1
|
Krasilnikova OA, Baranovskii DS, Yakimova AO, Arguchinskaya N, Kisel A, Sosin D, Sulina Y, Ivanov SA, Shegay PV, Kaprin AD, Klabukov ID. Intraoperative Creation of Tissue-Engineered Grafts with Minimally Manipulated Cells: New Concept of Bone Tissue Engineering In Situ. Bioengineering (Basel) 2022; 9:704. [PMID: 36421105 PMCID: PMC9687730 DOI: 10.3390/bioengineering9110704] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 07/22/2023] Open
Abstract
Transfer of regenerative approaches into clinical practice is limited by strict legal regulation of in vitro expanded cells and risks associated with substantial manipulations. Isolation of cells for the enrichment of bone grafts directly in the Operating Room appears to be a promising solution for the translation of biomedical technologies into clinical practice. These intraoperative approaches could be generally characterized as a joint concept of tissue engineering in situ. Our review covers techniques of intraoperative cell isolation and seeding for the creation of tissue-engineered grafts in situ, that is, directly in the Operating Room. Up-to-date, the clinical use of tissue-engineered grafts created in vitro remains a highly inaccessible option. Fortunately, intraoperative tissue engineering in situ is already available for patients who need advanced treatment modalities.
Collapse
Affiliation(s)
- Olga A. Krasilnikova
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Denis S. Baranovskii
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Anna O. Yakimova
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Nadezhda Arguchinskaya
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Anastas Kisel
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Dmitry Sosin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Pogodinskaya St. 10 Bld. 1, 119121 Moscow, Russia
| | - Yana Sulina
- Department of Obstetrics and Gynecology, Sechenov University, Bolshaya Pirogovskaya St. 2 Bld. 3, 119435 Moscow, Russia
| | - Sergey A. Ivanov
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Peter V. Shegay
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Ilya D. Klabukov
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering, National Research Nuclear University MEPhI, Studgorodok 1, 249039 Obninsk, Russia
| |
Collapse
|
2
|
Bouland C, Philippart P, Dequanter D, Corrillon F, Loeb I, Bron D, Lagneaux L, Meuleman N. Cross-Talk Between Mesenchymal Stromal Cells (MSCs) and Endothelial Progenitor Cells (EPCs) in Bone Regeneration. Front Cell Dev Biol 2021; 9:674084. [PMID: 34079804 PMCID: PMC8166285 DOI: 10.3389/fcell.2021.674084] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Bone regeneration is a complex, well-orchestrated process based on the interactions between osteogenesis and angiogenesis, observed in both physiological and pathological situations. However, specific conditions (e.g., bone regeneration in large quantity, immunocompromised regenerative process) require additional support. Tissue engineering offers novel strategies. Bone regeneration requires a cell source, a matrix, growth factors and mechanical stimulation. Regenerative cells, endowed with proliferation and differentiation capacities, aim to recover, maintain, and improve bone functions. Vascularization is mandatory for bone formation, skeletal development, and different osseointegration processes. The latter delivers nutrients, growth factors, oxygen, minerals, etc. The development of mesenchymal stromal cells (MSCs) and endothelial progenitor cells (EPCs) cocultures has shown synergy between the two cell populations. The phenomena of osteogenesis and angiogenesis are intimately intertwined. Thus, cells of the endothelial line indirectly foster osteogenesis, and conversely, MSCs promote angiogenesis through different interaction mechanisms. In addition, various studies have highlighted the importance of the microenvironment via the release of extracellular vesicles (EVs). These EVs stimulate bone regeneration and angiogenesis. In this review, we describe (1) the phenomenon of bone regeneration by different sources of MSCs. We assess (2) the input of EPCs in coculture in bone regeneration and describe their contribution to the osteogenic potential of MSCs. We discuss (3) the interaction mechanisms between MSCs and EPCs in the context of osteogenesis: direct or indirect contact, production of growth factors, and the importance of the microenvironment via the release of EVs.
