro-inflammatory stimuli bringing about chronic inflammation can lead to life-threatening or severely debilitating diseases. Mesenchymal stem cell (MSC) transplant has proved to be an effective therapy in preclinical studies which evaluated a vast diversity of inflammatory conditions. MSCs lead to resolution of inflammation, preparation for regeneration and actual regeneration, and then ultimate return to normal baseline or homeostasis. However, in clinical trials of transplanted MSCs, the expectations of great medical benefit have not yet been fulfilled. As a practical alternative to MSC transplant, a synthetic drug with the capacity to boost endogenous MSC expansion and/or activation may also be effective. Regarding this, IMT504, the prototype of a major class of immunomodulatory oligonucleotides, induces in vivo expansion of MSCs, resulting in a marked improvement in preclinical models of neuropathic pain, osteoporosis, diabetes and sepsis. IMT504 is easily manufactured and has an excellent preclinical safety record. In the small number of patients studied thus far, IMT504 has been well-tolerated, even at very high dosage. Further clinical investigation is necessary to demonstrate the utility of IMT504 for resolution of inflammation and regeneration in a broad array of human diseases that would likely benefit from an immunoprotective/immunoregenerative therapy.
Collapse
|
41
|
Garg P, Mazur MM, Buck AC, Wandtke ME, Liu J, Ebraheim NA. Prospective Review of Mesenchymal Stem Cells Differentiation into Osteoblasts. Orthop Surg 2017; 9:13-19. [PMID: 28276640 DOI: 10.1111/os.12304] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/27/2016] [Indexed: 01/02/2023] Open
Abstract
Stem cell research has been a popular topic in the past few decades. This review aims to discuss factors that help regulate, induce, and enhance mesenchymal stem cell (MSC) differentiation into osteoblasts for bone regeneration. The factors analyzed include bone morphogenic protein (BMP), transforming growth factor β (TGF-β), stromal cell-derived factor 1 (SDF-1), insulin-like growth factor type 1 (IGF-1), histone demethylase JMJD3, cyclin dependent kinase 1 (CDK1), fucoidan, Runx2 transcription factor, and TAZ transcriptional coactivator. Methods promoting bone healing are also evaluated in this review that have shown promise in previous studies. Methods tested using animal models include low intensity pulsed ultrasound (LIPUS) with MSC, micro motion, AMD3100 injections, BMP delivery, MSC transplantation, tissue engineering utilizing scaffolds, anti-IL-20 monoclonal antibody, low dose photodynamic therapy, and bone marrow stromal cell transplants. Human clinical trial methods analyzed include osteoblast injections, bone marrow grafts, bone marrow and platelet rich plasma transplantation, tissue engineering using scaffolds, and recombinant human BMP-2. These methods have been shown to promote and accelerate new bone formation. These various methods for enhanced bone regeneration have the potential to be used, following further research, in clinical practice.
Collapse
Affiliation(s)
- Priyanka Garg
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Matthew M Mazur
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Amy C Buck
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Meghan E Wandtke
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Jiayong Liu
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Nabil A Ebraheim
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| |
Collapse
|
42
|
Ba H, Li B, Li X, Li C, Feng A, Zhu Y, Wang J, Li Z, Yin B. Transmembrane tumor necrosis factor-α promotes the recruitment of MDSCs to tumor tissue by upregulating CXCR4 expression via TNFR2. Int Immunopharmacol 2017; 44:143-152. [PMID: 28092866 DOI: 10.1016/j.intimp.2016.12.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/12/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) accumulated in tumor sites promote immune evasion. We found that TNFR deficiency-induced rejection of transplanted tumor was accompanied with markedly decreased accumulation of MDSCs. However, the mechanism(s) behind this phenomenon is not completely understood. Here, we demonstrated that TNFR deficiency did not affect the amount of MDSCs in bone marrow (BM), but decreased accumulation of Gr-1+CD11b+ MDSCs in the spleen and tumor tissues. The chemotaxis of Tnfr-/- MDSCs was prominently decreased in response to both tumor cell culture supernatants and tumor tissue homogenates from Tnfr-/- and wild-type mice, indicating an effect of TNFR signaling on chemokine receptor expression in MDSCs. We used real-time PCR to detect gene expression for several chemokine receptors in MDSCs from BM and found that CXCR4 was the most affected molecule at the transcriptional level in Tnfr-/- MDSCs. Neutralizing CXCR4 in wild-type MDSCs by a specific antibody blocked their chemotactic migration. Interestingly, it was tmTNF-α, but not sTNF-α, that induced CXCR4 expression in MDSCs. This effect of tmTNF-α was totally blocked in TNFR2-/- but not in TNFR1-/- MDSCs, and partially inhibited by PDTC or SB203580, an inhibitor of NF-κB or p38 MAPK pathway, respectively. Adoptive transfer of wild-type MDSCs restored MDSCs accumulation in tumors of Tnfr-/- mice, but this could be partially blocked by treatment with a CXCR4 inhibitor AMD3100. Our data suggest that tmTNF-α upregulates CXCR4 expression that promotes chemotaxis of MDSCs to tumor, and give a new insight into a novel mechanism by which tmTNF-α facilitates tumor immune evasion.
