1
|
Wang TF, Liou YS, Yang SH, Lin GL, Chiang YW, Lien TS, Li CC, Wang JH, Chang HH, Sun DS. Platelet-derived circulating soluble P-selectin is sufficient to induce hematopoietic stem cell mobilization. Stem Cell Res Ther 2023; 14:300. [PMID: 37864264 PMCID: PMC10589967 DOI: 10.1186/s13287-023-03527-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 10/09/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Granulocyte colony-stimulating factor (G-CSF)-mediated mobilization of hematopoietic stem cells (HSCs) is a well-established method to prepare HSCs for transplantation nowadays. A sufficient number of HSCs is critical for successful HSC transplantation. However, approximately 2-6% of healthy stem cell donors are G-CSF-poor mobilizers for unknown reasons; thus increasing the uncertainties of HSC transplantation. The mechanism underlining G-CSF-mediated HSC mobilization remains elusive, so detailed mechanisms and an enhanced HSC mobilization strategy are urgently needed. Evidence suggests that P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) are one of the cell-cell adhesion ligand-receptor pairs for HSCs to keep contacting bone marrow (BM) stromal cells before being mobilized into circulation. This study hypothesized that blockage of PSGL-1 and P-selectin may disrupt HSC-stromal cell interaction and facilitate HSC mobilization. METHODS The plasma levels of soluble P-selectin (sP-sel) before and after G-CSF administration in humans and male C57BL/6J mice were analyzed using enzyme-linked immunosorbent assay. Male mice with P-selectin deficiency (Selp-/-) were further employed to investigate whether P-selectin is essential for G-CSF-induced HSC mobilization and determine which cell lineage is sP-sel derived from. Finally, wild-type mice were injected with either G-CSF or recombinant sP-sel to investigate whether sP-sel alone is sufficient for inducing HSC mobilization and whether it accomplishes this by binding to HSCs and disrupting their interaction with stromal cells in the BM. RESULTS A significant increase in plasma sP-sel levels was observed in humans and mice following G-CSF administration. Treatments of G-CSF induced a decrease in the level of HSC mobilization in Selp-/- mice compared with the wild-type (Selp+/+) controls. Additionally, the transfer of platelets derived from wild-type mice can ameliorate the defected HSC mobilization in the Selp-/- recipients. G-CSF induces the release of sP-sel from platelets, which is sufficient to mobilize BM HSCs into the circulation of mice by disrupting the PSGL-1 and P-selectin interaction between HSCs and stromal cells. These results collectively suggested that P-selectin is a critical factor for G-CSF-induced HSC mobilization. CONCLUSIONS sP-sel was identified as a novel endogenous HSC-mobilizing agent. sP-sel injections achieved a relatively faster and more convenient regimen to mobilize HSCs in mice than G-CSF. These findings may serve as a reference for developing and optimizing human HSC mobilization in the future.
Collapse
Grants
- MOST103-2321-B-320-001 Ministry of Science and Technology, Taiwan
- MOST105-2633-B-320-001 Ministry of Science and Technology, Taiwan
- MOST106-2633-B-320-001 Ministry of Science and Technology, Taiwan
- MOST108-2311-B-320-001 Ministry of Science and Technology, Taiwan
- TCMMP104-06 Buddhist Tzu Chi Medical Foundation
- TCMMP108-04 Buddhist Tzu Chi Medical Foundation
- TCMMP111-01 Buddhist Tzu Chi Medical Foundation
- TCRD106-42 Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- TCRD108-55 Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- TCRD110-61 Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- TCRD111-082 Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- TCRD112-054 Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- TCAS-112-02 Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
Collapse
Affiliation(s)
- Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan, Republic of China
- Buddhist Tzu Chi Stem Cells Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Yu-Shan Liou
- Department of Molecular Biology and Human Genetics, College of Medicine, Tzu Chi University, No. 701, Section 3, Zhong-Yang Road, Hualien, 97004, Taiwan, Republic of China
| | - Shang-Hsien Yang
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan, Republic of China
- Buddhist Tzu Chi Stem Cells Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
- Department of Pediatric Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Guan-Ling Lin
- Department of Molecular Biology and Human Genetics, College of Medicine, Tzu Chi University, No. 701, Section 3, Zhong-Yang Road, Hualien, 97004, Taiwan, Republic of China
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Ya-Wen Chiang
- Department of Molecular Biology and Human Genetics, College of Medicine, Tzu Chi University, No. 701, Section 3, Zhong-Yang Road, Hualien, 97004, Taiwan, Republic of China
| | - Te-Sheng Lien
- Department of Molecular Biology and Human Genetics, College of Medicine, Tzu Chi University, No. 701, Section 3, Zhong-Yang Road, Hualien, 97004, Taiwan, Republic of China
| | - Chi-Cheng Li
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
- Center of Stem Cell and Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Jen-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, College of Medicine, Tzu Chi University, No. 701, Section 3, Zhong-Yang Road, Hualien, 97004, Taiwan, Republic of China.
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, College of Medicine, Tzu Chi University, No. 701, Section 3, Zhong-Yang Road, Hualien, 97004, Taiwan, Republic of China.
| |
Collapse
|
2
|
Chang HH, Liou YS, Sun DS. Hematopoietic stem cell mobilization. Tzu Chi Med J 2022; 34:270-275. [PMID: 35912054 PMCID: PMC9333105 DOI: 10.4103/tcmj.tcmj_98_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 06/25/2021] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation has been used to treat hematopoietic diseases for over 50 years. HSCs can be isolated from bone marrow (BM), umbilical cord blood, or peripheral blood. Because of lower costs, shorter hospitalization, and faster engraftment, peripheral blood has become the predominant source of HSCs for transplantation. The major factors determining the rate of successful HSC transplantation include the degree of human leukocyte antigen matching between the donor and recipient and the number of HSCs for transplantation. Administration of granulocyte colony-stimulating factor (G-CSF) alone or combined with plerixafor (AMD3100) are clinical used methods to promote HSC mobilization from BM to the peripheral blood for HSC transplantations. However, a significant portion of healthy donors or patients may be poor mobilizers of G-CSF, resulting in an insufficient number of HSCs for the transplantation and necessitating alternative strategies to increase the apheresis yield. The detailed mechanisms underlying G-CSF-mediated HSC mobilization remain to be elucidated. This review summarizes the current research on deciphering the mechanism of HSC mobilization.
Collapse
Affiliation(s)
- Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Yu-Shan Liou
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
3
|
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
|
4
|
Daw S, Law S. The functional interplay of transcription factors and cell adhesion molecules in experimental myelodysplasia including hematopoietic stem progenitor compartment. Mol Cell Biochem 2020; 476:535-551. [PMID: 33011884 DOI: 10.1007/s11010-020-03920-6] [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: 06/19/2020] [Accepted: 09/19/2020] [Indexed: 12/30/2022]
Abstract
Myelodysplastic syndrome is a heterogenous group of disorder with clonal dysregulated hematopoiesis characterized by bone marrow failure, cytogenetic and molecular abnormalities and variable risk of progression to acute myeloid leukemia (AML). The bone marrow niche plays a major role in maintaining the homeostasis and is often injured by the chemotherapeutic drugs leading to catastrophic consequences like myelodysplastic syndrome. In the present study, we made an attempt to find out the osteoblastic niche related alterations in the myelodysplastic bone marrow through mainly flowcytometric and fluorescent microscopic studies. We have also checked the condition of the myelodysplastic bone through micro computed tomography. The results revealed that the affected osteoblasts of the myelodysplastic bone marrow compelled the hematopoietic stem cell to come out of quiescence and become actively proliferating, and in this scenario the decline in expression of cell adhesion molecules like N-Cadherin, Intercellular adhesion molecule 1 (ICAM) and upregulated focal adhesion kinase (FAK) played a major role. The hike in number of osteoclasts in myelodysplastic cases than control also shattered the balance between bone formation and resorption ratio. We have recorded a dysregulated expression of transcription factors GATA2 and CEBPα (CCAAT-enhancer-binding-protein) in the hematopoietic stem progenitor compartment of the myelodysplastic bone marrow, the main reason behind the presence of abnormal myeloblasts in myelodysplastic cases. Collectively, we can say the coordinated perturbations in the osteoblastic niche, cell adhesion molecules together with the transcription factors has resulted in the uncontrolled proliferation of hematopoietic stem cell, dysregulated myelopoiesis, early trafficking of hematopoietic progenitors to blood compartment and at the same time pancytopenic peripheral blood conditions during the progression of N-Ethyl N Nitroso Urea (ENU) induced myelodysplasia.