Collapse
Affiliation(s)
- Cyril Bouland
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Philippart
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Stomatology and Maxillofacial Surgery, IRIS South Hospital, Brussels, Belgium
| | - Didier Dequanter
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Florent Corrillon
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Loeb
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Dominique Bron
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
3
|
Higashi Y, Yusoff FM, Kishimoto S, Maruhashi T. Regenerative medicine for radiation emergencies. JOURNAL OF RADIATION RESEARCH 2021; 62:i21-i29. [PMID: 33978185 PMCID: PMC8114226 DOI: 10.1093/jrr/rraa091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 05/12/2023]
Abstract
Hiroshima University is a 'medical institution for tertiary radiation emergencies' and a 'medical support organization as a part of the International Atomic Emergency Agency Emergency Preparedness Response-Response and Assistance Network (IAEA EPR-RANET)'. To establish a system of regenerative medicine for radiation emergencies with treatment by implantation of various types of cells derived from induced pluripotent stem (iPS) cells, it is necessary to establish methods of defense against and treatment for radiation-induced damage from nuclear power plant accidents and nuclear terrorism. It is also necessary to develop cell therapy, cellular repair technology and regenerative biotechnology as regenerative medicine for radiation emergencies. Such applications have not been established yet. To develop a regenerative medical system, by using the existing one, for radiation emergencies, we will attempt to manage the cell-processing center to establish a safe and secured iPS cell bank for radiation medicine. By using this iPS cell bank as the central leverage, we will develop an education program for radiation emergency medicine and construct a network of regenerative medicine for radiation emergency medicine.
Collapse
Affiliation(s)
- Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Japan
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Japan
- Corresponding author. Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan. Tel: +81-82-257-5831; Fax: +81-82-257-5831;
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Japan
| |
Collapse
|
4
|
Du F, Wang Q, Ouyang L, Wu H, Yang Z, Fu X, Liu X, Yan L, Cao Y, Xiao R. Comparison of concentrated fresh mononuclear cells and cultured mesenchymal stem cells from bone marrow for bone regeneration. Stem Cells Transl Med 2020; 10:598-609. [PMID: 33341102 PMCID: PMC7980203 DOI: 10.1002/sctm.20-0234] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/20/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Autologous bone marrow mononuclear cell (BMMNC) transplantation has been widely studied in recent years. The fresh cell cocktail in BMMNCs, without going through the in vitro culture process, helps to establish a stable microenvironment for osteogenesis, and each cell type may play a unique role in bone regeneration. Our study compared the efficacy of concentrated fresh BMMNCs and cultured bone marrow‐derived mesenchymal stem cells (BMSCs) in Beagle dogs for the first time. Fifteen‐millimeter segmental bone defects were created in the animals' tibia bones. In BMMNCs group, the defects were repaired with concentrated fresh BMMNCs combined with β‐TCP (n = 5); in cultured BMSC group, with in vitro cultured and osteo‐induced BMSCs combined with β‐TCP (n = 5); in scaffold‐only group, with a β‐TCP graft alone (n = 5); and in blank group, nothing was grafted (n = 3). The healing process was monitored by X‐rays and single photon emission computed tomography. The animals were sacrificed 12 months after surgery and their tibias were harvested and analyzed by microcomputed tomography and hard tissue histology. Moreover, the microstructure, chemical components, and microbiomechanical properties of the regenerated bone tissue were explored by multiphoton microscopy, Raman spectroscopy and nanoindentation. The results showed that BMMNCs group promoted much more bone regeneration than cultured BMSC group. The grafts in BMMNCs group were better mineralized, and they had collagen arrangement and microbiomechanical properties similar to the contralateral native tibia bone. These results indicate that concentrated fresh bone marrow mononuclear cells may be superior to in vitro expanded stem cells in segmental bone defect repair.
