1
|
Wu H, Huang J, Wu H, Xu W, Zhong Q, Song J, Linghu X, Gao B, Wa Q. Enhancement of in vitro and in vivo bone repair performance of decalcified bone/gelma by desferrioxamine. Sci Rep 2025; 15:14092. [PMID: 40269226 PMCID: PMC12019368 DOI: 10.1038/s41598-025-99101-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Autologous and allogeneic bone grafting is currently the clinical gold standard for the treatment of bone defects; however, it is limited by the scarcity of autologous sources and the risk of secondary trauma, as well as the complications of disease transmission and immune rejection associated with allogeneic grafts. The clinical management of bone defects remains a significant challenge. In this study, we prepared a demineralized bone matrix/gelatin methacrylate composite hydrogel loaded with deferoxamine (GelMA/DBM/DFO) using a freeze-drying method and investigated its properties. Assessments using CCK-8, live-dead fluorescence staining, alkaline phosphatase staining, and Alizarin Red staining indicated that the GelMA/DBM/DFO composite hydrogel demonstrated superior biocompatibility and in vitro osteogenic differentiation capacity compared with the GelMA/DBM composite hydrogel. We established a cranial defect model in Sprague-Dawley (SD) rats and examined peripheral blood indices, micro-computed tomography (Micro-CT), hematoxylin and eosin (HE) staining, Masson's trichrome staining, and immunohistochemical staining for bone morphogenetic protein-2 (BMP-2) and collagen type I (COL-1). Both hydrogels exhibited good biosafety and the GelMA/DBM/DFO hydrogel showed more effective repair of cranial defects in SD rats. This study provides a novel material for bone-defect repair.
Collapse
Affiliation(s)
- Honghan Wu
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Jun Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510632, China
| | - Hengpeng Wu
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510632, China
| | - Qian Zhong
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Jiaxiang Song
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Xitao Linghu
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510632, China.
| | - Qingde Wa
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China.
| |
Collapse
|
2
|
Suda Y, Taguchi A, Matsumoto T, Okinaka Y, Hayashi S, Tsubosaka M, Kamenaga T, Kuroda Y, Nakano N, Onoi Y, Tachibana S, Wada K, Saito A, Maeda T, Araki S, Motono K, Kuroda R. Bone marrow mononuclear cell transplantation promotes bone healing via gap junction-mediated cell-cell interaction. Stem Cells 2025; 43:sxae090. [PMID: 39847452 DOI: 10.1093/stmcls/sxae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025]
Abstract
AIMS Bone marrow mononuclear cells (BM-MNCs) are a rich source of hematopoietic stem cells that have been widely used in experimental therapies for patients with various diseases, including fractures. Activation of angiogenesis is believed to be one of the major modes of action of BM-MNCs; however, the essential mechanism by which BM-MNCs activate angiogenesis remains elusive. This study aimed to demonstrate that BM-MNCs promote bone healing by enhancing angiogenesis through direct cell-to-cell interactions via gap junctions, in addition to a previously reported method. METHODS Using a murine fracture model, we aimed to elucidate the relationship between gap junction-mediated cell-to-cell interactions and enhanced fracture healing after BM-MNC transplantation. We evaluated the transfer of substances from BM-MNCs to vascular endothelial cells and osteoblasts in the tissues surrounding the fracture site and assessed the effects of BM-MNC transplantation on bone healing, angiogenesis, and osteogenesis. RESULTS Bone marrow mononuclear cells transferred substances to vascular endothelial cells and osteoblasts in the tissues surrounding the fracture site. Moreover, BM-MNC transplantation promoted bone healing via gap junction-mediated cell-to-cell interactions, accelerating both angiogenesis and osteogenesis. CONCLUSIONS Our findings provide a novel understanding of fracture healing mechanisms and suggest that BM-MNC transplantation enhances bone healing through gap junction-mediated cell-to-cell interactions, contributing to the development of regenerative medicine strategies targeting bone repair.
Collapse
Affiliation(s)
- Yoshihito Suda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
- Department of Regenerative Medicine Research, Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Yuka Okinaka
- Department of Regenerative Medicine Research, Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Kensuke Wada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Akira Saito
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Takuma Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Shotaro Araki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
- Department of Regenerative Medicine Research, Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kohei Motono
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
3
|
Chen H, Fang HQ, Liu JT, Chang SY, Cheng LB, Sun MX, Feng JR, Liu ZM, Zhang YH, Rosen CJ, Liu P. Atlas of Fshr expression from novel reporter mice. eLife 2025; 13:RP93413. [PMID: 39773308 PMCID: PMC11709436 DOI: 10.7554/elife.93413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
The FSH-FSHR pathway has been considered an essential regulator in reproductive development and fertility. But there has been emerging evidence of FSHR expression in extragonadal organs. This poses new questions and long-term debates regarding the physiological role of the FSH-FSHR, and underscores the need for reliable, in vivo analysis of FSHR expression in animal models. However, conventional methods have proven insufficient for examining FSHR expression due to several limitations. To address this challenge, we developed Fshr-ZsGreen reporter mice under the control of Fshr endogenous promoter using CRISPR-Cas9. With this novel genetic tool, we provide a reliable readout of Fshr expression at single-cell resolution level in vivo and in real time. Reporter animals were also subjected to additional analyses,to define the accurate expression profile of FSHR in gonadal and extragonadal organs/tissues. Our compelling results not only demonstrated Fshr expression in intragonadal tissues but also, strikingly, unveiled notably increased expression in Leydig cells, osteoblast lineage cells, endothelial cells in vascular structures, and epithelial cells in bronchi of the lung and renal tubes. The genetic decoding of the widespread pattern of Fshr expression highlights its physiological relevance beyond reproduction and fertility, and opens new avenues for therapeutic options for age-related disorders of the bones, lungs, kidneys, and hearts, among other tissues. Exploiting the power of the Fshr knockin reporter animals, this report provides the first comprehensive genetic record of the spatial distribution of FSHR expression, correcting a long-term misconception about Fshr expression and offering prospects for extensive exploration of FSH-FSHR biology.
Collapse
Affiliation(s)
- Hongqian Chen
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Hui-Qing Fang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Department of Dentistry, The 980th Hospital of the PLA Joint Logistic Support ForceShijiazhuangChina
| | - Jin-Tao Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Shi-Yu Chang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Li-Ben Cheng
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Ming-Xin Sun
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Jian-Rui Feng
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Ze-Min Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Shanxi Medical Universityersity, The Second Hospital, University Shanxi Medical UniversityTaiyuanChina
| | - Yong-Hong Zhang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Shanxi Medical Universityersity, The Second Hospital, University Shanxi Medical UniversityTaiyuanChina
| | | | - Peng Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
4
|
Bury MI, Fuller NJ, Wang X, Chan YY, Sturm RM, Oh SS, Sofer LA, Arora HC, Sharma TT, Nolan BG, Feng W, Rabizadeh RR, Barac M, Edassery SS, Goedegebuure MM, Wang LW, Ganesh B, Halliday LC, Seniw ME, Edassery SL, Mahmud NB, Hofer MD, McKenna KE, Cheng EY, Ameer GA, Sharma AK. Multipotent bone marrow cell-seeded polymeric composites drive long-term, definitive urinary bladder tissue regeneration. PNAS NEXUS 2024; 3:pgae038. [PMID: 38344009 PMCID: PMC10855019 DOI: 10.1093/pnasnexus/pgae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
To date, there are no efficacious translational solutions for end-stage urinary bladder dysfunction. Current surgical strategies, including urinary diversion and bladder augmentation enterocystoplasty (BAE), utilize autologous intestinal segments (e.g. ileum) to increase bladder capacity to protect renal function. Considered the standard of care, BAE is fraught with numerous short- and long-term clinical complications. Previous clinical trials employing tissue engineering approaches for bladder tissue regeneration have also been unable to translate bench-top findings into clinical practice. Major obstacles still persist that need to be overcome in order to advance tissue-engineered products into the clinical arena. These include scaffold/bladder incongruencies, the acquisition and utility of appropriate cells for anatomic and physiologic tissue recapitulation, and the choice of an appropriate animal model for testing. In this study, we demonstrate that the elastomeric, bladder biomechanocompatible poly(1,8-octamethylene-citrate-co-octanol) (PRS; synthetic) scaffold coseeded with autologous bone marrow-derived mesenchymal stem cells and CD34+ hematopoietic stem/progenitor cells support robust long-term, functional bladder tissue regeneration within the context of a clinically relevant baboon bladder augmentation model simulating bladder trauma. Partially cystectomized baboons were independently augmented with either autologous ileum or stem-cell-seeded small-intestinal submucosa (SIS; a commercially available biological scaffold) or PRS grafts. Stem-cell synergism promoted functional trilayer bladder tissue regeneration, including whole-graft neurovascularization, in both cell-seeded grafts. However, PRS-augmented animals demonstrated fewer clinical complications and more advantageous tissue characterization metrics compared to ileum and SIS-augmented animals. Two-year study data demonstrate that PRS/stem-cell-seeded grafts drive bladder tissue regeneration and are a suitable alternative to BAE.