Collapse
Affiliation(s)
- Hongping Ba
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Baihua Li
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Hematology, Yichang Central People's Hospital, Yichang, Hubei 443003, China.
| | - Xiaoyan Li
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Cheng Li
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Anlin Feng
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yazhen Zhu
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Jing Wang
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Zhuoya Li
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Bingjiao Yin
- Department of Immunology, Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
43
|
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School, Royal North Shore Hospital, The University of Sydney, Camperdown, NSW, Australia
- School of Biomedical Engineering, The University of New South Wales, Kensington, NSW, Australia
| |
Collapse
|
44
|
Diehl R, Ferrara F, Müller C, Dreyer AY, McLeod DD, Fricke S, Boltze J. Immunosuppression for in vivo research: state-of-the-art protocols and experimental approaches. Cell Mol Immunol 2016; 14:146-179. [PMID: 27721455 PMCID: PMC5301156 DOI: 10.1038/cmi.2016.39] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 05/30/2016] [Accepted: 05/30/2016] [Indexed: 02/06/2023] Open
Abstract
Almost every experimental treatment strategy using non-autologous cell, tissue or organ transplantation is tested in small and large animal models before clinical translation. Because these strategies require immunosuppression in most cases, immunosuppressive protocols are a key element in transplantation experiments. However, standard immunosuppressive protocols are often applied without detailed knowledge regarding their efficacy within the particular experimental setting and in the chosen model species. Optimization of such protocols is pertinent to the translation of experimental results to human patients and thus warrants further investigation. This review summarizes current knowledge regarding immunosuppressive drug classes as well as their dosages and application regimens with consideration of species-specific drug metabolization and side effects. It also summarizes contemporary knowledge of novel immunomodulatory strategies, such as the use of mesenchymal stem cells or antibodies. Thus, this review is intended to serve as a state-of-the-art compendium for researchers to refine applied experimental immunosuppression and immunomodulation strategies to enhance the predictive value of preclinical transplantation studies.
Collapse
Affiliation(s)
- Rita Diehl
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | - Fabienne Ferrara
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany.,Institute of Vegetative Physiology, Charite University Medicine and Center for Cardiovascular Research, Berlin 10115, Germany
| | - Claudia Müller
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | - Antje Y Dreyer
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | | | - Stephan Fricke
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | - Johannes Boltze
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany.,Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck 23562, Germany
| |
Collapse
|
45
|
Intramembranous bone regeneration and implant placement using mechanical femoral marrow ablation: rodent models. BONEKEY REPORTS 2016; 5:837. [PMID: 27648259 DOI: 10.1038/bonekey.2016.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/19/2016] [Indexed: 12/16/2022]
Abstract
In this paper, we provide a detailed protocol for a model of long bone mechanical marrow ablation in the rodent, including surgical procedure, anesthesia, and pre- and post-operative care. In addition, frequently used experimental end points are briefly discussed. This model was developed to study intramembranous bone regeneration following surgical disruption of the marrow contents of long bones. In this model, the timing of the appearance of bone formation and remodeling is well-characterized and therefore the model is well-suited to evaluate the in vivo effects of various agents which influence these processes. When biomaterials such as tissue engineering scaffolds or metal implants are placed in the medullary cavity after marrow ablation, end points relevant to tissue engineering and implant fixation can also be analyzed. By sharing a detailed protocol, we hope to improve inter-laboratory reproducibility.