Collapse
Affiliation(s)
- Fengzhou Du
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China.,Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Long Ouyang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Huanhuan Wu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Yilin Cao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
5
|
Li T, Peng M, Yang Z, Zhou X, Deng Y, Jiang C, Xiao M, Wang J. 3D-printed IFN-γ-loading calcium silicate-β-tricalcium phosphate scaffold sequentially activates M1 and M2 polarization of macrophages to promote vascularization of tissue engineering bone. Acta Biomater 2018; 71:96-107. [PMID: 29549051 DOI: 10.1016/j.actbio.2018.03.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
To promote vascularization of tissue-engineered bone, IFN-γ polarizing macrophages to M1 was loaded on 5% calcium silicate/β-tricalcium phosphate (CaSiO3-β-TCP) scaffolds. IFN-γ and Si released from the scaffold were designed to polarize M1 and M2 macrophages, respectively. β-TCP, CaSiO3-β-TCP, and IFN-γ@CaSiO3-β-TCP were fabricated and biocompatibilities were evaluated. Polarizations of macrophages were detected by flow cytometry. Human umbilical vein endothelial cells with GFP were cultured and induced on Matrigel with conditioned culture medium extracted from culture of macrophages loaded on scaffolds for evaluating angiogenesis. Four weeks after the scaffolds were subcutaneously implanted into C57B1/6, vascularization was evaluated by visual observation, hematoxylin and eosin staining, as well as immunohistochemistry of CD31. The results showed that IFN-γ@CaSiO3-β-TCP scaffolds released IFN-γ in the early stage (1-3 days) to stimulate macrophages to M1 polarization, followed by release of Si inducing macrophages to M2 polarization while scaffolds degraded. The activation of M1/M2 allows macrophages to secrete more cytokines, including VEGF, CXCL12 and PDGF-BB. The IFN-γ@CaSiO3-β-TCP scaffolds formed more blood vessels in vitro and in vivo compared to the control groups. The study indicated that the design of tissue-engineered scaffolds with immunomodulatory function utilized host macrophages to increase vascularization of tissue-engineered bone, providing a new strategy for accelerating vascularization and osteogenesis of tissue-engineered scaffolds and showing the potential for treatment of major bone defects. STATEMENT OF SIGNIFICANCE A 3-D printed immunomodulatory scaffold was designed for repair of massive bone defects. Through the release of interferon γ and silicon ions, the new immunomodulatory scaffold promoted the M1 and M2 polarization of macrophages, boosting angiogenesis. This scaffold provided a new strategy for accelerating vascularization and osteogenesis of tissue-engineered scaffolds and showing the potential for treatment of major bone defects.
Collapse
|
6
|
Staruch R, Glass GE, Butler PEM. Tissue engineering through the UK Defence Medical Services: lessons learned from the Armed Forces Institute of Regenerative Medicine (AFIRM). J ROY ARMY MED CORPS 2016; 162:234-5. [PMID: 26937023 DOI: 10.1136/jramc-2015-000406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2016] [Indexed: 11/03/2022]
Affiliation(s)
- Robert Staruch
- School of Engineering & Applied Sciences, Harvard University, Boston, Massachusetts, USA
| | - G E Glass
- Department of Plastic & Reconstructive Surgery, The Kennedy Institute, Oxford University, Oxford, UK
| | - P E M Butler
- Department of Plastic & Reconstructive Surgery, University College London & The Royal Free Hospital, London, UK
| |
Collapse
|
7
|
Fukui T, Mifune Y, Matsumoto T, Shoji T, Kawakami Y, Kawamoto A, Ii M, Akimaru H, Kuroda T, Horii M, Yokoyama A, Alev C, Kuroda R, Kurosaka M, Asahara T. Superior Potential of CD34-Positive Cells Compared to Total Mononuclear Cells for Healing of Nonunion following Bone Fracture. Cell Transplant 2015; 24:1379-93. [DOI: 10.3727/096368914x681586] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We recently demonstrated that the local transplantation of human peripheral blood (PB) CD34+ cells, an endothelial/hematopoietic progenitor cell-rich population, contributes to fracture repair via vasculogenesis/angiogenesis and osteogenesis. Human PB mononuclear cells (MNCs) are also considered a potential cell fraction for neovascularization. We