Collapse
Affiliation(s)
- Suchismita Daw
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical, Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata, 700073, West Bengal, India
| | - Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical, Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata, 700073, West Bengal, India.
| |
Collapse
|
5
|
Zhou B, Ge T, Zhou L, Jiang L, Zhu L, Yao P, Yu Q. Dimethyloxalyl Glycine Regulates the HIF-1 Signaling Pathway in Mesenchymal Stem Cells. Stem Cell Rev Rep 2020; 16:702-710. [DOI: 10.1007/s12015-019-09947-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
6
|
Moll G, Drzeniek N, Kamhieh-Milz J, Geissler S, Reinke P. Editorial comment: variables affecting the presence of mesenchymal stromal cells in the peripheral blood and their relationship with apheresis product. Br J Haematol 2020; 189:593-596. [PMID: 32037526 DOI: 10.1111/bjh.16389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Norman Drzeniek
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Geissler
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany.,Julius Wolff Institute (JWI), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies (BECAT), all Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Hussien NI, Ebrahim N, Mohammed OM, Sabry D. Combination of Obestatin and Bone Marrow Mesenchymal Stem Cells Prevents Aggravation of Endocrine Pancreatic Damage in Type II Diabetic Rats. Int J Stem Cells 2017; 10:129-143. [PMID: 29186652 PMCID: PMC5741194 DOI: 10.15283/ijsc17035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2017] [Indexed: 12/16/2022] Open
Abstract
One of the new promising therapies in treatment of diabetes mellitus is mesenchymal stem cells (MSCs) which have an interesting therapeutic potentiality based on their paracrine effect and transdifferentiation potentiality. Also obestatin improves the generation of functional β cells/islet-like cell clusters in vitro, suggesting implications for cell-based replacement therapy in diabetes. So the aim of this study was to evaluate the effect of combination of both MSCs and obestatin on an experimental model of type II diabetes mellitus (T2DM). Sixty male rats were divided into; group I (control group), group II (T2DM group) induced by administration of high fat diet (HFD) and injection of streptozotocin (STZ) in low dose, group III (T2DM treated with MSCs), group IV (T2DM treated with obestatin), group V (T2DM treated with MSCs and obestatin). Fasting blood glucose, C-peptide, insulin and lipid profile were measured. HOMA-IR and HOMA-β were calculated. Pancreatic expression of insulin, glucagon like peptide -1 (GLP-1) and pancreatic duodenal homeobox 1 (Pdx1) mRNA levels were measured. In addition pancreatic histological changes, insulin and Bax were analyzed by immunohistochemical examination of islets of Langerhans. Diabetic rats showed significant increase in HOMA-IR, serum glucose and lipid profile levels with significant decrease in insulin, HOMA-β, GLP-1 and Pdx1 levels. MSCs and obestatin caused significant improvement in all parameters with more significant improvement in combined therapy. The protective effects afforded by MSCs and obestatin may derive from improvement of the metabolic profile, antiapoptosis and by increase in pancreatic GLP-1and Pdx1 gene expression.
Collapse
Affiliation(s)
- Noha I Hussien
- Department of Physiology, Faculty of Medicine, Benha University, Cairo, Egypt
| | - Nesrine Ebrahim
- Department of Histology, Faculty of Medicine, Benha University, Cairo, Egypt
| | - Ola M Mohammed
- Department of Histology, Faculty of Medicine, Benha University, Cairo, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
Gilevich IV, Fedorenko TV, Pashkova IA, Porkhanov VA, Chekhonin VP. Effects of Growth Factors on Mobilization of Mesenchymal Stem Cells. Bull Exp Biol Med 2017; 162:684-686. [DOI: 10.1007/s10517-017-3687-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Indexed: 01/13/2023]
|
13
|
Dunne PD, O'Reilly PG, Coleman HG, Gray RT, Longley DB, Johnston PG, Salto-Tellez M, Lawler M, McArt DG. Stratified analysis reveals chemokine-like factor (CKLF) as a potential prognostic marker in the MSI-immune consensus molecular subtype CMS1 of colorectal cancer. Oncotarget 2016; 7:36632-36644. [PMID: 27153559 PMCID: PMC5095027 DOI: 10.18632/oncotarget.9126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/16/2016] [Indexed: 11/25/2022] Open
Abstract
The Colorectal Cancer (CRC) Subtyping Consortium (CRCSC) recently published four consensus molecular subtypes (CMS's) representing the underlying biology in CRC. The Microsatellite Instable (MSI) immune group, CMS1, has a favorable prognosis in early stage disease, but paradoxically has the worst prognosis following relapse, suggesting the presence of factors enabling neoplastic cells to circumvent this immune response. To identify the genes influencing subsequent poor prognosis in CMS1, we analyzed this subtype, centered on risk of relapse. In a cohort of early stage colon cancer (n=460), we examined, in silico, changes in gene expression within the CMS1 subtype and demonstrated for the first time the favorable prognostic value of chemokine-like factor (CKLF) gene expression in the adjuvant disease setting [HR=0.18, CI=0.04-0.89]. In addition, using transcription profiles originating from cell sorted CRC tumors, we delineated the source of CKLF transcription within the colorectal tumor microenvironment to the leukocyte component of these tumors. Further to this, we confirmed that CKLF gene expression is confined to distinct immune subsets in whole blood samples and primary cell lines, highlighting CKLF as a potential immune cell-derived factor promoting tumor immune-surveillance of nascent neoplastic cells, particularly in CMS1 tumors. Building on the recently reported CRCSC data, we provide compelling evidence that leukocyte-infiltrate derived CKLF expression is a candidate biomarker of favorable prognosis, specifically in MSI-immune stage II/III disease.
Collapse
Affiliation(s)
- Philip D. Dunne
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Paul G. O'Reilly
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Helen G. Coleman
- Centre for Public Health, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Ronan T. Gray
- Centre for Public Health, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Daniel B. Longley
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Patrick G. Johnston
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Manuel Salto-Tellez
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Mark Lawler
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Darragh G. McArt
- Centre for Cancer Research and Cell Biology, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| |
Collapse
|
14
|
The angiogenic related functions of bone marrow mesenchymal stem cells are promoted by CBDL rat serum via the Akt/Nrf2 pathway. Exp Cell Res 2016; 344:86-94. [PMID: 27105936 DOI: 10.1016/j.yexcr.2016.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/01/2016] [Accepted: 04/17/2016] [Indexed: 12/22/2022]
Abstract
Hepatopulmonary syndrome (HPS) is a complication of severe liver disease. It is characterized by an arterial oxygenation defect. Recent studies have demonstrated that pulmonary angiogenesis contributes to the abnormal gas exchange found in HPS. Additionally, mesenchymal stem cells (MSCs) are considered the stable source of VEGF-producing cells and have the potential to differentiate into multiple cell types. However, it has not been determined whether bone marrow mesenchymal stem cells (BM-MSCs) are mobilized and involved in the pulmonary angiogenesis in HPS. In this study, a CFU-F assay showed that the number of peripheral blood MSCs was increased in common bile duct ligation (CBDL) rats; however, there was no significant difference found in the number of BM-MSCs. In vitro, CBDL rat serum induced the overexpression of CXCR4 and PCNA in BM-MSCs. Consistently, the directional migration as well as the proliferation ability of BM-MSCs were enhanced by CBDL rat serum, as determined by a transwell migration and MTT assays. Moreover, the secretion of VEGF by BM-MSCs increased after treatment with CBDL rat serum. We also found that the expression of phospho-Akt, phospho-ERK, and Nrf2 in BM-MSCs was significantly up-regulated by CBDL rat serum in a time dependent manner, and the blockage of the Akt/Nrf2 signalling pathway with an Akt Inhibitor or Nrf2 siRNA, instead of an ERK inhibitor, attenuated the migration, proliferation and paracrine capacity of BM-MSCs. In conclusion, these findings indicated that the number of MSCs increased in the peripheral blood of CBDL rats, and the Akt/Nrf2 pathway plays a vital role in promoting the angiogenic related functions of BM-MSCs, which could be a potent contributor to pulmonary angiogenesis in HPS.