Collapse
Affiliation(s)
- Matthew I Bury
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Natalie J Fuller
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Xinlong Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yvonne Y Chan
- Department of Urologic Surgery, University of California at Davis, Davis, CA 95817, USA
| | - Renea M Sturm
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sang Su Oh
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Laurel A Sofer
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hans C Arora
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Tiffany T Sharma
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Bonnie G Nolan
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Wei Feng
- Flow Cytometry Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rebecca R Rabizadeh
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Milica Barac
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Sonia S Edassery
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Madeleine M Goedegebuure
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Larry W Wang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Balaji Ganesh
- Flow Cytometry Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lisa C Halliday
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark E Seniw
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
| | - Seby L Edassery
- Center for Translational Research and Education, Loyola University Chicago, Chicago, IL 60153, USA
| | - Nadim B Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois Cancer Center, Chicago, IL 60612, USA
| | | | - Kevin E McKenna
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60612, USA
| | - Earl Y Cheng
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Louis A. Simpson and Kimberly K. Querrey Biomedical Research Center, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Vascular Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60612, USA
| | - Arun K Sharma
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Louis A. Simpson and Kimberly K. Querrey Biomedical Research Center, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
5
|
Radu P, Zurzu M, Paic V, Bratucu M, Garofil D, Tigora A, Georgescu V, Prunoiu V, Pasnicu C, Popa F, Surlin P, Surlin V, Strambu V. CD34-Structure, Functions and Relationship with Cancer Stem Cells. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:938. [PMID: 37241170 PMCID: PMC10220851 DOI: 10.3390/medicina59050938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023]
Abstract
The CD34 protein was identified almost four decades ago as a biomarker for hematopoietic stem cell progenitors. CD34 expression of these stem cells has been exploited for therapeutic purposes in various hematological disorders. In the last few decades, studies have revealed the presence of CD34 expression on other types of cells with non-hematopoietic origins, such as interstitial cells, endothelial cells, fibrocytes, and muscle satellite cells. Furthermore, CD34 expression may also be found on a variety of cancer stem cells. Nowadays, the molecular functions of this protein have been involved in a variety of cellular functions, such as enhancing proliferation and blocking cell differentiation, enhanced lymphocyte adhesion, and cell morphogenesis. Although a complete understanding of this transmembrane protein, including its developmental origins, its stem cell connections, and other functions, is yet to be achieved. In this paper, we aimed to carry out a systematic analysis of the structure, functions, and relationship with cancer stem cells of CD34 based on the literature overview.
Collapse
Affiliation(s)
- Petru Radu
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Mihai Zurzu
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Vlad Paic
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Mircea Bratucu
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Dragos Garofil
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Anca Tigora
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
| | - Valentin Georgescu
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
| | - Virgiliu Prunoiu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
- Oncological Institute “Prof. Dr. Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Costin Pasnicu
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Florian Popa
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| | - Petra Surlin
- Department of Periodontology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Valeriu Surlin
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania
| | - Victor Strambu
- General Surgery Department, Carol Davila Nephrology Hospital Bucharest, 020021 Bucharest, Romania
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| |
Collapse
|
6
|
Sadek AA, Abd-Elkareem M, Abdelhamid HN, Moustafa S, Hussein K. Repair of critical-sized bone defects in rabbit femurs using graphitic carbon nitride (g-C 3N 4) and graphene oxide (GO) nanomaterials. Sci Rep 2023; 13:5404. [PMID: 37012344 PMCID: PMC10070441 DOI: 10.1038/s41598-023-32487-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Various biomaterials have been evaluated to enhance bone formation in critical-sized bone defects; however, the ideal scaffold is still missing. The objective of this study was to investigate the in vitro and in vivo regenerative capacity of graphitic carbon nitride (g-C3N4) and graphene oxide (GO) nanomaterials to stimulate critical-sized bone defect regeneration. The in vitro cytotoxicity and hemocompatibility of g-C3N4 and GO were evaluated, and their potential to induce the in vitro osteogenesis of human fetal osteoblast (hFOB) cells was assessed using qPCR. Then, bone defect in femoral condyles was created in rabbits and left empty as control or filled with either g-C3N4 or GO. The osteogenesis of the different implanted scaffolds was evaluated after 4, 8, and 12 weeks of surgery using X-ray, computed tomography (CT), macro/microscopic examinations, and qPCR analysis of osteocalcin (OC) and osteopontin (OP) expressions. Both materials displayed good cell viability and hemocompatibility with enhanced collagen type-I (Col-I), OC, and OP expressions of the hFOB cells. Compared to the control group, the bone healing process in g-C3N4 and GO groups was promoted in vivo. Moreover, complete healing of the bone defect was observed radiologically and grossly in g-C3N4 implanted group. Additionally, g-C3N4 implanted group showed higher percentages of osteoid tissue, mature collagen, biodegradation, and expressions of OC and OP. In conclusion, our results revealed that g-C3N4 and GO nanomaterials could induce osteogenesis in critical-sized bone defects.
Collapse
Affiliation(s)
- Ahmed Abdelrahiem Sadek
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mahmoud Abd-Elkareem
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Hani Nasser Abdelhamid
- Advanced Multifunctional Materials Laboratory, Department of Chemistry, Faculty of Science, Assiut University, Assiut, Egypt
- Proteomics Laboratory for Clinical Research and Materials Science, Department of Chemistry, Faculty of Science, Assiut University, Assiut, Egypt
- Nanotechnology Research Centre (NTRC), The British University in Egypt (BUE), Suez Desert Road, El-Sherouk City, 11837, Cairo, Egypt
| | - Samia Moustafa
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Kamal Hussein
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| |
Collapse
|
7
|
Haeusner S, Jauković A, Kupczyk E, Herrmann M. Review: cellularity in bone marrow autografts for bone and fracture healing. Am J Physiol Cell Physiol 2023; 324:C517-C531. [PMID: 36622067 DOI: 10.1152/ajpcell.00482.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of autografts, as primary cell and tissue source, is the current gold standard approach to treat critical size bone defects and nonunion defects. The unique mixture of the autografts, containing bony compartments and bone marrow (BM), delivers promising results. Although BM mesenchymal stromal cells (BM-MSCs) still represent a major target for various healing approaches in current preclinical research and respective clinical trials, their occurrence in the human BM is typically low. In vitro expansion of this cell type is regulatory challenging as well as time and cost intensive. Compared with marginal percentages of resident BM-MSCs in BM, BM mononuclear cells (BM-MNCs) contained in BM aspirates, concentrates, and bone autografts represent a readily available abundant cell source, applicable within hours during surgical procedures without the need for time-consuming and regulatory challenging cell expansion. This benefit is one reason why autografting has become a clinical standard procedure. However, the exact anatomy and cellularity of BM-MNCs in humans, which is strongly correlated to their unique mode of action and wide application range remains to be elucidated. The aim of this review was to present an overview of the current knowledge on these specific cell types found in human BM, emphasize the contribution of BM-MNCs in bone healing, highlight donor site dependence, and discuss limitations in the current isolation and subsequent characterization procedures. Hereby, the most recent and relevant examples of human BM-MNC cell characterization, flow cytometric analyses, and findings are summarized, with a strong focus on bone therapy.
Collapse
Affiliation(s)
- S Haeusner
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital of Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - A Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - E Kupczyk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - M Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital of Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
8
|
Yuan G, Li Z, Lin X, Li N, Xu R. New perspective of skeletal stem cells. BIOMATERIALS TRANSLATIONAL 2022; 3:280-294. [PMID: 36846511 PMCID: PMC9947737 DOI: 10.12336/biomatertransl.2022.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/29/2022] [Accepted: 12/19/2022] [Indexed: 03/01/2023]
Abstract
Tissue-resident stem cells are a group of stem cells distinguished by their capacity for self-renewal and multilineage differentiation capability with tissue specificity. Among these tissue-resident stem cells, skeletal stem cells (SSCs) were discovered in the growth plate region through a combination of cell surface markers and lineage tracing series. With the process of unravelling the anatomical variation of SSCs, researchers were also keen to investigate the developmental diversity outside the long bones, including in the sutures, craniofacial sites, and spinal regions. Recently, fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing have been used to map lineage trajectories by studying SSCs with different spatiotemporal distributions. The SSC niche also plays a pivotal role in regulating SSC fate, such as cell-cell interactions mediated by multiple signalling pathways. This review focuses on discussing the spatial and temporal distribution of SSCs, and broadening our understanding of the diversity and plasticity of SSCs by summarizing the progress of research into SSCs in recent years.