Collapse
|
46
|
Brennen WN, Kisteman LN, Isaacs JT. Rapid selection of mesenchymal stem and progenitor cells in primary prostate stromal cultures. Prostate 2016; 76:552-64. [PMID: 26732992 PMCID: PMC4856028 DOI: 10.1002/pros.23145] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Carcinoma-associated fibroblasts (CAFs) are a dominant component of the tumor microenvironment with pro-tumorigenic properties. Despite this knowledge, their physiologic origins remain poorly understood. Mesenchymal stem cells (MSCs) can be recruited from the bone marrow to areas of tissue damage and inflammation, including prostate cancer. MSCs can generate and have many overlapping properties with CAFs in preclinical models. METHODS Multiparameter flow cytometry and multipotent differentiation assays used to define MSCs in primary prostate stromal cultures derived from young (<25 yrs) organ donors and prostate cancer patients compared with bone marrow-derived stromal cultures. Population doubling times, population doublings, cell size, and differentiation potential determined under multiple culture conditions, including normoxia, hypoxia, and a variety of media. TGF-β measured by ELISA. RESULTS MSCs and stromal progenitors are not only present in normal and malignant prostate tissue, but are quickly selected for in primary stromal cultures derived from these tissues; becoming the dominant population within just a few passages. Growth potential inversely associated with TGF-β concentrations. All conditions generated populations with an average cell diameter >15 µm. All cultures tested had the ability to undergo osteogenic and chondrogenic differentiation, but unlike bone marrow-derived MSCs, primary stromal cultures derived from normal prostate tissue lack adipogenic differentiation potential. In contrast, a subset of stromal cultures derived from prostate cancer patients retain the ability to differentiate into adipocytes; a property that is significantly suppressed under hypoxic conditions in both bone marrow- and prostate-derived MSCs. CONCLUSIONS Primary prostate stromal cultures are highly enriched in cells with an MSC or stromal progenitor phenotype. The use of primary cultures such as these to study CAFs raises interesting implications when considering their overlapping properties. The lack of adipogenesis in stromal cultures derived from normal prostates suggests they have a lineage-restricted progenitor phenotype. The retention of adipogenic differentiation in cultures from a subset of prostate cancer patients suggests the active recruitment of less committed progenitors or MSCs from the bone marrow as a function of disease progression. This recruitment can potentially be exploited for prognostic purposes or a cell-based platform for the systemic delivery of cytotoxic agents to sites of prostate cancer.
Collapse
Affiliation(s)
- W. Nathaniel Brennen
- Correspondence to: W. Nathaniel Brennen, Department of Oncology, Prostate Cancer Program, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1650 Orleans St., CRB-I, Rm 1M87, Baltimore, MD 21287.
| | | | | |
Collapse
|
47
|
Current View on Osteogenic Differentiation Potential of Mesenchymal Stromal Cells Derived from Placental Tissues. Stem Cell Rev Rep 2016; 11:570-85. [PMID: 25381565 PMCID: PMC4493719 DOI: 10.1007/s12015-014-9569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells (MSC) isolated from human term placental tissues possess unique characteristics, including their peculiar immunomodulatory properties and their multilineage differentiation potential. The osteogenic differentiation capacity of placental MSC has been widely disputed, and continues to be an issue of debate. This review will briefly discuss the different MSC populations which can be obtained from different regions of human term placenta, along with their unique properties, focusing specifically on their osteogenic differentiation potential. We will present the strategies used to enhance osteogenic differentiation potential in vitro, such as through the selection of subpopulations more prone to differentiate, the modification of the components of osteo-inductive medium, and even mechanical stimulation. Accordingly, the applications of three-dimensional environments in vitro and in vivo, such as non-synthetic, polymer-based, and ceramic scaffolds, will also be discussed, along with results obtained from pre-clinical studies of placental MSC for the regeneration of bone defects and treatment of bone-related diseases.
Collapse
|
48
|
Almeida CR, Caires HR, Vasconcelos DP, Barbosa MA. NAP-2 Secreted by Human NK Cells Can Stimulate Mesenchymal Stem/Stromal Cell Recruitment. Stem Cell Reports 2016; 6:466-473. [PMID: 27052313 PMCID: PMC4834048 DOI: 10.1016/j.stemcr.2016.02.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 12/21/2022] Open
Abstract
Strategies for improved homing of mesenchymal stem cells (MSCs) to a place of injury are being sought and it has been shown that natural killer (NK) cells can stimulate MSC recruitment. Here, we studied the chemokines behind this recruitment. Assays were performed with bone marrow human MSCs and NK cells freshly isolated from healthy donor buffy coats. Supernatants from MSC-NK cell co-cultures can induce MSC recruitment but not to the same extent as when NK cells are present. Antibody arrays and ELISA assays confirmed that NK cells secrete RANTES (CCL5) and revealed that human NK cells secrete NAP-2 (CXCL7), a chemokine that can induce MSC migration. Inhibition with specific antagonists of CXCR2, a receptor that recognizes NAP-2, abolished NK cell-mediated MSC recruitment. This capacity of NK cells to produce chemokines that stimulate MSC recruitment points toward a role for this immune cell population in regulating tissue repair/regeneration. Primary unstimulated human NK cells produce NAP-2 (CXCL7) NAP-2 is a chemokine that can promote recruitment of bone marrow MSCs Inhibiting the NAP-2 receptor CXCR2 abolishes NK cell-mediated MSC recruitment
Collapse
Affiliation(s)
- Catarina R Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.