Collapse
|
15
|
Human Very Small Embryonic-Like Stem Cells Are Present in Normal Peripheral Blood of Young, Middle-Aged, and Aged Subjects. Stem Cells Int 2015; 2016:7651645. [PMID: 26633977 PMCID: PMC4655065 DOI: 10.1155/2016/7651645] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/27/2015] [Indexed: 01/10/2023] Open
Abstract
The purpose of our study was to determine whether the number of human very small embryonic-like stem cells (huVSELs) would vary depending on the age of humans. HuVSELs frequency was evaluated into the steady-state (SS) peripheral blood (PB) of healthy volunteers using flow cytometry analysis. Their numbers were compared with volunteers' age. Blood samples were withdrawn from 28 volunteers (age ranging from 20 to 70 years), who were distributed among three groups of age: “young” (mean age, 27.8 years), “middle” (mean age, 49 years), and “older” (mean age, 64.2 years). Comparing the three groups, we did not observe any statistically significant difference in huVSELs numbers between them. The difference in mRNA expression for PSC markers as SSEA-4, Oct-4, Nanog, and Sox2 between the three groups of age was not statistically significant. A similar frequency of huVSELs into the SS-PB of young, middle-aged, and aged subjects may indicate that the VSELs pool persists all along the life as a reserve for tissue repair in case of minor injury and that there is a continuous efflux of these cells from the BM into the PB.
Collapse
|
16
|
Bueno PDG, Yochite JNU, Derigge-Pisani GF, Malmegrim de Farias KCR, de Avó LRDS, Voltarelli JC, Leal ÂMDO. Metabolic and pancreatic effects of bone marrow mesenchymal stem cells transplantation in mice fed high-fat diet. PLoS One 2015; 10:e0124369. [PMID: 25923733 PMCID: PMC4414281 DOI: 10.1371/journal.pone.0124369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/27/2015] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to investigate the effects of multiple infusions of allogeneic MSCs on glucose homeostasis and morphometry of pancreatic islets in high- fat diet (HFD) fed mice. Swiss mice were fed standard diet (C group) or HFD (HFD group). After 8 weeks, animals of HFD group received sterile phosphate-buffered saline infusions (HFD-PBS) or four infusions of MSCs one week apart (HFD-MSCs). Fasting glycemia (FG) was determined weekly and glucose (GTT) and insulin (ITT) tolerance tests were performed 4, 8, 12, and 16 weeks after the infusions of MSCs. The MSCs transplanted mice were classified as responder (FG < 180 mg/dL, 72.2% of transplanted mice) or non-responder (FG > 180mg/dL, 28.8%) Seven weeks after MSCs infusions, FG decreased in HFD-MSCs responder mice compared with the HFD-PBS group. Sixteen weeks post MSCs infusions, GTT and ITT areas under the curve (AUC) decreased in HFD-MSCs responder mice compared to HFD-PBS group. Serum insulin concentration was higher in HFD-PBS group than in control animals and was not different compared with the other groups. The relative volume of α-cells was significantly smaller in HFD-PBS group than in C group and significantly higher in HFD-MSCs-NR than in HFD-PBS and HFD-MSCs-R groups. Cell apoptosis in the islets was higher in HFD-PBS group than in C group, and lower in HFD-MSCs responder mice than in HFD-PBS group and non-responder animals. The results demonstrate the ability of multiple infusions of MSCs to promote prolonged decrease in hyperglycemia and apoptosis in pancreatic islets and increase in insulin sensitivity in HFD fed mice.
Collapse
Affiliation(s)
- Patricia de Godoy Bueno
- Department of Physiological Science, Center of Biological Sciences and Health, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Juliana Navarro Ueda Yochite
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Graziela Fernanda Derigge-Pisani
- Department of Physiological Science, Center of Biological Sciences and Health, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Kelen Cristina Ribeiro Malmegrim de Farias
- Department of Clinical, Toxicological and Bromatological Analyses, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | | | | | | |
Collapse
|
17
|
Zheng F, Cheng L, Yu Q, Liu Q, Cheng F. The Primary Study of CD90(+)CD34(-)and Sca-1(+) Stem Cells Mobilized by EPO Plus G-CSF in Mice. Int J Stem Cells 2014; 2:129-34. [PMID: 24855532 DOI: 10.15283/ijsc.2009.2.2.129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2009] [Indexed: 12/27/2022] Open
Abstract
To investigate effects of recombinant human erythropoietin (rhEPO) plus recombinant human granulocyte colony stimulating factor (rhG-CSF) on mobilization of CD90(+)CD34(-) and Sca-1(+) stem cells in mice. rhEPO 1000 IU·kg(-1)·d(-1) per mice alone or plus rhG-CSF 250 μg·kg(-1)·d-1(-1)per mice was administered to mice for five days, the peripheral blood was collected at 6 hours, 1, 3,5 and 7 days after the last administration. The number of white blood cells (WBC) and mononuclear cells (MNC) was counted. The level of CD34, CD90 and Sca-1 mRNA were detected by reverse transcription-polymerase chain reaction (RT-PCR).The expressions of CD90(+)CD34(-) in absolute nuclear cells of peripheral blood was detected by flow cytometry. The results indicate that compared to mobilizing by rhG-CSF, rhEPO or plus rhG-CSF did not significantly enhance the number of WBC and MNC, on the fifth and the seventh day after the last administration, both the expression of CD34,CD90 and Sca-1 mRNA and the proportion of CD90(+)CD34(-) cells dramatically raised in rhEPO or in addition to rhG-CSF groups. We conclude that rhEPO or plus rhG-CSF had a strong capacity of mobilization of stem cells. The alteration of expression of CD34, CD90 and Sca-1 mRNA after the administration of rhEPO or combined with rhG-CSF indicated that this agents might potentially alter the peripheral blood graft content.