Collapse
Affiliation(s)
- Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China
| | - Zan Li
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xixi Lin
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China
| | - Na Li
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Corresponding authors: Ren Xu, ; Na Li,
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Corresponding authors: Ren Xu, ; Na Li,
| |
Collapse
|
9
|
Changes in interstitial fluid flow, mass transport and the bone cell response in microgravity and normogravity. Bone Res 2022; 10:65. [PMID: 36411278 PMCID: PMC9678891 DOI: 10.1038/s41413-022-00234-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
In recent years, our scientific interest in spaceflight has grown exponentially and resulted in a thriving area of research, with hundreds of astronauts spending months of their time in space. A recent shift toward pursuing territories farther afield, aiming at near-Earth asteroids, the Moon, and Mars combined with the anticipated availability of commercial flights to space in the near future, warrants continued understanding of the human physiological processes and response mechanisms when in this extreme environment. Acute skeletal loss, more severe than any bone loss seen on Earth, has significant implications for deep space exploration, and it remains elusive as to why there is such a magnitude of difference between bone loss on Earth and loss in microgravity. The removal of gravity eliminates a critical primary mechano-stimulus, and when combined with exposure to both galactic and solar cosmic radiation, healthy human tissue function can be negatively affected. An additional effect found in microgravity, and one with limited insight, involves changes in dynamic fluid flow. Fluids provide the most fundamental way to transport chemical and biochemical elements within our bodies and apply an essential mechano-stimulus to cells. Furthermore, the cell cytoplasm is not a simple liquid, and fluid transport phenomena together with viscoelastic deformation of the cytoskeleton play key roles in cell function. In microgravity, flow behavior changes drastically, and the impact on cells within the porous system of bone and the influence of an expanding level of adiposity are not well understood. This review explores the role of interstitial fluid motion and solute transport in porous bone under two different conditions: normogravity and microgravity.
Collapse
|
10
|
Sadek AA, Abd-Elkareem M, Abdelhamid HN, Moustafa S, Hussein K. Enhancement of critical-sized bone defect regeneration using UiO-66 nanomaterial in rabbit femurs. BMC Vet Res 2022; 18:260. [PMID: 35791016 PMCID: PMC9254639 DOI: 10.1186/s12917-022-03347-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Repair of large-sized bone defects is a challengeable obstacle in orthopedics and evoked the demand for the development of biomaterials that could induce bone repair in such defects. Recently, UiO-66 has emerged as an attractive metal–organic framework (MOF) nanostructure that is incorporated in biomedical applications due to its biocompatibility, porosity, and stability. In addition, its osteogenic properties have earned a great interest as a promising field of research. Thus, the UiO-66 was prepared in this study and assessed for its potential to stimulate and support osteogenesis in vitro and in vivo in a rabbit femoral condyle defect model. The nanomaterial was fabricated and characterized using x-ray diffraction (XRD) and transmission electron microscopy (TEM). Afterward, in vitro cytotoxicity and hemolysis assays were performed to investigate UiO-66 biocompatibility. Furthermore, the material in vitro capability to upregulate osteoblast marker genes was assessed using qPCR. Next, the in vivo new bone formation potential of the UiO-66 nanomaterial was evaluated after induction of bone defects in rabbit femoral condyles. These defects were left empty or filled with UiO-66 nanomaterial and monitored at weeks 4, 8, and 12 after bone defect induction using x-ray, computed tomography (CT), histological examinations, and qPCR analysis of osteocalcin (OC) and osteopontin (OP) expressions.
Results
The designed UiO-66 nanomaterial showed excellent cytocompatibility and hemocompatibility and stimulated the in vitro osteoblast functions. The in vivo osteogenesis was enhanced in the UiO-66 treated group compared to the control group, whereas evidence of healing of the treated bone defects was observed grossly and histologically. Interestingly, UiO-66 implanted defects displayed a significant osteoid tissue and collagen deposition compared to control defects. Moreover, the UiO-66 nanomaterial demonstrated the potential to upregulate OC and OP in vivo.
Conclusions
The UiO-66 nanomaterial implantation possesses a stimulatory impact on the healing process of critical-sized bone defects indicating that UiO-66 is a promising biomaterial for application in bone tissue engineering.
Collapse
|
11
|
Oliveira CS, Carreira M, Correia CR, Mano JF. The Therapeutic Potential of Hematopoietic Stem Cells in Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:379-392. [PMID: 33683146 DOI: 10.1089/ten.teb.2021.0019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The repair process of bone fractures is a complex biological mechanism requiring the recruitment and in situ functionality of stem/stromal cells from the bone marrow (BM). BM mesenchymal stem/stromal cells have been widely explored in multiple bone tissue engineering applications, whereas the use of hematopoietic stem cells (HSCs) has been poorly investigated in this context. A reasonable explanation is the fact that the role of HSCs and their combined effect with other elements of the hematopoietic niches in the bone-healing process is still elusive. Therefore, in this review we intend to highlight the influence of HSCs in the bone repair process, mainly through the promotion of osteogenesis and angiogenesis at the bone injury site. For that, we briefly describe the main biological characteristics of HSCs, as well as their hematopoietic niches, while reviewing the biomimetic engineered BM niche models. Moreover, we also highlighted the role of HSCs in translational in vivo transplantation or implantation as promoters of bone tissue repair.
Collapse
Affiliation(s)
- Cláudia S Oliveira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Mariana Carreira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Clara R Correia
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
12
|
Abbasi N, Lee RSB, Ivanovski S, Love RM, Hamlet S. In vivo bone regeneration assessment of offset and gradient melt electrowritten (MEW) PCL scaffolds. Biomater Res 2020; 24:17. [PMID: 33014414 PMCID: PMC7529514 DOI: 10.1186/s40824-020-00196-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/21/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Biomaterial-based bone tissue engineering represents a promising solution to overcome reduced residual bone volume. It has been previously demonstrated that gradient and offset architectures of three-dimensional melt electrowritten poly-caprolactone (PCL) scaffolds could successfully direct osteoblast cells differentiation toward an osteogenic lineage, resulting in mineralization. The aim of this study was therefore to evaluate the in vivo osteoconductive capacity of PCL scaffolds with these different architectures. METHODS Five different calcium phosphate (CaP) coated melt electrowritten PCL pore sized scaffolds: 250 μm and 500 μm, 500 μm with 50% fibre offset (offset.50.50), tri layer gradient 250-500-750 μm (grad.250top) and 750-500-250 μm (grad.750top) were implanted into rodent critical-sized calvarial defects. Empty defects were used as a control. After 4 and 8 weeks of healing, the new bone was assessed by micro-computed tomography and immunohistochemistry. RESULTS Significantly more newly formed bone was shown in the grad.250top scaffold 8 weeks post-implantation. Histological investigation also showed that soft tissue was replaced with newly formed bone and fully covered the grad.250top scaffold. While, the bone healing did not happen completely in the 250 μm, offset.50.50 scaffolds and blank calvaria defects following 8 weeks of implantation. Immunohistochemical analysis showed the expression of osteogenic markers was present in all scaffold groups at both time points. The mineralization marker Osteocalcin was detected with the highest intensity in the grad.250top and 500 μm scaffolds. Moreover, the expression of the endothelial markers showed that robust angiogenesis was involved in the repair process. CONCLUSIONS These results suggest that the gradient pore size structure provides superior conditions for bone regeneration.
Collapse
Affiliation(s)
- Naghmeh Abbasi
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| | - Ryan S. B. Lee
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- School of Dentistry, University of Queensland, Herston Campus, Herston, Queensland 4006 Australia
| | - Saso Ivanovski
- School of Dentistry, University of Queensland, Herston Campus, Herston, Queensland 4006 Australia
| | - Robert M. Love
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| | - Stephen Hamlet
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| |
Collapse
|
13
|
Hellwinkel JE, Miclau T, Provencher MT, Bahney CS, Working ZM. The Life of a Fracture: Biologic Progression, Healing Gone Awry, and Evaluation of Union. JBJS Rev 2020; 8:e1900221. [PMID: 32796195 PMCID: PMC11147169 DOI: 10.2106/jbjs.rvw.19.00221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
New knowledge about the molecular biology of fracture-healing provides opportunities for intervention and reduction of risk for specific phases that are affected by disease and medications. Modifiable and nonmodifiable risk factors can prolong healing, and the informed clinician should optimize each patient to provide the best chance for union. Techniques to monitor progression of fracture-healing have not changed substantially over time; new objective modalities are needed.
Collapse
Affiliation(s)
- Justin E Hellwinkel
- Department of Orthopedic Surgery, New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Theodore Miclau
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Matthew T Provencher
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Chelsea S Bahney
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Zachary M Working
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
- Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
14
|
Valid Presumption of Shiga Toxin-Mediated Damage of Developing Erythrocytes in EHEC-Associated Hemolytic Uremic Syndrome. Toxins (Basel) 2020; 12:toxins12060373. [PMID: 32512916 PMCID: PMC7354503 DOI: 10.3390/toxins12060373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
The global emergence of clinical diseases caused by enterohemorrhagic Escherichia coli (EHEC) is an issue of great concern. EHEC release Shiga toxins (Stxs) as their key virulence factors, and investigations on the cell-damaging mechanisms toward target cells are inevitable for the development of novel mitigation strategies. Stx-mediated hemolytic uremic syndrome (HUS), characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal injury, is the most severe outcome of an EHEC infection. Hemolytic anemia during HUS is defined as the loss of erythrocytes by mechanical disruption when passing through narrowed microvessels. The formation of thrombi in the microvasculature is considered an indirect effect of Stx-mediated injury mainly of the renal microvascular endothelial cells, resulting in obstructions of vessels. In this review, we summarize and discuss recent data providing evidence that HUS-associated hemolytic anemia may arise not only from intravascular rupture of erythrocytes, but also from the extravascular impairment of erythropoiesis, the development of red blood cells in the bone marrow, via direct Stx-mediated damage of maturing erythrocytes, leading to “non-hemolytic” anemia.