have previously shown the feasibility of human PB MNCs to enhance fracture healing. However, there is no report directly comparing the efficacy for fracture repair between CD34+ cells and MNCs. In addition, an unhealing fracture model, which does not accurately resemble a clinical setting, was used in our previous studies. To overcome these issues, we compared the capacity of human granulocyte colony-stimulating factor-mobilized PB (GM-PB) CD34+ cells and human GM-PB MNCs in a nonunion model, which more closely resembles a clinical setting. First, the effect of local transplantation of 1 × 105 GM-PB CD34+ cells (CD34+ group), 1 × 107 GM-PB MNCs (containing approximately 1 × 105 GM-PB CD34+ cells) (MNC group), and phosphate-buffered saline (PBS) (PBS group) on nonunion healing was compared. Similar augmentation of blood flow recovery at perinonunion sites was observed in the CD34+ and MNC groups. Meanwhile, a superior effect on nonunion repair was revealed by radiological, histological, and functional assessment in the CD34+ group compared with the other groups. Moreover, through in vivo and in vitro experiments, excessive inflammation induced by GM-PB MNCs was confirmed and believed to be one of the mechanisms underlying this potency difference. These results strongly suggest that local transplantation of GM-PB CD34+ cells is a practical and effective strategy for treatment of nonunion after fracture.
Collapse
Affiliation(s)
- Tomoaki Fukui
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yutaka Mifune
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Tomoyuki Matsumoto
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Taro Shoji
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yohei Kawakami
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Atsuhiko Kawamoto
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Masaaki Ii
- Group of Translational Stem Cell Research, Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Hiroshi Akimaru
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Tomoya Kuroda
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Miki Horii
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Ayumi Yokoyama
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Cantas Alev
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Takayuki Asahara
- Group of Vascular Regeneration Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
8
|
Corona BT, Rathbone CR. Accelerated functional recovery after skeletal muscle ischemia-reperfusion injury using freshly isolated bone marrow cells. J Surg Res 2014; 188:100-9. [PMID: 24485153 DOI: 10.1016/j.jss.2013.12.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/18/2013] [Accepted: 12/30/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Relatively little information exists regarding the usefulness of bone marrow-derived cells for skeletal muscle ischemia-reperfusion injury (I/R), especially when compared with I/R that occurs in other tissues. The objectives of this study were to evaluate the ability of freshly isolated bone marrow cells to home to injured skeletal muscle and to determine their effects on muscle regeneration. MATERIALS AND METHODS Freshly isolated lineage-depleted bone marrow cells (Lin(-) BMCs) were injected intravenously 2 d after I/R. Bioluminescent imaging was used to evaluate cell localization for up to 28 d after injury. Muscle function, the percentage of fibers with centrally located nuclei, and the capillary-to-fiber ratio were evaluated 14 d after delivery of either saline (Saline) or saline containing Lin(-) BMCs (Lin(-) BMCs). RESULTS Bioluminescence was higher in the injured leg than the contralateral control leg for up to 7 d after injection (P < 0.05) suggestive of cell homing to the injured skeletal muscle. Fourteen days after injury, there was a significant improvement in maximal tetanic torque (40% versus 22% deficit; P < 0.05), a faster rate of force production (+dP/dt) (123.6 versus 94.5 Nmm/S; P < 0.05), and a reduction in the percentage of fibers containing centrally located nuclei (40 versus 17%; P < 0.05), but no change in the capillary-to-fiber ratio in the Lin(-) BMC as compared with the Saline group. CONCLUSIONS The homing of freshly isolated BMCs to injured skeletal muscle after I/R is associated with an increase in functional outcomes.