Collapse
Affiliation(s)
- Fang Zheng
- Institute of Hematology, Dongfeng Hospital affiliated to Yunyang Medical college, 16 Daling Avenue, Shiyan 442008, China. ; Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Long Cheng
- Institute of Hematology, Dongfeng Hospital affiliated to Yunyang Medical college, 16 Daling Avenue, Shiyan 442008, China
| | - Qiang Yu
- Institute of Hematology, Dongfeng Hospital affiliated to Yunyang Medical college, 16 Daling Avenue, Shiyan 442008, China
| | - Qihuan Liu
- Institute of Hematology, Dongfeng Hospital affiliated to Yunyang Medical college, 16 Daling Avenue, Shiyan 442008, China
| | - Fanjun Cheng
- Institute of Hematology, Dongfeng Hospital affiliated to Yunyang Medical college, 16 Daling Avenue, Shiyan 442008, China
| |
Collapse
|
18
|
Lymphohematopoietic cancers induced by chemicals and other agents and their implications for risk evaluation: An overview. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 761:40-64. [PMID: 24731989 DOI: 10.1016/j.mrrev.2014.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/13/2022]
Abstract
Lymphohematopoietic neoplasia are one of the most common types of cancer induced by therapeutic and environmental agents. Of the more than 100 human carcinogens identified by the International Agency for Research on Cancer, approximately 25% induce leukemias or lymphomas. The objective of this review is to provide an introduction into the origins and mechanisms underlying lymphohematopoietic cancers induced by xenobiotics in humans with an emphasis on acute myeloid leukemia, and discuss the implications of this information for risk assessment. Among the agents causing lymphohematopoietic cancers, a number of patterns were observed. Most physical and chemical leukemia-inducing agents such as the therapeutic alkylating agents, topoisomerase II inhibitors, and ionizing radiation induce mainly acute myeloid leukemia through DNA-damaging mechanisms that result in either gene or chromosomal mutations. In contrast, biological agents and a few immunosuppressive chemicals induce primarily lymphoid neoplasms through mechanisms that involve alterations in immune response. Among the environmental agents examined, benzene was clearly associated with acute myeloid leukemia in humans, with increasing but still limited evidence for an association with lymphoid neoplasms. Ethylene oxide and 1,3-butadiene were linked primarily to lymphoid cancers. Although the association between formaldehyde and leukemia remains controversial, several recent evaluations have indicated a potential link between formaldehyde and acute myeloid leukemia. The four environmental agents examined in detail were all genotoxic, inducing gene mutations, chromosomal alterations, and/or micronuclei in vivo. Although it is clear that rapid progress has been made in recent years in our understanding of leukemogenesis, many questions remain for future research regarding chemically induced leukemias and lymphomas, including the mechanisms by which the environmental agents reviewed here induce these diseases and the risks associated with exposures to such agents.
Collapse
|
19
|
Zhang C, Chen XH, Gao L, Liu Y, Gao L, Kong PY, Zeng DF, Peng XG, Sun AH, Zhang X. High-dose methotrexate in the mobilization of hematopoietic stem cells for patients with non-Hodgkin's lymphoma: a twelve-year study in a single center. Transfusion 2013; 54:1251-5. [PMID: 24329982 DOI: 10.1111/trf.12516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/20/2013] [Accepted: 08/23/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (auto-HSCT) is a promising approach for non-Hodgkin's lymphoma (NHL). Higher cell doses have been associated with a faster blood count recovery and a reduction in transfusion requirements, infection rates, and hospitalization times. Mobilization failure constitutes one of the main reasons for avoiding auto-HSCT. The role of high-dose methotrexate (MTX) as mobilization regimen is still unclear. STUDY DESIGN AND METHODS The effect of high-dose MTX as a mobilization regimen for 67 adult patients with NHL who received auto-HSCT was studied between January 2001 and October 2012. The stem cells were mobilized using combination chemotherapy including MTX plus granulocyte-colony-stimulating factor (G-CSF) in 33 patients (Group A), and the stem cells of the other 34 patients were mobilized using the same combination chemotherapy plus G-CSF without MTX (Group B). RESULTS All of the patients were successfully mobilized in Group A; however, two patients failed in Group B. The median numbers of CD34+ cells collected were 14.36 × 10(6) and 5.3 × 10(6) cells/kg for Groups A and B, respectively (p < 0.05). All of the patients experienced a stable neutrophil and platelet (PLT) engraftment. The times to white blood cell engraftment were 8.0 days in Group A and 11.0 days in Group B, and the times to PLT engraftment were 12.0 days in Group A and 13.0 days in Group B (p < 0.05 for both variables). CONCLUSION High-dose MTX is a powerful regimen component for stem cell mobilization in adult patients with NHL.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Toupadakis CA, Granick JL, Sagy M, Wong A, Ghassemi E, Chung DJ, Borjesson DL, Yellowley CE. Mobilization of endogenous stem cell populations enhances fracture healing in a murine femoral fracture model. Cytotherapy 2013; 15:1136-47. [PMID: 23831362 DOI: 10.1016/j.jcyt.2013.05.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 03/26/2013] [Accepted: 05/08/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Delivery of bone marrow-derived stem and progenitor cells to the site of injury is an effective strategy to enhance bone healing. An alternate approach is to mobilize endogenous, heterogeneous stem cells that will home to the site of injury. AMD3100 is an antagonist of the chemokine receptor 4 (CXCR4) that rapidly mobilizes stem cell populations into peripheral blood. Our hypothesis was that increasing circulating numbers of stem and progenitor cells using AMD3100 will improve bone fracture healing. METHODS A transverse femoral fracture was induced in C57BL/6 mice, after which they were subcutaneously injected for 3 d with AMD3100 or saline control. Mesenchymal stromal cells, hematopoietic stem and progenitor cells and endothelial progenitor cells in the peripheral blood and bone marrow were evaluated by means of flow cytometry, automated hematology analysis and cell culture 24 h after injection and/or fracture. Healing was assessed up to 84 d after fracture by histomorphometry and micro-computed tomography. RESULTS AMD3100 injection resulted in higher numbers of circulating mesenchymal stromal cells, hematopoietic stem cells and endothelial progenitor cells. Micro-computed tomography data demonstrated that the fracture callus was significantly larger compared with the saline controls at day 21 and significantly smaller (remodeled) at day 84. AMD3100-treated mice have a significantly higher bone mineral density than do saline-treated counterparts at day 84. CONCLUSIONS Our data demonstrate that early cell mobilization had significant positive effects on healing throughout the regenerative process. Rapid mobilization of endogenous stem cells could provide an effective alternative strategy to cell transplantation for enhancing tissue regeneration.
Collapse
Affiliation(s)
- Chrisoula A Toupadakis
- Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
McNulty MA, Virdi AS, Christopherson KW, Sena K, Frank RR, Sumner DR. Adult stem cell mobilization enhances intramembranous bone regeneration: a pilot study. Clin Orthop Relat Res 2012; 470:2503-12. [PMID: 22528386 PMCID: PMC3830081 DOI: 10.1007/s11999-012-2357-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Stem cell mobilization, which is defined as the forced egress of stem cells from the bone marrow to the peripheral blood (PB) using chemokine receptor agonists, is an emerging concept for enhancing tissue regeneration. However, the effect of stem cell mobilization by a single injection of the C-X-C chemokine receptor type 4 (CXCR4) antagonist AMD3100 on intramembranous bone regeneration is unclear. QUESTIONS/PURPOSES We therefore asked: Does AMD3100 mobilize adult stem cells in C57BL/6 mice? Are stem cells mobilized to the PB after marrow ablation? And does AMD3100 enhance bone regeneration? METHODS Female C57BL/6 mice underwent femoral marrow ablation surgery alone (n = 25), systemic injection of AMD3100 alone (n = 15), or surgery plus AMD3100 (n = 57). We used colony-forming unit assays, flow cytometry, and micro-CT to investigate mobilization of mesenchymal stem cells, endothelial progenitor cells, and hematopoietic stem cells to the PB and bone regeneration. RESULTS AMD3100 induced mobilization of stem cells to the PB, resulting in a 40-fold increase in mesenchymal stem cells. The marrow ablation injury mobilized all three cell types to the PB over time. Administration of AMD3100 led to a 60% increase in bone regeneration at Day 21. CONCLUSIONS A single injection of a CXCR4 antagonist lead to stem cell mobilization and enhanced bone volume in the mouse marrow ablation model of intramembranous bone regeneration.