Collapse
|
15
|
Xu GP, Zhang XF, Sun L, Chen EM. Current and future uses of skeletal stem cells for bone regeneration. World J Stem Cells 2020; 12:339-350. [PMID: 32547682 PMCID: PMC7280866 DOI: 10.4252/wjsc.v12.i5.339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/07/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
The postnatal skeleton undergoes growth, modeling, and remodeling. The human skeleton is a composite of diverse tissue types, including bone, cartilage, fat, fibroblasts, nerves, blood vessels, and hematopoietic cells. Fracture nonunion and bone defects are among the most challenging clinical problems in orthopedic trauma. The incidence of nonunion or bone defects following fractures is increasing. Stem and progenitor cells mediate homeostasis and regeneration in postnatal tissue, including bone tissue. As multipotent stem cells, skeletal stem cells (SSCs) have a strong effect on the growth, differentiation, and repair of bone regeneration. In recent years, a number of important studies have characterized the hierarchy, differential potential, and bone formation of SSCs. Here, we describe studies on and applications of SSCs and/or mesenchymal stem cells for bone regeneration.
Collapse
Affiliation(s)
- Guo-Ping Xu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiang-Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Lu Sun
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, United States
| | - Er-Man Chen
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
16
|
Doppelt O, Cohen G, Tamari T, Elimelech R, Sabbah N, Zigdon-Giladi H. Endothelial progenitors increase vascularization and improve fibroblasts function that prevent medication-related osteonecrosis of the jaw. Oral Dis 2020; 26:1523-1531. [PMID: 32400918 DOI: 10.1111/odi.13412] [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: 02/03/2020] [Revised: 04/22/2020] [Accepted: 05/03/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES In a previous rat model, MRONJ occurrence was 50%. Our aim was to investigate the potential of endothelial progenitor cells (EPCs) to improve fibroblasts function and prevent MRONJ. METHODS Human gingival fibroblasts were cultured with EPC-conditioned media (EPC-CM); endothelial growth media (EGM-2) or DMEM followed by incubation with 10 µM zoledronic (ZOL) and dexamethasone (DEX). Cell proliferation and migration were assessed by XTT and scratch wound healing assays. In vivo, ten Lewis rats were treated weekly with ZOL and DEX for 11 weeks. After a week, EPCs or EGM-2 were injected to the gingiva around the molars. At 3 weeks, bilateral molars were extracted. After 8 weeks, wound healing was assessed, and serum VEGF and blood vessels were quantified. RESULTS ZOL/DEX significantly reduced fibroblasts proliferation and wound healing. Treatment with EPC-CM before ZOL/DEX improved cell proliferation, and scratch healing (p = .007, p = .023). In vivo, local EPC injection before tooth extraction increased serum VEGF (p = .01) and soft tissue vascularization (p = .05). Normal healing was similar (80%) in EPCs and EGM-2 groups. CONCLUSION EPC rescued fibroblasts from the cytotoxic effect of ZOL/DEX and elevated serum VEGF and vessel density that might reduce MRONJ occurrence to 20% compared to 50% in a similar model.
Collapse
Affiliation(s)
- Ofri Doppelt
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Gal Cohen
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Rina Elimelech
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel.,Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel
| | - Nadin Sabbah
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Hadar Zigdon-Giladi
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.,Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
17
|
Tamari T, Elimelech R, Cohen G, Cohen T, Doppelt O, Eskander-Hashoul L, Zigdon-Giladi H. Endothelial Progenitor Cells inhibit jaw osteonecrosis in a rat model: A major adverse effect of bisphosphonate therapy. Sci Rep 2019; 9:18896. [PMID: 31827217 PMCID: PMC6906486 DOI: 10.1038/s41598-019-55383-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 11/22/2019] [Indexed: 01/27/2023] Open
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a serious adverse effect of antiresorptive and antiangiogenic therapies. MRONJ is identified by chronic wounds in the oral mucosa associated with exposed necrotic bone. We hypothesized that zoledronic acid (ZOL) impairs keratinocyte and fibroblast function and reduces soft tissue vascularization; therefore, treating MRONJ with proangiogenic cells may benefit MRONJ patients. The effect of ZOL and dexamethasone (DEX) on gingival fibroblasts and keratinocytes was investigated. In-vitro, ZOL inhibited fibroblast and keratinocyte proliferation, delaying scratch healing. In-vivo, exposed bone was detected at tooth extraction sites, mainly in ZOL(+)/DEX(+) rats; and was associated with significantly decreased soft tissue vascularization, serum-VEGF, and tissue-VEGF. Local injection of early and late endothelial progenitor cells (EPCs) healed 13 of 14 MRONJ lesions compared with 2/7 lesions in the mesenchymal stem cells, and 2/6, in culture-medium group. The EPCs reduced necrotic bone area, increased serum and tissue VEGF levels. EPCs engraftment was minimal, suggesting their paracrine role in MRONJ healing. The EPC-conditioned medium improved scratch healing of keratinocytes and fibroblasts via VEGF pathway and elevated mRNA of VEGFA and collagen1A1. In conclusion, a novel MRONJ treatment with EPCs, increased vascularization and improved epithelial and fibroblast functions as well as cured the lesion.
Collapse
Affiliation(s)
- Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Rina Elimelech
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel.,Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Gal Cohen
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Talia Cohen
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ofri Doppelt
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Lana Eskander-Hashoul
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel.,Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Hadar Zigdon-Giladi
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel. .,Laboratory for Bone Repair, Rambam Health Care Campus, Haifa, Israel. .,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The emergence of cell-based therapies has brought much excitement to the field of orthopedic sports medicine. However, the significant inconsistency of reporting has led to the poor understanding, misinformation, and false expectations for patients and clinicians alike. In this paper, we aim to clarify the available cell-therapy treatments and summarize some of the latest research. RECENT FINDINGS Although this technology is in early development, our understanding of cell biology has grown significantly over the last decade. Furthermore, it is becoming evident that tissue specificity may play a significant role in determining the effectiveness and overall clinical benefit attributed to cell therapy. Cell therapy is an emerging field with tremendous potential for clinically significant benefit. However, in its current state, clinical application of these treatments is limited by federal regulations, variability in formulation, and limited understanding of the biologic activity of various cell formulations.
Collapse
Affiliation(s)
- Bijan Dehghani
- Hospital for Special Surgery, 525 East 71 Street, 1st floor, New York, NY 10021 USA
| | - Scott Rodeo
- Hospital for Special Surgery, 525 East 71 Street, 1st floor, New York, NY 10021 USA
| |
Collapse
|
19
|
Meeson R, Sanghani-Keri A, Coathup M, Blunn G. CXCR4 Antagonism to Treat Delayed Fracture Healing. Tissue Eng Part A 2019; 25:1242-1250. [PMID: 30612520 PMCID: PMC6864747 DOI: 10.1089/ten.tea.2018.0265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A significant number of fractures develop nonunion. Stem cell homing is regulated through stromal cell-derived factor 1 (SDF1) and its receptor CXCR4. Stem/progenitor cell populations can be endogenously mobilized by administering growth factors with a pharmacological antagonist of CXCR4, AMD3100, which may be a means to improve fracture healing. A 1.5 mm femoral osteotomy in Wistar rats was stabilized with an external fixator. Rats were pretreated with phosphate buffered saline [PBS(P)], vascular endothelial growth factor [VEGF(V)], insulin-like growth factor-1 [IGF1(I)], or granulocyte colony stimulating factor [GCSF(G)] before AMD3100. A control group (C) did not receive growth factors or AMD3100. Bone formation after 5 weeks was analyzed. Group P had a significant increase in total bone volume (BV) (p = 0.01) and group I in percentage bone in the fracture gap (p = 0.035). Group G showed a decrease in BV. All treated groups had an increase in trabecular thickness. Histology showed decreased cartilage tissue associated with increased bone in groups with improved healing, and increased fibrous tissue in poorly performing groups. Antagonism of SDF1-CXCR4 axis can boost impaired fracture healing. AMD3100 given alone was the most effective means to boost healing, whereas pretreatment with GCSF reduced healing. AMD3100 is likely mobilizing stem cells into the blood stream that home to the fracture site enhancing healing.