Collapse
Affiliation(s)
- Benjamin T Corona
- Department of Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Christopher R Rathbone
- Department of Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, Texas.
| |
Collapse
|
9
|
Zhong W, Sumita Y, Ohba S, Kawasaki T, Nagai K, Ma G, Asahina I. In vivo comparison of the bone regeneration capability of human bone marrow concentrates vs. platelet-rich plasma. PLoS One 2012; 7:e40833. [PMID: 22808272 PMCID: PMC3395629 DOI: 10.1371/journal.pone.0040833] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 06/15/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone marrow aspirate concentrate (BMAC) including high densities of stem cells and progenitor cells may possess a stronger bone regenerative capability compared with Platelet-rich plasma (PRP), which contains enriched growth factors. The objective of this study was to evaluate the effects of human BMAC and PRP in combination with β-tricalcium phosphate (β-TCP) on promoting initial bone augmentation in an immunodeficient mouse model. METHODOLOGY/PRINCIPAL FINDINGS BMAC and PRP were concentrated with an automated blood separator from the bone marrow and peripheral blood aspirates. β-TCP particles were employed as a scaffold to carry cells. After cell counting and FACS characterization, three groups of nude mice (BMAC+TCP, PRP+TCP, and a TCP control) were implanted with graft materials for onlay placement on the cranium. Samples were harvested after 4 weeks, and serial sections were prepared. We observed the new bone on light microscopy and performed histomorphometric analysis. After centrifugation, the concentrations of nucleated cells and platelets in BMAC were increased by factors of 2.8 ± 0.8 and 5.3 ± 2.4, respectively, whereas leucocytes and platelets in PRP were increased by factors of 4.1 ± 1.8 and 4.4 ± 1.9, respectively. The concentrations of CD34-, CD271-, CD90-, CD105-, and CD146-positive cells were markedly increased in both BMAC and PRP. The percentage of new bone in the BMAC group (7.6 ± 3.9%) and the PRP group (7.2 ± 3.8%) were significantly higher than that of TCP group (2.7 ± 1.4%). Significantly more bone cells in the new bone occurred in sites transplanted with BMAC (552 ± 257) and PRP (491 ± 211) compared to TCP alone (187 ± 94). But the difference between the treatment groups was not significant. CONCLUSIONS/SIGNIFICANCE Both human BMACs and PRP may provide therapeutic benefits in bone tissue engineering applications. These fractions possess a similar ability to enhance early-phase bone regeneration.
Collapse
Affiliation(s)
- Weijian Zhong
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Seigo Ohba
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takako Kawasaki
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazuhiro Nagai
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan
| | - Guowu Ma
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
10
|
Exhaustive expansion: A novel technique for analyzing complex data generated by higher-order polychromatic flow cytometry experiments. J Transl Med 2010; 8:106. [PMID: 21034498 PMCID: PMC2988720 DOI: 10.1186/1479-5876-8-106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 10/30/2010] [Indexed: 01/14/2023] Open
Abstract
Background The complex data sets generated by higher-order polychromatic flow cytometry experiments are a challenge to analyze. Here we describe Exhaustive Expansion, a data analysis approach for deriving hundreds to thousands of cell phenotypes from raw data, and for interrogating these phenotypes to identify populations of biological interest given the experimental context. Methods We apply this approach to two studies, illustrating its broad applicability. The first examines the longitudinal changes in circulating human memory T cell populations within individual patients in response to a melanoma peptide (gp100209-2M) cancer vaccine, using 5 monoclonal antibodies (mAbs) to delineate subpopulations of viable, gp100-specific, CD8+ T cells. The second study measures the mobilization of stem cells in porcine bone marrow that may be associated with wound healing, and uses 5 different staining panels consisting of 8 mAbs each. Results In the first study, our analysis suggests that the cell surface markers CD45RA, CD27 and CD28, commonly used in historical lower order (2-4 color) flow cytometry analysis to distinguish memory from naïve and effector T cells, may not be obligate parameters in defining central memory T cells (TCM). In the second study, we identify novel phenotypes such as CD29+CD31+CD56+CXCR4+CD90+Sca1-CD44+, which may characterize progenitor cells that are significantly increased in wounded animals as compared to controls. Conclusions Taken together, these results demonstrate that Exhaustive Expansion supports thorough interrogation of complex higher-order flow cytometry data sets and aids in the identification of potentially clinically relevant findings.