Collapse
Affiliation(s)
- Margaret A. McNulty
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | - Amarjit S. Virdi
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | | | - Kotaro Sena
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | - Robin R. Frank
- Division of Hematology & Oncology, Rush University Medical Center, Chicago, IL USA
| | - Dale R. Sumner
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| |
Collapse
|
22
|
Herbert KE, Gambell P, Link EK, Mouminoglu A, Wall DM, Harrison SJ, Ritchie DS, Seymour JF, Prince HM. Pegfilgrastim compared with filgrastim for cytokine-alone mobilization of autologous haematopoietic stem and progenitor cells. Bone Marrow Transplant 2012; 48:351-6. [PMID: 22858510 DOI: 10.1038/bmt.2012.145] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Haematopoietic stem and progenitor cells (HSPC) mobilization, using cytokine-alone, is a well-tolerated regimen with predictable mobilization kinetics. Single-dose pegfilgrastim mobilizes HSPC efficiently; however, there is surprisingly little comparative data on its use without chemotherapy for HSPC mobilization. Pegfilgrastim-alone and filgrastim-alone mobilization regimens were compared in 52 patients with haematological malignancy. Pegfilgrastim 12 mg (n=20) or 6 mg (n=2) was administered Day 1 (D1) in 22 patients (lymphoma n=17; myeloma n=5). Thirty historical controls (lymphoma n=18; myeloma n=12) received filgrastim 10 mcg/kg daily from D1. Peripheral blood (PB) CD34(+) counts reached threshold (5 × 10(6)/L) and apheresis commenced on D4(4-5) and D4(4-6). Median PB CD34(+) cell count on D1 of apheresis was similar (26.0 × 10(6)/L (2.5-125.0 × 10(6)/L) and 16.2 × 10(6)/L (2.6-50.7 × 10(6)/L); P=0.06), for pegfilgrastim and filgrastim groups, respectively. Target yield (2 × 10(6) per kg CD34(+) cells) was collected in 20/22 (91%) pegfilgrastim patients and 24/30 (80%) in the filgrastim group (P=0.44), in a similar median number of aphereses (3(1-4) versus 3(2-6), respectively; P=0.85). A higher proportion of pegfilgrastim patients tended to yield 4 × 10(6) per kg CD34(+) cells; 16/22 (73%) versus 14/30 (47%) filgrastim patients (P=0.09). One pegfilgrastim patient developed hyperleukocytosis that resolved without incident. Pegfilgrastim-alone is a simple, well-tolerated, and attractive option for outpatient-based HSPC mobilization with similar mobilization kinetics and efficacy to regular filgrastim.
Collapse
Affiliation(s)
- K E Herbert
- Department of Haematology and Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hematopoietic stem and progenitor cells as effectors in innate immunity. BONE MARROW RESEARCH 2012; 2012:165107. [PMID: 22762001 PMCID: PMC3385697 DOI: 10.1155/2012/165107] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 04/22/2012] [Accepted: 04/28/2012] [Indexed: 12/17/2022]
Abstract
Recent research has shed light on novel functions of hematopoietic stem and progenitor cells (HSPC). While they are critical for maintenance and replenishment of blood cells in the bone marrow, these cells are not limited to the bone marrow compartment and function beyond their role in hematopoiesis. HSPC can leave bone marrow and circulate in peripheral blood and lymph, a process often manipulated therapeutically for the purpose of transplantation. Additionally, these cells preferentially home to extramedullary sites of inflammation where they can differentiate to more mature effector cells. HSPC are susceptible to various pathogens, though they may participate in the innate immune response without being directly infected. They express pattern recognition receptors for detection of endogenous and exogenous danger-associated molecular patterns and respond not only by the formation of daughter cells but can themselves secrete powerful cytokines. This paper summarizes the functional and phenotypic characterization of HSPC, their niche within and outside of the bone marrow, and what is known regarding their role in the innate immune response.
Collapse
|
24
|
Liu L, Yu Q, Lin J, Lai X, Cao W, Du K, Wang Y, Wu K, Hu Y, Zhang L, Xiao H, Duan Y, Huang H. Hypoxia-Inducible Factor-1α Is Essential for Hypoxia-Induced Mesenchymal Stem Cell Mobilization into the Peripheral Blood. Stem Cells Dev 2011; 20:1961-71. [PMID: 21275821 DOI: 10.1089/scd.2010.0453] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Lizhen Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Yu
- Institute of Bioengineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Lin
- Institute of Bioengineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoyu Lai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijie Cao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaili Du
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingjia Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kangni Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifei Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haowen Xiao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanping Duan
- Institute of Bioengineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
25
|
Identification of Very Small Embryonic/Epiblast-Like Stem Cells (VSELs) Circulating in Peripheral Blood During Organ/Tissue Injuries. Methods Cell Biol 2011; 103:31-54. [DOI: 10.1016/b978-0-12-385493-3.00003-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
26
|
Higashino K, Viggeswarapu M, Bargouti M, Liu H, Titus L, Boden SD. Stromal cell-derived factor-1 potentiates bone morphogenetic protein-2 induced bone formation. Tissue Eng Part A 2010; 17:523-30. [PMID: 21043834 DOI: 10.1089/ten.tea.2010.0168] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The mechanisms driving bone marrow stem cell mobilization are poorly understood. A recent murine study found that circulating bone marrow-derived osteoprogenitor cells (MOPCs) were recruited to the site of recombinant human bone morphogenetic protein-2 (BMP-2)-induced bone formation. Stromal cell-derived factor-1α (SDF-1α) and its cellular receptor CXCR4 have been shown to mediate the homing of stem cells to injured tissues. We hypothesized that chemokines, such as SDF-1, are also involved with mobilization of bone marrow cells. The CD45(-) fraction is a major source of MOPCs. In this report we determined that the addition of BMP-2 or SDF-1 to collagen implants increased the number of MOPCs in the peripheral blood. BMP-2-induced mobilization was blocked by CXCR4 antibody, confirming the role of SDF-1 in mobilization. We determined for the first time that addition of SDF-1 to implants containing BMP-2 enhances mobilization, homing of MOPCs to the implant, and ectopic bone formation induced by suboptimal BMP-2 doses. These results suggest that SDF-1 increases the number of osteoprogenitor cells that are mobilized from the bone marrow and then home to the implant. Thus, addition of SDF-1 to BMP-2 may improve the efficiency of BMPs in vivo, making their routine use for orthopaedic applications more affordable and available to more patients.
Collapse
Affiliation(s)
- Kosaku Higashino
- Atlanta Veteran Affairs Medical Center and Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
27
|
Lévesque JP, Helwani FM, Winkler IG. The endosteal 'osteoblastic' niche and its role in hematopoietic stem cell homing and mobilization. Leukemia 2010; 24:1979-92. [PMID: 20861913 DOI: 10.1038/leu.2010.214] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The concept of hematopoietic stem cell (HSC) niche was formulated in 1978, but HSC niches remained unidentified for the following two decades largely owing to technical limitations. Sophisticated live microscopy techniques and genetic manipulations have identified the endosteal region of the bone marrow (BM) as a preferential site of residence for the most potent HSC - able to reconstitute in serial transplants - with osteoblasts and their progenitors as critical cellular elements of these endosteal niches. This article reviews the path to the discovery of these endosteal niches (often called 'osteoblastic' niches) for HSC, what cell types contribute to these niches with their known physical and biochemical features. In the past decade, a first wave of research uncovered many mechanisms responsible for HSC homing to, and mobilization from, the whole BM tissue. However, the recent discovery of endosteal HSC niches has initiated a second wave of research focusing on the mechanisms by which most primitive HSC lodge into and migrate out of their endosteal niches. The second part of this article reviews the current knowledge of the mechanisms of HSC lodgment into, retention in and mobilization from osteoblastic niches.