Collapse
Affiliation(s)
- Richard Meeson
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,Department of Clinical Services and Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Anita Sanghani-Keri
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom
| | - Melanie Coathup
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,University of Central Florida, Orlando, Florida
| | - Gordon Blunn
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
20
|
Bian Y, Du Y, Wang R, Chen N, Du X, Wang Y, Yuan H. A comparative study of HAMSCs/HBMSCs transwell and mixed coculture systems. IUBMB Life 2019; 71:1048-1055. [PMID: 31112365 DOI: 10.1002/iub.2074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/16/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022]
Abstract
Our previous studies indicated that a coculture system containing human amnion-derived mesenchymal stem cells (HAMSCs) and human bone marrow mesenchymal stem cells (HBMSCs) has the potential of application for bone regeneration. However, there is currently no enough comparative investigation between HAMSCs/HBMSCs transwell and mixed coculture systems. This study aimed to assess the phenotype and mechanisms regulated by indirect and direct coculture systems, respectively. Two in vitro models were employed with HAMSCs and HBMSCs at a ratio of 3:1, and then were analyzed by a series of processes, including flow cytometry, alkaline phosphatase (ALP) substrate assays, Alizarin red S staining, quantitative reverse transcription polymerase chain reaction (RT-qPCR), and Western blot analysis. We found that cell proliferation, ALP activity, mineralized matrix formation, and osteoblast-related mRNA expression were accelerated in transwell coculture system compared with mixed coculture system. Conditioned medium from transwell coculture system achieved an elevated level of vascular endothelial growth factor and induced more vascular structures in human umbilical vein endothelial cells than those of mixed coculture system. Moreover, we observed that transwell coculture system, promoted osteogenesis and angiogenesis by maintaining stemness through extracellular regulated protein kinases 1/2 (ERK1/2) mitogen-activated protein kinase (MAPK) signaling pathway. U0126, a selective inhibitor of ERK1/2 MAPK signaling, significantly suppressed maintaining of the stemness-based effects on transwell coculture system. Taken together, our results compared the merits of two different models and clarified the role of HAMSCs/HBMSCs transwell coculture system in the development of bone tissue engineering. © 2019 IUBMB Life, 2019.
Collapse
Affiliation(s)
- Yifeng Bian
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yifei Du
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Ruixia Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Dental Implant, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Ning Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xin Du
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Yuli Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hua Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Pasqualini L, Ministrini S, Lombardini R, Bagaglia F, Paltriccia R, Pippi R, Collebrusco L, Reginato E, Sbroma Tomaro E, Marini E, D'Abbondanza M, Scarponi AM, De Feo P, Pirro M. Effects of a 3-month weight-bearing and resistance exercise training on circulating osteogenic cells and bone formation markers in postmenopausal women with low bone mass. Osteoporos Int 2019; 30:797-806. [PMID: 30809725 DOI: 10.1007/s00198-019-04908-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Abstract
UNLABELLED Osteoporosis is a health issue in postmenopausal women. Physical activity is recommended in these subjects, since it has positive effects on bone mass. Cellular mechanisms underlying this effect are still unclear. Osteogenic cells, released after physical exertion, could be a key factor in exercise-induced bone formation. INTRODUCTION The aim of our research was to explore if a weight-bearing and resistance exercise program could positively affect circulating osteogenic cells (OCs), markers of bone formation and quality of life (QoL) in osteopenic postmenopausal women. METHODS We recruited 33 postmenopausal women with a T-score at lumbar spine or femoral neck between - 1 and - 2.5 SD. Anthropometric and fitness parameters, bone-remodeling markers, OCs, and QoL were evaluated at the time of enrolment, after 1-month run-in period, and after 3 months of weight-bearing and resistance exercise. RESULTS After 3 months of training, the pro-collagen type 1 N-terminal peptide (P1NP) and the number of OCs were significantly increased, with no significant increase of the type 1 collagen cross-linked C-telopeptide (sCTX). We also observed a significant increase in body height, one-repetition maximum (1RM) on the pull-down lat machine and leg press, and mean VO2max. The increase of immature circulating OCs was significantly correlated with the improvement of 1RM both of the upper and lower limbs. Moreover, QoL was significantly improved with regard to pain, physical function, mental function, and general QoL. The improvement in QoL, namely in the overall score and in the pain score, was significantly correlated with the increase in height. CONCLUSIONS The exercise program we trialed is able to increase the markers of bone formation and the commitment of immature OCs with no significant increase in the markers of bone resorption. Our results confirm that combined weight-bearing and resistance physical activity is an effective tool to improve QoL of postmenopausal women with low bone mass. TRIAL REGISTRATION NCT03195517.
Collapse
Affiliation(s)
- L Pasqualini
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - S Ministrini
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy.
| | - R Lombardini
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - F Bagaglia
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - R Paltriccia
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - R Pippi
- Healthy Lifestyle Institute CURIAMO, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - L Collebrusco
- Rehabilitation Unit, National Health Service of Umbria, Perugia, Italy
| | - E Reginato
- Healthy Lifestyle Institute CURIAMO, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - E Sbroma Tomaro
- Healthy Lifestyle Institute CURIAMO, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - E Marini
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - M D'Abbondanza
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - A M Scarponi
- Medicina Interna, Azienda Ospedaliera "S.M. della Misericordia", Perugia, Italy
| | - P De Feo
- Healthy Lifestyle Institute CURIAMO, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - M Pirro
- Sezione di Medicina Interna e Angiologia, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
22
|
Asiedu KO, Ferdousi M, Ton PT, Adler SS, Choyke PL, Sato N. Bone marrow cell homing to sites of acute tibial fracture: 89Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model. EJNMMI Res 2018; 8:109. [PMID: 30547233 PMCID: PMC6292830 DOI: 10.1186/s13550-018-0463-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Background Bone fracture healing is dependent upon the rapid migration and engraftment of bone marrow (BM) progenitor and stem cells to the site of injury. Stromal cell-derived factor-1 plays a crucial role in recruiting BM cells expressing its receptor CXCR4. Recently, a CXCR4 antagonist, plerixafor, has been used to mobilize BM cells into the blood in efforts to enhance cell migration to sites of injury presumably improving healing. In this study, we employed zirconium-89 (89Zr)-oxine-labeled BM cells imaged with positron emission tomography (PET)/computed tomography (CT) to visualize and quantitate BM cell trafficking following acute bone injury and to investigate the effect of plerixafor on BM cell homing. Unilateral 1-mm incisions were created in the distal tibia of mice either on the same day (d0) or 24 h (d1) after 89Zr-oxine-labeled BM cell transfer (n = 4–6, 2–2.3 × 107 cells at 9.65–15.7 kBq/106 cells). Serial microPET/CT imaging was performed and migration of 89Zr-labeled cells to the bone injury was quantified. The effects of three daily doses of plerixafor on cell trafficking were evaluated beginning on the day of fracture generation (n = 4–6). The labeled cells localizing to the fracture were analyzed by flow cytometry and immunohistochemistry. Results In d0- and d1-fracture groups, 0.7% and 1.7% of administered BM cells accumulated within the fracture, respectively. Plerixafor treatment reduced BM cell migration to the fracture by approximately one-third (p < 0.05 for both fracture groups). Flow cytometry analysis of donor cells collected from the injured site revealed a predominance of CD45+ stem/progenitor cell populations and subsequent histological analysis demonstrated the presence of donor cells engrafted within sites of fracture repair. Conclusion 89Zr-oxine labeling enabled visualization and quantitation of BM cell recruitment to acute fractures and further demonstrated that plerixafor plays an inhibitory role in this recruitment. Electronic supplementary material The online version of this article (10.1186/s13550-018-0463-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kingsley O Asiedu
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Munira Ferdousi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Phuongnga T Ton
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Stephen S Adler
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, 21702, USA
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA
| | - Noriko Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Building 10, Room B3B406, Bethesda, MD, 20892-1002, USA.
| |
Collapse
|
23
|
Cheng Q, Lin S, Bi B, Jiang X, Shi H, Fan Y, Lin W, Zhu Y, Yang F. Bone Marrow-derived Endothelial Progenitor Cells Are Associated with Bone Mass and Strength. J Rheumatol 2018; 45:1696-1704. [PMID: 30173148 DOI: 10.3899/jrheum.171226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Blood vessels of bone are thought to influence osteogenesis of bone. No clinical studies have determined whether angiogenesis is related to bone mass and gene expression of growth factors. We compared bone marrow endothelial progenitor cells (EPC), which control angiogenesis of bone in postmenopausal women incurring fragility fracture, with osteoporosis or traumatic fracture with normal bone mass (COM). METHODS Bone specimens were obtained from age-matched women with osteoporosis or COM. Mononuclear cells were isolated and EPC were detected by flow cytometry. The expression levels of specific genes were measured. Bone mineral density (BMD) was determined, and serum markers of bone turnover also were measured. Differences between OP and COM were assessed with Student t test or Mann-Whitney U test, and correlations were determined using Spearman's correlation. RESULTS Compared with COM, patients with OP had significantly lower levels of serum osteocalcin, procollagen type-1 N-terminal propeptide, and 25-hydroxy vitamin D, as well as decreased BMD of total hip and femoral neck and fewer bone marrow EPC. Expression levels of vascular endothelial growth factor, angiopoietin-1 (Ang-1), angiopoietin 2 (Ang-2), and the osteoblast-specific genes runt-related transcription factor 2 (RUNX2) and osterix in bone were significantly lower in OP than in COM. We determined that mature EPC were correlated positively with BMD of the femoral neck and total hip, gene expression of Ang-1, RUNX2, and CD31, and negatively with gene expression of receptor activator of nuclear factor-κB ligand and Ang-2. CONCLUSION Our results demonstrate correlations of bone marrow EPC with bone mass and gene expression of growth factors, which support a hypothesis of crosstalk between angiogenesis and osteogenesis in bone health.