Collapse
|
11
|
Autologous stem cell therapy for peripheral arterial disease meta-analysis and systematic review of the literature. Atherosclerosis 2009; 209:10-7. [PMID: 19740466 DOI: 10.1016/j.atherosclerosis.2009.08.033] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 08/05/2009] [Accepted: 08/17/2009] [Indexed: 11/21/2022]
Abstract
BACKGROUND Peripheral arterial disease (PAD) is a common cause of disability and mortality. Up to one third of patients are not susceptible to traditional revascularization and may benefit from stem cell therapies. OBJECTIVE In this meta-analysis, we sought to determine whether autologous cell therapy is effective in the treatment of PAD. METHODS We searched the English literature in Medline, Excerpta Medica and the Cochrane database for trials of autologous cell therapy in patients with PAD published before 31 January 2009. We included controlled and non-controlled, randomized and non-randomized trials using autologous bone marrow or granulocyte colony stimulating factor (G-CSF) mobilized peripheral blood cells to treat PAD. We also collected data from trials of G-CSF monotherapy, as a control treatment. RESULTS In a meta-analysis of 37 trials, autologous cell therapy was effective in improving surrogate indexes of ischemia, subjective symptoms and hard endpoints (ulcer healing and amputation). On the contrary, G-CSF monotherapy was not associated with significant improvement in the same endpoints. Patients with thromboangiitis obliterans showed some larger benefits than patients with atherosclerotic PAD. The intramuscular route of administration and the use of bone marrow cells seemed somehow more effective than intrarterial administration and the use of mobilized peripheral blood cells. The procedures were well tolerated and generally safe. CONCLUSION This meta-analysis indicates that intramuscular autologous bone marrow cell therapy is a feasible, relatively safe and potentially effective therapeutic strategy for PAD patients, who are not candidate for traditional revascularization. Larger, placebo-controlled, randomized multicenter trials need to be planned and conducted to confirm these findings.
Collapse
|
12
|
Yip HK, Chang LT, Wu CJ, Sheu JJ, Youssef AA, Pei SN, Lee FY, Sun CK. Autologous bone marrow-derived mononuclear cell therapy prevents the damage of viable myocardium and improves rat heart function following acute anterior myocardial infarction. Circ J 2008; 72:1336-45. [PMID: 18654023 DOI: 10.1253/circj.72.1336] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND We examined the effects of bone marrow-derived mononuclear cells (BMDMNCs) on preventing viable myocardium damage from myocardial infarction (MI) in a rat MI model. METHODS AND RESULTS Saline (group 1) or BMDMNCs (group 2) were implanted into the infarct area (IA) of 1-week-old anterior wall MI Sprague-Dawley (SD) rats. Twenty SD rats without MI served as the controls (group 3). The results demonstrated that in remote viable myocardium, the integrated area (microm2) of connexin43 spots was lower, whereas the number of apoptotic nuclei were higher in group 1 than in groups 2 and 3 on day 90 following BMDMNC implantation (all p<0.001). Additionally, the number of vessels and survival myocardium in the IA was lower in group 1 than in groups 2 and 3 (all p<0.005). Furthermore, the mRNA expressions of nitric oxide synthase, interleukin-8/Gro-alpha, interleukin-10 and matrix metalloproteinase-9 were higher in group 2 than in groups 1 and 3 in peri-IA (all p<0.05). On days 42 and 90, the left ventricular (LV) function was lower in group 1 than in groups 2 and 3 (p<0.001). CONCLUSIONS Autologous BMDMNC therapy improves LV function, and mitigates molecular and cellular perturbation following MI.
Collapse
Affiliation(s)
- Hon-Kan Yip
- Division of Cardiology, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|