Collapse
Affiliation(s)
- J-P Lévesque
- Biotherapies Program, Haematopoietic Stem Cell Laboratory, Mater Medical Research Institute, South Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
28
|
Furuya Y, Okazaki Y, Kaji K, Sato S, Takehara K, Kuwana M. Mobilization of endothelial progenitor cells by intravenous cyclophosphamide in patients with systemic sclerosis. Rheumatology (Oxford) 2010; 49:2375-80. [PMID: 20724431 DOI: 10.1093/rheumatology/keq259] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE To evaluate the effects of i.v. CYC on the number of circulating endothelial progenitor cells (EPCs) in patients with SSc, and the potential association of the EPC response with CYC's effect for treating interstitial lung disease (ILD). METHODS This open-label, prospective study involved 12 patients with SSc and alveolitis (CYC group). All patients received six courses of i.v. CYC (0.5 g/m2) at 4-week intervals in combination with low-dose prednisolone. Ten patients were followed for 24 months. Seven SSc patients treated with low-dose prednisolone alone were used as a control for the EPC measurement (control group). Five patients with non-SSc CTD who received i.v. CYC and prednisolone also served as disease controls. EPCs were quantified by the partial enrichment of CD34+ cells followed by three-colour flow cytometry. The circulating levels of vascular injury markers were measured by immunoassay. RESULTS The EPC count was significantly increased at 2 weeks after treatment in the CYC group (P=0.02), but not in the control group, while CYC increased EPC count in all disease controls. The SSc patients in the CYC group were divided into five EPC responders and seven EPC non-responders. Circulating vascular injury markers were reduced in the responders, but not in the non-responders. During the 24-month follow-up, 3 of 10 patients developed end-stage lung disease, and all of them were EPC non-responders. CONCLUSION A low-dose i.v. CYC induces EPC mobilization, which may contribute to the efficacy for treating SSc-associated ILD.
Collapse
Affiliation(s)
- Yoshiaki Furuya
- Department of Internal Medicine, Division of Rheumatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Positioning of bone marrow hematopoietic and stromal cells relative to blood flow in vivo: serially reconstituting hematopoietic stem cells reside in distinct nonperfused niches. Blood 2010; 116:375-85. [DOI: 10.1182/blood-2009-07-233437] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Abstract
Hematopoietic stem cell (HSC) niches have been reported at the endosteum or adjacent to bone marrow (BM) vasculature. To investigate functional attributes of these niches, mice were perfused with Hoechst 33342 (Ho) in vivo before BM cell collection in presence of pump inhibitors and antibody stained. We report that the position of phenotypic HSCs, multipotent and myeloid progenitors relative to blood flow, follows a hierarchy reflecting differentiation stage, whereas mesenchymal stromal cells are perivascular. Furthermore, during granulocyte colony-stimulating factor–induced mobilization, HSCs migrated closer to blood flow, whereas stromal cells did not. Interestingly, phenotypic Lin−Sca1+KIT+CD41−CD48−CD150+ HSCs segregated into 2 groups (Honeg or Homed), based on degree of blood/Ho perfusion of their niche. HSCs capable of serial transplantation and long-term bromodeoxyuridine label retention were enriched in Honeg HSCs, whereas Homed HSCs cycled more frequently and only reconstituted a single host. This suggests that the most potent HSC niches are enriched in locally secreted factors and low oxygen tension due to negligible blood flow. Importantly, blood perfusion of niches correlates better with HSC function than absolute distance from vasculature. This technique enables prospective isolation of serially reconstituting HSCs distinct from other less potent HSCs of the same phenotype, based on the in vivo niche in which they reside.
Collapse
|
30
|
Delgaudine M, Lambermont B, Lancellotti P, Roelants V, Walrand S, Vanoverschelde JL, Pierard L, Gothot A, Beguin Y. Effects of granulocyte-colony-stimulating factor on progenitor cell mobilization and heart perfusion and function in normal mice. Cytotherapy 2010; 13:237-47. [PMID: 20524771 DOI: 10.3109/14653249.2010.491820] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS Mobilization of stem cells and progenitor cells from the bone marrow (BM) into the peripheral blood (PB) by granulocyte-colony-stimulating factor (G-CSF) is being investigated for cardiac regeneration in ischemic heart disease. However, hematopoietic (HPC), mesenchymal (MPC) and endothelial (EPC) progenitor mobilization have not been optimized and the effect of G-CSF on myocardial perfusion and cardiac function in a normal heart has never been studied. METHODS Normal mice were injected daily for 1-10 days with subcutaneous recombinant human G-CSF. PB and BM were evaluated for HPC and EPC by flow cytometry and HPC and MPC by hematopoietic (CFU-GM) and mesenchymal (CFU-F) colony assays. Echocardiography, microSPECT imaging, cardiac catheterization and immunohistochemistry were performed in mice treated for 10 days. RESULTS HPC and CFU-GM in PB peaked after 2 days, CFU-F after 4 days and EPC after 3 days. Thereafter, while HPC temporally decreased before showing a second peak, EPC remained detectable only at low levels. In BM, hematopoietic stem cells (HSC) and CFU-GM did not increase much overall but peaked twice on days 2 and 7. EPC (peak on day 7) production increased in the BM, but CFU-F formation declined considerably after day 2. G-CSF enhanced myocardial perfusion and vascularization but impaired hemodynamic performance of the heart through apparently increased ventricular wall rigidity. CONCLUSIONS G-CSF induces the mobilization of HPC, EPC and CFU-F progenitors in PB according to very different patterns, and has a significant impact on perfusion and function of the normal heart.
Collapse
Affiliation(s)
- Marie Delgaudine
- Giga-Research, Hematology Unit, University of Liège, Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Shirvaikar N, Marquez-Curtis LA, Shaw AR, Turner AR, Janowska-Wieczorek A. MT1-MMP association with membrane lipid rafts facilitates G-CSF--induced hematopoietic stem/progenitor cell mobilization. Exp Hematol 2010; 38:823-35. [PMID: 20471446 DOI: 10.1016/j.exphem.2010.05.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 05/04/2010] [Accepted: 05/06/2010] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Soluble matrix metalloproteinases (MMPs) facilitate the egress of hematopoietic stem/progenitor cells (HSPC) from the bone marrow (BM) during granulocyte colony-stimulating factor (G-CSF)-induced mobilization. Because membrane-type (MT)1-MMP, which is localized on the leading edge of migrating cells, activates the latent forms of soluble MMPs, we investigated its role in HSPC mobilization. MATERIALS AND METHODS We examined the effect of G-CSF on the expression of MT1-MMP and its activities (proMMP-2 activation and migration) in hematopoietic cells. We also investigated the subcellular localization of MT1-MMP and the signaling pathways that regulate its expression and function in hematopoietic cells after exposure to G-CSF. RESULTS We found that G-CSF increases MT1-MMP transcription and protein synthesis in hematopoietic cells; proMMP-2 activation in cocultures of HSPC with BM fibroblasts; chemoinvasion across reconstituted basement membrane Matrigel toward a stromal cell-derived factor-1 gradient, which is reduced by small interfering RNA silencing of MT1-MMP; and localization of MT1-MMP to membrane lipid rafts through a mechanism that is regulated by the phosphatidylinositol 3-kinase signaling pathway. Disruption of raft formation (by the cholesterol-sequestering agent methyl-beta-cyclodextrin) abrogated phosphatidylinositol 3-kinase phosphorylation and MT1-MMP incorporation into lipid rafts resulting in reduced proMMP-2 activation and HSPC migration. CONCLUSION G-CSF-induced upregulation of MT1-MMP in hematopoietic cells and its enhanced incorporation into membrane lipid rafts contributes to proMMP-2 activation, which facilitates mobilization of HSPC.
Collapse
Affiliation(s)
- Neeta Shirvaikar
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
32
|
Zaruba MM, Franz WM. Role of the SDF-1-CXCR4 axis in stem cell-based therapies for ischemic cardiomyopathy. Expert Opin Biol Ther 2010; 10:321-35. [PMID: 20132055 DOI: 10.1517/14712590903460286] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE OF THE FIELD Ischemic disorders are the leading cause of mortality worldwide, current therapies only delay progression of the disease. Data suggest a role of the SDF-1-CXCR4 axis in attenuation of ischemic disorders. AREAS COVERED IN THIS REVIEW We discuss the importance of SDF-1-CXCR4 interactions during development and postnatal mobilization and migration of stem cells. We focus on the role of the SDF-1-CXCR4 axis in stem-cell-based applications for attenuation of ischemic cardiomyopathy. WHAT THE READER WILL GAIN During development the SDF-1-CXCR4 axis plays a critical role in gradient-guided cell movements. In adults, the SDF-1-CXCR4 axis is involved in retention and mobilization of stem cells. Since SDF-1 is upregulated during hypoxic tissue damage, strategies to augment or stabilize SDF-1 have been utilized to target blood-derived stem cells to ischemic tissue. We exploited this concept by preventing SDF-1 degradation with dipeptidylpeptidaseIV (DPPIV) inhibition and mobilization of stem cells by G-CSF after acute myocardial infarction. This targeted CD34(+)CXCR4(+) cells to ischemic heart and attenuated ischemic cardiomyopathy. TAKE HOME MESSAGE The SDF-1-CXCR4 axis plays a role in stem cell homing during embryogenesis and adulthood especially after ischemia. Preserving functional SDF-1 by DPPIV inhibition after ischemia may enhance stem cell therapies.