Collapse
Affiliation(s)
- Qun Cheng
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China. .,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article.
| | - Shangjin Lin
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Bo Bi
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Xin Jiang
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Hongli Shi
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Yongqian Fan
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Weilong Lin
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Yuefeng Zhu
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| | - Fengjian Yang
- From the Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; the Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; and the Central Laboratory, Huadong Hospital, affiliated to Fudan University, Shanghai, China.,Q. Cheng, MD, PhD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; S. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; B. Bi, MD, PhD, Central Lab, Huadong Hospital, affiliated to Fudan University; X. Jiang, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; H. Shi, MD, Department of Osteoporosis and Bone Disease, Huadong Hospital, affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute; Y. Fan, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; W. Lin, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; Y. Zhu, MD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University; F. Yang, MD, PhD, Department of Orthopedics, Huadong Hospital, affiliated to Fudan University. Qun Cheng and Shangjin Lin are co-first authors of this article
| |
Collapse
|
24
|
Xie Y, Gao Y, Zhang L, Chen Y, Ge W, Tang P. Involvement of serum-derived exosomes of elderly patients with bone loss in failure of bone remodeling via alteration of exosomal bone-related proteins. Aging Cell 2018; 17:e12758. [PMID: 29603567 PMCID: PMC5946082 DOI: 10.1111/acel.12758] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2018] [Indexed: 12/31/2022] Open
Abstract
Exosomes are secreted into the blood by various types of cells. These extracellular vesicles are involved in the contribution of exosomal proteins to osteoblastic or osteoclastic regulatory networks during the failure of bone remodeling, which results in age-related bone loss. However, the molecular changes in serum-derived exosomes (SDEs) from aged patients with low bone density and their functions in bone remodeling remain to be fully elucidated. We present a quantitative proteomics analysis of exosomes purified from the serum of the elderly patients with osteoporosis/osteopenia and normal volunteers; these data are available via Proteome Xchange with the identifier PXD006463. Overall, 1,371 proteins were identified with an overlap of 1,160 Gene IDs among the ExoCarta proteins. Bioinformatics analysis and in vitro studies suggested that protein changes in SDEs of osteoporosis patients are not only involved in suppressing the integrin-mediated mechanosensation and activation of osteoblastic cells, but also trigger the differentiation and resorption of osteoclasts. In contrast, the main changes in SDEs of osteopenia patients facilitated both activation of osteoclasts and formation of new bone mass, which could result in a compensatory elevation in bone remodeling. While the SDEs from aged normal volunteers might play a protective role in bone health through facilitating adhesion of bone cells and suppressing aging-associated oxidative stress. This information will be helpful in elucidating the pathophysiological functions of SDEs and aid in the development of senile osteoporosis diagnostics and therapeutics.
Collapse
Affiliation(s)
- Yong Xie
- Department of Orthopedics; Chinese PLA General Hospital; Beijing China
| | - Yanpan Gao
- State Key Laboratory of Medical Molecular Biology; Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing China
| | - Licheng Zhang
- Department of Orthopedics; Chinese PLA General Hospital; Beijing China
| | - Yanyu Chen
- State Key Laboratory of Medical Molecular Biology; Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology; Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing China
| | - Peifu Tang
- Department of Orthopedics; Chinese PLA General Hospital; Beijing China
| |
Collapse
|
25
|
Herrmann M, Zeiter S, Eberli U, Hildebrand M, Camenisch K, Menzel U, Alini M, Verrier S, Stadelmann VA. Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study. Front Bioeng Biotechnol 2018; 6:5. [PMID: 29484293 PMCID: PMC5816045 DOI: 10.3389/fbioe.2018.00005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022] Open
Abstract
Bone is an organ with high natural regenerative capacity and most fractures heal spontaneously when appropriate fracture fixation is provided. However, additional treatment is required for patients with large segmental defects exceeding the endogenous healing potential and for patients suffering from fracture non-unions. These cases are often associated with insufficient vascularization. Transplantation of CD34+ endothelial progenitor cells (EPCs) has been successfully applied to promote neovascularization of bone defects, however including extensive ex vivo manipulation of cells. Here, we hypothesized, that treatment with granulocyte colony-stimulating factor (G-CSF) may improve bone healing by mobilization of CD34+ progenitor cells into the circulation, which in turn may facilitate vascularization at the defect site. In this pilot study, we aimed to characterize the different cell populations mobilized by G-CSF and investigate the influence of cell mobilization on the healing of a critical size femoral defect in rats. Cell mobilization was investigated by flow cytometry at different time points after five consecutive daily G-CSF injections. In a pilot study, bone healing of a 4.5-mm critical femoral defect in F344 rats was compared between a saline-treated control group and a G-CSF treatment group. In vivo microcomputed tomography and histology were applied to compare bone formation in both treatment groups. Our data revealed that leukocyte counts show a peak increase at the first day after the last G-CSF injection. In addition, we found that CD34+ progenitor cells, including EPCs, were significantly enriched at day 1, and further increased at day 5 and day 11. Upregulation of monocytes, granulocytes and macrophages peaked at day 1. G-CSF treatment significantly increased bone volume and bone density in the defect, which was confirmed by histology. Our data show that different cell populations are mobilized by G-CSF treatment in cell specific patterns. Although in this pilot study no bridging of the critical defect was observed, significantly improved bone formation by G-CSF treatment was clearly shown.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | | |
Collapse
|
26
|
Hoff P, Gaber T, Strehl C, Schmidt-Bleek K, Lang A, Huscher D, Burmester GR, Schmidmaier G, Perka C, Duda GN, Buttgereit F. Immunological characterization of the early human fracture hematoma. Immunol Res 2017; 64:1195-1206. [PMID: 27629117 DOI: 10.1007/s12026-016-8868-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The initial inflammatory phase of fracture healing is of great importance for the clinical outcome. We aimed to develop a detailed time-dependent analysis of the initial fracture hematoma. We analyzed the composition of immune cell subpopulations by flow cytometry and the concentration of cytokines and chemokines by bioplex in 42 samples from human fractures of long bones <72 h post-trauma. The early human fracture hematoma is characterized by maturation of granulocytes and migration of monocytes/macrophages and hematopoietic stem cells. Both T helper cells and cytotoxic T cells proliferate within the fracture hematoma and/or migrate to the fracture site. Humoral immunity characteristics comprise high concentration of pro-inflammatory cytokines such as IL-6, IL-8, IFNγ and TNFα, but also elevated concentration of anti-inflammatory cytokines, e.g., IL-1 receptor antagonist and IL-10. Furthermore, we found that cells of the fracture hematoma represent a source for key chemokines. Even under the bioenergetically restricted conditions that exist in the initial fracture hematoma, immune cells are not only present, but also survive, mature, function and migrate. They secrete a cytokine/chemokine cocktail that contributes to the onset of regeneration. We hypothesize that this specific microenvironment of the initial fracture hematoma is among the crucial factors that determine fracture healing.
Collapse
Affiliation(s)
- Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany.
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany.
| | - T Gaber
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| | - C Strehl
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
| | - K Schmidt-Bleek
- Julius Wolff Institute, Charité University Hospital, 13353, Berlin, Germany
| | - A Lang
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353, Berlin, Germany
| | - D Huscher
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
| | - G R Burmester
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
| | - G Schmidmaier
- Department of Orthopedics, University Hospital Heidelberg, 69118, Heidelberg, Germany
| | - C Perka
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
- Center for Musculoskeletal Surgery, Charité University Hospital, 10117, Berlin, Germany
| | - G N Duda
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
- Julius Wolff Institute, Charité University Hospital, 13353, Berlin, Germany
| | - F Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| |
Collapse
|
27
|
Joensuu K, Uusitalo-Kylmälä L, Hentunen TA, Heino TJ. Angiogenic potential of human mesenchymal stromal cell and circulating mononuclear cell cocultures is reflected in the expression profiles of proangiogenic factors leading to endothelial cell and pericyte differentiation. J Tissue Eng Regen Med 2017; 12:775-783. [PMID: 28593699 DOI: 10.1002/term.2496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/17/2022]
Abstract
Endothelial progenitors found among the peripheral blood (PB) mononuclear cells (MNCs) are interesting cells for their angiogenic properties. Mesenchymal stromal cells (MSCs) in turn can produce proangiogenic factors as well as differentiate into mural pericytes, making MSCs and MNCs an attractive coculture setup for regenerative medicine. In this study, human bone marrow-derived MSCs and PB-derived MNCs were cocultured in basal or osteoblastic medium without exogenously supplied growth factors to demonstrate endothelial cell, pericyte and osteoblastic differentiation. The expression levels of various proangiogenic factors, as well as endothelial cell, pericyte and osteoblast markers in cocultures were determined by quantitative polymerase chain reaction. Immunocytochemistry for vascular endothelial growth factor receptor-1 and α-smooth muscle actin as well as staining for alkaline phosphatase were performed after 10 and 14 days. Messenger ribonucleic acid expression of endothelial cell markers was highly upregulated in both basal and osteoblastic conditions after 5 days of coculture, indicating an endothelial cell differentiation, which was supported by immunocytochemistry for vascular endothelial growth factor receptor-1. Stromal derived factor-1 and vascular endothelial growth factor were highly expressed in MSC-MNC coculture in basal medium but not in osteoblastic medium. On the contrary, the expression levels of bone morphogenetic protein-2 and angiopoietin-1 were significantly higher in osteoblastic medium. Pericyte markers were highly expressed in both cocultures after 5 days. In conclusion, it was demonstrated endothelial cell and pericyte differentiation in MSC-MNC cocultures both in basal and osteoblastic medium indicating a potential for neovascularization for tissue engineering applications.