| | - Hugo R Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Daniela P Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Mário A Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
49
|
Ge T, Yu Q, Liu W, Cong L, Liu L, Wang Y, Zhou L, Lin D. Characterization of bone marrow-derived mesenchymal stem cells from dimethyloxallyl glycine-preconditioned mice: Evaluation of the feasibility of dimethyloxallyl glycine as a mobilization agent. Mol Med Rep 2016; 13:3498-506. [PMID: 26935134 PMCID: PMC4805059 DOI: 10.3892/mmr.2016.4945] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 01/27/2016] [Indexed: 12/23/2022] Open
Abstract
The prolyl hydroxylase inhibitor dimethyloxallyl glycine (DMOG) has been increasingly studied with regards to stem cell therapy. Previous studies have demonstrated that endogenous mesenchymal stem cells (MSCs) may be mobilized into peripheral circulation by pharmaceutical preconditioning. In addition, our previous study confirmed that DMOG, as a novel mobilization agent, could induce mouse/rat MSC migration into peripheral blood circulation. Therefore, the present study conducted studies to characterize bone marrow-derived MSCs (BM-MSCs) collected from mice following DMOG intraperitoneal injection. The surface antigen immune phenotype, differentiation capability, proliferative ability, migratory capacity and paracrine capacity of the BM-MSCs collected from DMOG-preconditioned mice (DBM-MSCs) or normal saline-treated mice (NBM-MSCs) were evaluated by means of flow cytometry, differentiation induction, Cell Counting kit-8, Transwell assay and enzyme-linked immunosorbent assay, respectively. Compared with NBM-MSCs, DBM-MSCs displayed a similar immune phenotype and multilineage differentiation capability, reduced proliferative ability and migratory capacity, and similar transforming growth factor and platelet-derived growth factor secretion capacity. These results provide a novel insight into the biological properties of BM-MSCs from mice preconditioned with DMOG. DBM-MSCs exhibited slightly distinct characteristics to NBM-MSCs; however, they may have therapeutic potential for future stem cell therapy. In addition, the present study suggested that DMOG may be used as a novel mobilization agent in future clinical trials as no adverse effects were observed.
Collapse
Affiliation(s)
- Tingting Ge
- Department of Bioengineering, College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Qin Yu
- Department of Bioengineering, College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Wei Liu
- Department of Bioengineering, College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Li Cong
- Department of Pediatrics, The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Lizhen Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Medical School of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yan Wang
- Department of Pediatrics, The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Liping Zhou
- Department of Bioengineering, College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Deju Lin
- Department of Bioengineering, College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
50
|
Mele L, Vitiello PP, Tirino V, Paino F, De Rosa A, Liccardo D, Papaccio G, Desiderio V. Changing Paradigms in Cranio-Facial Regeneration: Current and New Strategies for the Activation of Endogenous Stem Cells. Front Physiol 2016; 7:62. [PMID: 26941656 PMCID: PMC4764712 DOI: 10.3389/fphys.2016.00062] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022] Open
Abstract
Craniofacial area represent a unique district of human body characterized by a very high complexity of tissues, innervation and vascularization, and being deputed to many fundamental function such as eating, speech, expression of emotions, delivery of sensations such as taste, sight, and earing. For this reasons, tissue loss in this area following trauma or for example oncologic resection, have a tremendous impact on patients' quality of life. In the last 20 years regenerative medicine has emerged as one of the most promising approach to solve problem related to trauma, tissue loss, organ failure etc. One of the most powerful tools to be used for tissue regeneration is represented by stem cells, which have been successfully implanted in different tissue/organs with exciting results. Nevertheless, both autologous and allogeneic stem cell transplantation raise many practical and ethical concerns that make this approach very difficult to apply in clinical practice. For this reason different cell free approaches have been developed aiming to the mobilization, recruitment, and activation of endogenous stem cells into the injury site avoiding exogenous cells implant but instead stimulating patients' own stem cells to repair the lesion. To this aim many strategies have been used including functionalized bioscaffold, controlled release of stem cell chemoattractants, growth factors, BMPs, Platelet-Rich-Plasma, and other new strategies such as ultrasound wave and laser are just being proposed. Here we review all the current and new strategies used for activation and mobilization of endogenous stem cells in the regeneration of craniofacial tissue.
Collapse
Affiliation(s)
- Luigi Mele
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Pietro Paolo Vitiello
- Medical Oncology, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara," Second University of Naples Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Francesca Paino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Alfredo De Rosa
- Department of Odontology and Surgery, Second University of Naples Naples, Italy
| | - Davide Liccardo
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| |
Collapse
|