Collapse
Affiliation(s)
- Marc-Michael Zaruba
- Riley Heart Research Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
33
|
Infusion of autologous bone marrow mononuclear cells through hepatic artery results in a short-term improvement of liver function in patients with chronic liver disease: a pilot randomized controlled study. Eur J Gastroenterol Hepatol 2010; 22:33-42. [PMID: 19654548 DOI: 10.1097/meg.0b013e32832eb69a] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIM This randomized controlled study evaluated the effect of autologous infusion of bone marrow cells (BMC) in patients with hepatic cirrhosis. METHODS Thirty patients on the liver transplant waiting list were randomly assigned to receive BMC therapy or no treatment. They were followed up for 1 year. The study was nonblinded. Autologous mononuclear-enriched BMC were infused into the hepatic artery; liver function scores/tests were chosen as endpoints to assess efficacy. Statistical analysis calculated mean relative changes (RC) from baseline and fitted a random-effects model. RESULTS Mean age, baseline model for end-stage liver disease, and Child-Pugh score were similar in both groups. Child-Pugh score improved in the first 90 days in the cell therapy group compared with controls (P = 0.017, BMC group RC = -8%, controls RC = +5%). The model for end-stage liver disease score remained stable in the treated patients (RC -2 to +6%), whereas it increased during follow-up in the control group (RC +6 to +18%). Albumin levels improved in the treatment arm, whereas they remained stable among controls in the first 90 days (P = 0.034; BMC group RC = +16%, control group RC = +2%). Bilirubin levels increased among controls, whereas they decreased in the therapy arm during the first 60 days; INR RC differences between groups reached up to 10%. The changes observed did not persist beyond 90 days. CONCLUSION Transplantation of autologous BMC into the hepatic artery improved liver function in patients with advanced cirrhosis in the first 90 days. However, larger studies are necessary to define the role of BMC therapy in cirrhotic patients. Repeated autologous BMC infusions or combination therapy with granulocyte-colony-stimulating factor might improve or sustain the treatment response.
Collapse
|
34
|
Mazo M, Gavira JJ, Abizanda G, Moreno C, Ecay M, Soriano M, Aranda P, Collantes M, Alegría E, Merino J, Peñuelas I, García Verdugo JM, Pelacho B, Prósper F. Transplantation of mesenchymal stem cells exerts a greater long-term effect than bone marrow mononuclear cells in a chronic myocardial infarction model in rat. Cell Transplant 2009; 19:313-28. [PMID: 19919732 DOI: 10.3727/096368909x480323] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The aim of this study is to assess the long-term effect of mesenchymal stem cells (MSC) transplantation in a rat model of chronic myocardial infarction (MI) in comparison with the effect of bone marrow mononuclear cells (BM-MNC) transplant. Five weeks after induction of MI, rats were allocated to receive intramyocardial injection of 10(6) GFP-expressing cells (BM-MNC or MSC) or medium as control. Heart function (echocardiography and (18)F-FDG-microPET) and histological studies were performed 3 months after transplantation and cell fate was analyzed along the experiment (1 and 2 weeks and 1 and 3 months). The main findings of this study were that both BM-derived populations, BM-MNC and MSC, induced a long-lasting (3 months) improvement in LVEF (BM-MNC: 26.61 +/- 2.01% to 46.61 +/- 3.7%, p < 0.05; MSC: 27.5 +/- 1.28% to 38.8 +/- 3.2%, p < 0.05) but remarkably, only MSC improved tissue metabolism quantified by (18)F-FDG uptake (71.15 +/- 1.27 to 76.31 +/- 1.11, p < 0.01), which was thereby associated with a smaller infarct size and scar collagen content and also with a higher revascularization degree. Altogether, results show that MSC provides a long-term superior benefit than whole BM-MNC transplantation in a rat model of chronic MI.
Collapse
Affiliation(s)
- Manuel Mazo
- Hematology and Cell Therapy and Division of Cancer, Clinica Universitaria and Foundation for Applied Medical Research, University of Navarra, Navarra, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Dawn B, Abdel-Latif A, Sanganalmath SK, Flaherty MP, Zuba-Surma EK. Cardiac repair with adult bone marrow-derived cells: the clinical evidence. Antioxid Redox Signal 2009; 11:1865-82. [PMID: 19203221 PMCID: PMC2848520 DOI: 10.1089/ars.2009.2462] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
On the basis of strong evidence from animal studies, numerous clinical trials of cardiac repair with adult bone marrow-derived cells (BMC) have been completed. These relatively smaller studies employed different BMC types with highly variable numbers, routes, and timings of transplantation, and included patients with acute myocardial infarction (MI), chronic ischemic heart disease (IHD), as well as ischemic cardiomyopathy. Although the outcomes have been predictably disparate, analysis of pooled data indicates that BMC therapy in patients with acute MI and chronic IHD results in modest improvements in left ventricular function and infarct scar size without any increase in untoward effects. However, the precise mechanisms underlying these benefits remain to be ascertained, and the specific advantages of one BMC type over another remain to be determined. The long-term benefit and safety issues with different BMC types are currently being evaluated critically in larger randomized controlled trials with a view to applying this novel therapeutic strategy to broader patient populations. The purpose of this review is to summarize the available clinical evidence regarding the efficacy and safety of therapeutic cardiac repair with different types of adult BMCs, and to discuss the key variables that need optimization to further enhance the benefits of BMC therapy.
Collapse
Affiliation(s)
- Buddhadeb Dawn
- Division of Cardiovascular Medicine and the Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky 40292, USA.
| | | | | | | | | |
Collapse
|
36
|
Nehlin JO, Barington T. Strategies for future histocompatible stem cell therapy. Biogerontology 2009; 10:339-76. [PMID: 19219637 DOI: 10.1007/s10522-009-9213-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 01/19/2009] [Indexed: 02/07/2023]
Abstract
Stem cell therapy based on the safe and unlimited self-renewal of human pluripotent stem cells is envisioned for future use in tissue or organ replacement after injury or disease. A gradual decline of regenerative capacity has been documented among the adult stem cell population in some body organs during the aging process. Recent progress in human somatic cell nuclear transfer and inducible pluripotent stem cell technologies has shown that patient-derived nuclei or somatic cells can be reprogrammed in vitro to become pluripotent stem cells, from which the three germ layer lineages can be generated, genetically identical to the recipient. Once differentiation protocols and culture conditions can be defined and optimized, patient-histocompatible pluripotent stem cells could be directed towards virtually every cell type in the human body. Harnessing this capability to enrich for given cells within a developmental lineage, would facilitate the transplantation of organ/tissue-specific adult stem cells or terminally differentiated somatic cells to improve the function of diseased organs or tissues in an individual. Here, we present an overview of various experimental cell therapy technologies based on the use of patient-histocompatible stem cells, the pending issues needed to be dealt with before clinical trials can be initiated, evidence for the loss and/or aging of the stem cell pool and some of the possible uses of human pluripotent stem cell-derivatives aimed at curing disease and improving health.