Collapse
Affiliation(s)
- Katriina Joensuu
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Liina Uusitalo-Kylmälä
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Teuvo A Hentunen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Terhi J Heino
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
28
|
Krasna M, Malicev E, Rozman JZ, Vrtovec B. Assessment of stability of CD34+ cell products enriched by immunoselection from peripheral blood mononuclear cells during refrigerated storage. Transfus Apher Sci 2017; 56:566-570. [PMID: 28800845 DOI: 10.1016/j.transci.2017.07.011] [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] [Received: 03/14/2017] [Revised: 06/15/2017] [Accepted: 07/10/2017] [Indexed: 01/24/2023]
Abstract
Durable engraftment of transplanted CD34+ cells largely depends on the quality of the cell product. Limited data are currently available about extended storage of immunoselected CD34+ cells. The aim of our study was to assess the stability of CD34+ cell product with the cells stored in high concentration (80×106 in 6mL) in small bags intended for cell implantation. Cell products were prepared by leukapheresis and immunoselection (Clinimacsplus procedure) from 13 patients with chronic dilated cardiomyopathy. CD34+ cell products were stored at 2-8°C and analyzed at time 0 (fresh products), 24, 48h, 4 and 6 days. Product viability, absolute number of viable CD34+ cells and apoptosis were determined by flow cytometry. Microbiological contamination of the cell products was tested by BACTEC system. The mean viability of CD34+ cells decreased by 2.7% within 24h, by 13.4% within 48h and by 37.5% within 6 days. The mean recovery of viable CD34+ cells was 91.1%, 74.8%, 66.3% and 56.2% at 24, 48h, 4 and 6 days, respectively. The mean fraction of early apoptotic cells in fresh and stored products was 4.9±3.5% at 0h, 5.9±3.8% at 24h, 4.2±3.1% at 48h, 6.3±2.6% at 4 days and 9.3±4.6% at 6 days. All products were negative for microbial contamination.
Collapse
Affiliation(s)
- Metka Krasna
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia.
| | - Elvira Malicev
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | | | - Bojan Vrtovec
- Department of Cardiology, University Medical Centre Ljubljana, Slovenia
| |
Collapse
|
29
|
Mifuji K, Ishikawa M, Kamei N, Tanaka R, Arita K, Mizuno H, Asahara T, Adachi N, Ochi M. Angiogenic conditioning of peripheral blood mononuclear cells promotes fracture healing. Bone Joint Res 2017; 6:489-498. [PMID: 28835445 PMCID: PMC5579315 DOI: 10.1302/2046-3758.68.bjr-2016-0338.r1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022] Open
Abstract
Objectives The objective of this study was to investigate the therapeutic effect of peripheral blood mononuclear cells (PBMNCs) treated with quality and quantity control culture (QQ-culture) to expand and fortify angiogenic cells on the acceleration of fracture healing. Methods Human PBMNCs were cultured for seven days with the QQ-culture method using a serum-free medium containing five specific cytokines and growth factors. The QQ-cultured PBMNCs (QQMNCs) obtained were counted and characterised by flow cytometry and real-time polymerase chain reaction (RT-PCR). Angiogenic and osteo-inductive potentials were evaluated using tube formation assays and co-culture with mesenchymal stem cells with osteo-inductive medium in vitro. In order to evaluate the therapeutic potential of QQMNCs, cells were transplanted into an immunodeficient rat femur nonunion model. The rats were randomised into three groups: control; PBMNCs; and QQMNCs. The fracture healing was evaluated radiographically and histologically. Results The total number of PBMNCs was decreased after QQ-culture, however, the number of CD34+ and CD206+ cells were found to have increased as assessed by flow cytometry analysis. In addition, gene expression of angiogenic factors was upregulated in QQMNCs. In the animal model, the rate of bone union was higher in the QQMNC group than in the other groups. Radiographic scores and bone volume were significantly associated with the enhancement of angiogenesis in the QQMNC group. Conclusion We have demonstrated that QQMNCs have superior potential to accelerate fracture healing compared with PBMNCs. The QQMNCs could be a promising option for fracture nonunion. Cite this article: K. Mifuji, M. Ishikawa, N. Kamei, R. Tanaka, K. Arita, H. Mizuno, T. Asahara, N. Adachi, M. Ochi. Angiogenic conditioning of peripheral blood mononuclear cells promotes fracture healing. Bone Joint Res 2017;6: 489–498. DOI: 10.1302/2046-3758.68.BJR-2016-0338.R1.
Collapse
Affiliation(s)
- K Mifuji
- Hiroshima University, Hiroshima, Japan
| | | | - N Kamei
- Hiroshima University, Hiroshima, Japan
| | - R Tanaka
- Juntendo University School of Medicine, Tokyo, Japan
| | - K Arita
- Juntendo University School of Medicine, Tokyo, Japan
| | - H Mizuno
- Juntendo University School of Medicine, Tokyo, Japan
| | - T Asahara
- Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - N Adachi
- Hiroshima University, Hiroshima, Japan
| | - M Ochi
- Hiroshima University, Hiroshima, Japan
| |
Collapse
|
30
|
Hertweck J, Ritz U, Götz H, Schottel PC, Rommens PM, Hofmann A. CD34 + cells seeded in collagen scaffolds promote bone formation in a mouse calvarial defect model. J Biomed Mater Res B Appl Biomater 2017; 106:1505-1516. [PMID: 28730696 DOI: 10.1002/jbm.b.33956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/29/2017] [Accepted: 07/04/2017] [Indexed: 11/10/2022]
Abstract
Bone tissue engineering (BTE) holds promise for managing the clinical problem of large bone defects. However, clinical adoption of BTE is limited due to limited vascularization of constructs, which could be circumvented by pre-cultivation of osteogenic and endothelial derived cells in natural-based polymer scaffolds. However, until now not many studies compared the effect of mono- and cocultures pre-seeded in collagen before implantation. We utilized a mouse calvarial defect model and compared five groups of collagen scaffolds: a negative control of a collagen scaffold alone, a positive control treated with BMP-7, monocultures of either human osteoblasts (hOBs) or CD34+ cells, and a coculture of hOB and CD34+ cells. Each pre-seeded collagen scaffold was implanted in mice. After 6 weeks mice were sacrificed and their skulls prepared for volumetric and histologic analysis. We found that a monoculture of CD34+ cells and a coculture of hOB and CD34+ cells pre-cultured in the collagen scaffold increased bone regeneration to a similar extend. In these groups, greater amounts of new bone were found compared with hOB monocultures. Interestingly, monoculture of CD34+ cells demonstrated better fracture healing than monoculture of hOBs, emphasizing the possible role of angiogenesis. Our results are promising regarding a cellular based collagen BTE construct, but more work is needed to understand the complex interaction between the osteogenic and endothelial cells. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1505-1516, 2018.
Collapse
Affiliation(s)
- Jens Hertweck
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hermann Götz
- Platform for Biomaterial Research, Biomatics, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | - Patrick C Schottel
- Department of Orthopedics and Rehabilitation, University of Vermont Medical Center, Burlington, Vermont
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
31
|
Abstract
Unlike many other postnatal tissues, bone can regenerate and repair itself; nevertheless, this capacity can be overcome. Traditionally, surgical reconstructive strategies have implemented autologous, allogeneic, and prosthetic materials. Autologous bone--the best option--is limited in supply and also mandates an additional surgical procedure. In regenerative tissue engineering, there are myriad issues to consider in the creation of a functional, implantable replacement tissue. Importantly, there must exist an easily accessible, abundant cell source with the capacity to express the phenotype of the desired tissue, and a biocompatible scaffold to deliver the cells to the damaged region. A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key advances in stem and progenitor cell contribution to the field of bone tissue engineering. In this review, we briefly introduce various adult stem cells implemented in bone tissue engineering such as mesenchymal stem cells (including bone marrow- and adipose-derived stem cells), endothelial progenitor cells, and induced pluripotent stem cells. We then discuss numerous advances associated with their application and subsequently focus on technological advances in the field, before addressing key regenerative strategies currently used in clinical practice. Stem and progenitor cell implementation in bone tissue engineering strategies have the ability to make a major impact on regenerative medicine and reduce patient morbidity. As the field of regenerative medicine endeavors to harness the body's own cells for treatment, scientific innovation has led to great advances in stem cell-based therapies in the past decade.