Collapse
Affiliation(s)
- Jan O Nehlin
- Center for Stem Cell Treatment, Department of Clinical Immunology, University of Southern Denmark, Denmark.
| | | |
Collapse
|
37
|
Suppression of CXCL12 production by bone marrow osteoblasts is a common and critical pathway for cytokine-induced mobilization. Blood 2009; 114:1331-9. [PMID: 19141863 DOI: 10.1182/blood-2008-10-184754] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Current evidence suggests that hematopoietic stem/progenitor cell (HSPC) mobilization by granulocyte colony-stimulating factor (G-CSF) is mediated by induction of bone marrow proteases, attenuation of adhesion molecule function, and disruption of CXCL12/CXCR4 signaling in the bone marrow. The relative importance and extent to which these pathways overlap or function independently are uncertain. Despite evidence of protease activation in the bone marrow, HSPC mobilization by G-CSF or the chemokine Grobeta was abrogated in CXCR4(-/-) bone marrow chimeras. In contrast, HSPC mobilization by a VLA-4 antagonist was intact. To determine whether other mobilizing cytokines disrupt CXCR4 signaling, we characterized CXCR4 and CXCL12 expression after HSPC mobilization with Flt3 ligand (Flt3L) and stem cell factor (SCF). Indeed, treatment with Flt3L or SCF resulted in a marked decrease in CXCL12 expression in the bone marrow and a loss of surface expression of CXCR4 on HSPCs. RNA in situ and sorting experiments suggested that the decreased CXCL12 expression is secondary to a loss of osteoblast lineage cells. Collectively, these data suggest that disruption of CXCR4 signaling and attenuation of VLA-4 function are independent mechanisms of mobilization by G-CSF. Loss of CXCL12 expression by osteoblast appears to be a common and key step in cytokine-induced mobilization.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) normally reside in the bone marrow but can be forced into the blood, a process termed mobilization used clinically to harvest large numbers of HSCs for transplantation. Currently the mobilizing agent of choice is granulocyte colony-stimulating factor; however, not all patients mobilize well. This article reviews recent advances in understanding the molecular mechanisms responsible for the retention of HSCs in the bone marrow, which are perturbed during HSC mobilization, and the clinical application of these findings. RECENT FINDINGS The interaction between the chemokine SDF-1/CXCL12 and its receptor CXCR4 is critical to retain HSCs within the bone marrow, leading to the discovery that small synthetic CXCR4 antagonists are potent mobilizing agents that synergize with granulocyte colony-stimulating factor. Separate research has shown that HSC numbers in the bone marrow can be boosted by increasing the number of osteoblasts that support HSCs. SUMMARY HSC mobilization induced by granulocyte colony-stimulating factor may be enhanced by directly targeting the chemotactic interaction between HSCs and bone marrow stroma with CXCR4 antagonists. When the primary problem is reduced, however, HSC numbers in the bone marrow, due to repeated chemotherapy/radiotherapy treatments, an alternative is to enhance HSC content by enhancing bone formation prior to mobilization.
Collapse
|
39
|
Sari A, Gurbuz O, Yilmaz N, Eskandari G, Tasdelen B, Basterzi Y. Granulocyte-colony stimulating factor improves the survival of ischaemic skin flaps by the induction of angiogenesis. J Plast Reconstr Aesthet Surg 2008; 62:1035-41. [PMID: 18789779 DOI: 10.1016/j.bjps.2008.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 01/27/2008] [Accepted: 02/02/2008] [Indexed: 11/16/2022]
Abstract
In this study the effects of granulocyte-colony stimulating factor (G-CSF) on angiogenesis and the survival of ischaemic skin flaps are evaluated. Thirty adult Wistar rats were equally randomised into three groups. Caudal-based, ischaemic skin flaps of 10 x 3 cm were designed on the back and injected with saline in group 1 and with 100 microg/kg G-CSF in groups 2 and 3. The injections were performed just prior to flap elevation in groups 1 and 2 and 2 days earlier in group 3. Peripheral leukocyte counts, tissue myeloperoxidase enzyme assays, necrotic to total flap area ratio (NA/TA) calculations, flap tissue inflammation gradings, immunohistochemical vessel counts, and electron microscopic evaluation of endothelial cells were performed on the 8th day. No significant difference was encountered between the groups in terms of the leukocyte counts, myeloperoxidase enzyme assays and inflammation gradings (P > 0.05), demonstrating the absence of an increased inflammatory response within the flap tissue. The surviving flap portions were observed to be increased with the application of G-CSF. The mean NA/TA results (when measured in situ) were 0.44+/-0.13 for group 1, 0.30+/-0.17 for group 2, and 0.22+/-0.16 for group 3. The difference between groups 1 and 3 was statistically significant (P = 0.009). The mean vessel count was 3.53+/-1.20 in group 1, 7.36+/-1.41 in group 2 and 7.43+/-0.92 in group 3. The differences between groups 1 and 2 and groups 1 and 3 were statistically significant (P < or = 0.001). Early apoptotic changes were encountered in the endothelial cells of group 1, while activated and proliferating endothelial cells were seen in groups 2 and 3 with electron microscopy. G-CSF promotes angiogenesis by increasing the number of activated and proliferating endothelial cells within the ischaemic flaps by the resettlement of G-CSF-stimulated endothelial progenitor cells into the ischaemic tissue. The overall result is an improved survival of ischaemic skin flaps. These effects are more pronounced when G-CSF is introduced 2 days prior to flap elevation.
Collapse
Affiliation(s)
- Alper Sari
- Department of Plastic and Reconstructive Surgery, Mersin University, Faculty of Medicine, Mersin, Turkey.
| | | | | | | | | | | |
Collapse
|
40
|
Touyz RM. Targeting the ailing endothelium – current concepts and future prospects. Can J Cardiol 2008. [DOI: 10.1016/s0828-282x(08)71037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
41
|
Sakurada K, McDonald F, Shimada F. Regenerative Medicine and Stem Cell Based Drug Discovery. Angew Chem Int Ed Engl 2008; 47:5718-38. [DOI: 10.1002/anie.200700724] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
42
|
|
43
|
Abstract
PURPOSE OF REVIEW Granulocyte colony-stimulating factor-mobilized peripheral blood stem cells are widely used to reconstitute hematopoiesis; however, preclinical and clinical studies show that improvements to this mobilization can be achieved. We discuss the development of new mobilizing regimens and evaluation of new findings on mobilized stem cell populations that may improve the utility and convenience of peripheral blood stem cell transplant. RECENT FINDINGS Chemokines and their receptors regulate leukocyte trafficking, and altering chemokine signaling pathways mobilizes stem cells. In recent trials, combination use of the chemokine (C-X-C motif) receptor 4 antagonist AMD3100 and granulocyte colony-stimulating factor mobilized more CD34 cells in fewer days than granulocyte colony-stimulating factor alone and allowed more patients to proceed to autotransplant. In preclinical studies the chemokine GRObeta synergizes with granulocyte colony-stimulating factor and when used alone or with granulocyte colony-stimulating factor mobilizes more primitive hematopoietic stem cells with less apoptosis, higher integrin activation, lower CD26 expression and enhanced marrow homing compared with granulocyte colony-stimulating factor. Hematopoietic stem cells mobilized by GRObeta or AMD3100 demonstrate superior engraftment and contribution to chimerism in primary and secondary transplant studies in mice, and peripheral blood stem cells mobilized by AMD3100 and granulocyte colony-stimulating factor in patients demonstrate enhanced engraftment capabilities in immunodeficient mice. SUMMARY Alternate regimens differentially mobilize stem cell populations with unique intrinsic properties with the potential to expand the utility of hematopoietic transplantation. Continued mechanistic evaluation will be critical to our understanding of mechanisms of mobilization and their use in regenerative medicine.
Collapse
Affiliation(s)
- Louis M Pelus
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
44
|
Strong TD, Gebska MA, Champion HC, Burnett AL, Bivalacqua TJ. Stem and endothelial progenitor cells in erection biology. Int J Impot Res 2008; 20:243-54. [DOI: 10.1038/sj.ijir.3901635] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|