Collapse
|
32
|
D'Antimo C, Biggi F, Borean A, Di Fabio S, Pirola I. Combining a novel leucocyte-platelet-concentrated membrane and an injectable collagen scaffold in a single-step AMIC procedure to treat chondral lesions of the knee: a preliminary retrospective study. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2016; 27:673-681. [PMID: 27803980 DOI: 10.1007/s00590-016-1869-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/04/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Different surgical approaches are currently available to treat knee chondral defects. Microfracture is the most commonly applied, but it often leads to a mechanically inferior fibrocartilaginous tissue. To overcome this shortcoming, the Autologous, Matrix-Induced Chondrogenesis (AMIC) technique has been proposed. To further enhance the outcome of AMIC, the addition of haemoderivatives containing growth factors that stimulate cartilage healing has emerged as a new treatment method. Recently, a novel leucocyte-platelet-concentrated membrane (CLP-MB), highly enriched in platelets, monocytes/macrophages, fibrinogen, CD34+ and CD34+VEGFR-2+CD133+ cells, has been developed. Additionally, an injectable collagen scaffold (Cartifill) has been proposed as a replacement of the AMIC standard collagen membrane. AIMS This preliminary study is aimed to evaluate the short-term safety and efficacy of the use of the CLP-MB membrane and injectable collagen scaffold when combined in single-step AMIC procedures for the treatment of knee chondral lesions. METHODS Medical records of patients who underwent an AMIC procedure with the CLP-MB membrane combined with Cartifill were reviewed retrospectively. Follow-up assessments were conducted at 6 and 12 months after surgery. Clinical function was assessed on the basis of the International Knee Documentation Committee (IKDC) score. Pain was evaluated using the visual analogue scale (VAS). RESULTS Twenty-five patients were identified as meeting the inclusion criteria. Mean IKDC and VAS scores significantly improved during the follow-up time. The postoperative course was uneventful. CONCLUSIONS AMIC combined with the CLP-MB membrane, and Cartifill seems to be a promising approach to treat knee chondral defects.
Collapse
Affiliation(s)
- Corrado D'Antimo
- Orthopaedics and Traumatology Department, San Martino Hospital, Belluno, Italy.
| | - Francesco Biggi
- Orthopaedics and Traumatology Department, San Martino Hospital, Belluno, Italy
| | - Alessio Borean
- Immunohematology and Transfusion Medicine Department, San Martino Hospital, Belluno, Italy
| | - Stefano Di Fabio
- Orthopaedics and Traumatology Department, San Martino Hospital, Belluno, Italy
| | - Ivan Pirola
- Immunohematology and Transfusion Medicine Department, San Martino Hospital, Belluno, Italy
| |
Collapse
|
33
|
Park S, Zhao H, Urata M, Chai Y. Sutures Possess Strong Regenerative Capacity for Calvarial Bone Injury. Stem Cells Dev 2016; 25:1801-1807. [PMID: 27762665 DOI: 10.1089/scd.2016.0211] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Repair of calvarial bony defects remains challenging for craniofacial surgeons. Injury experiments on animal calvarial bones are widely used to study healing mechanisms and test tissue engineering approaches. Previously, we identified Gli1+ cells within the calvarial sutures as stem cells supporting calvarial bone turnover and injury repair. In this study, we tested the regenerative capacity of the suture region compared with other areas of calvarial bone. Injuries were made to mouse sagittal sutures or other areas of the calvarial bone at varying distances from the suture. Samples were collected at different time points after injury for evaluation. MicroCT and histological analyses were conducted. EdU incorporation analysis was performed to assay cell proliferation. Gli1-CreERT2;Tdtomatoflox mice were used to trace the fate of Gli1+ stem cells after injury. Calvarial sutures possess much stronger regeneration capability than the nonsuture bony areas of the calvaria. The healing rate of the calvarial bone is inversely proportional to the distance between the suture and injury site: injuries closer to the suture heal faster. After complete removal of the sagittal suture, regeneration and restoration of normal organization occur within 6 weeks. Gli1+ cells within the suture mesenchyme are the cellular source for injury repair and bone regeneration. These results demonstrate that calvarial bone healing is not an evenly distributed event on the calvarial surface. Sutures contain stem cells and are the origin of calvarial bone tissue regeneration. Therefore, current practice in calvarial surgery needs to be reevaluated and modified. These findings also necessitate the design of new approaches for repairing calvarial bony defects.
Collapse
Affiliation(s)
- Shery Park
- 1 Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California , Los Angeles, California
| | - Hu Zhao
- 2 Baylor College of Dentistry, Texas A&M University , Houston, Texas
| | - Mark Urata
- 1 Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California , Los Angeles, California
| | - Yang Chai
- 1 Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California , Los Angeles, California
| |
Collapse
|
34
|
Fortunato TM, Beltrami C, Emanueli C, De Bank PA, Pula G. Platelet lysate gel and endothelial progenitors stimulate microvascular network formation in vitro: tissue engineering implications. Sci Rep 2016; 6:25326. [PMID: 27141997 PMCID: PMC4855184 DOI: 10.1038/srep25326] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 04/14/2016] [Indexed: 12/22/2022] Open
Abstract
Revascularisation is a key step for tissue regeneration and complete organ engineering. We describe the generation of human platelet lysate gel (hPLG), an extracellular matrix preparation from human platelets able to support the proliferation of endothelial colony forming cells (ECFCs) in 2D cultures and the formation of a complete microvascular network in vitro in 3D cultures. Existing extracellular matrix preparations require addition of high concentrations of recombinant growth factors and allow only limited formation of capillary-like structures. Additional advantages of our approach over existing extracellular matrices are the absence of any animal product in the composition hPLG and the possibility of obtaining hPLG from patients to generate homologous scaffolds for re-implantation. This discovery has the potential to accelerate the development of regenerative medicine applications based on implantation of microvascular networks expanded ex vivo or the generation of fully vascularised organs.
Collapse
Affiliation(s)
- Tiago M Fortunato
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Cristina Beltrami
- Bristol Heart Institute, School of Clinical Sciences University of Bristol, Bristol, UK
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Sciences University of Bristol, Bristol, UK
| | - Paul A De Bank
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Giordano Pula
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|
35
|
Joensuu K, Uusitalo L, Alm JJ, Aro HT, Hentunen TA, Heino TJ. Enhanced osteoblastic differentiation and bone formation in co-culture of human bone marrow mesenchymal stromal cells and peripheral blood mononuclear cells with exogenous VEGF. Orthop Traumatol Surg Res 2015; 101:381-6. [PMID: 25813558 DOI: 10.1016/j.otsr.2015.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Despite recent advances in bone tissue engineering, efficient bone formation and vascularization remains a challenge for clinical applications. HYPOTHESIS The aim of this study was to investigate if the osteoblastic differentiation of human mesenchymal stromal cells (MSCs) can be enhanced by co-culturing them with peripheral blood (PB) mononuclear cells (MNCs), with and without vascular endothelial growth factor (VEGF), a coupling factor of bone formation and angiogenesis. MATERIALS AND METHODS Human bone marrow (BM) derived MSCs were co-cultured with PB-MNCs in osteogenic medium with or without VEGF. Osteoblastic differentiation and mineral deposition were studied by staining for alkaline phosphatase (ALP), and von Kossa, respectively, and measurements for ALP activity and calcium concentration (Ca). Cell proliferation was assayed with Alamar blue. The mechanism(s) were further studied by Transwell(®) cell culture experiments. RESULTS Both ALP and mineralization (von Kossa and Ca) were significantly higher in the MSC-MNC co-cultures compared to plain MSC cultures. VEGF alone had no effect on osteoblastic differentiation of MSCs, but further enhanced differentiation in co-culture settings. The mechanism was shown to require cell-cell contact between MSCs and MNCs and the factors contributing to further differentiation appear to be soluble. No differences were observed in cell proliferation. CONCLUSION Our study demonstrates that the in vitro ALP activity and mineralization of human BM-MSCs is more efficient in the presence of PB-MNCs, and exogenously added VEGF further enhances the stimulatory effect. This indicates that PB-MNCs could be a potential cell source in development of co-culture systems for novel tissue engineering applications for enhanced bone healing.
Collapse
Affiliation(s)
- K Joensuu
- Department of Cell Biology and Anatomy, Kiinamyllynkatu 10 C3, 20520 Turku, Finland.
| | - L Uusitalo
- Department of Cell Biology and Anatomy, Kiinamyllynkatu 10 C3, 20520 Turku, Finland
| | - J J Alm
- Orthopedic Research Unit, Department of Orthopedic Surgery and Traumatology, University of Turku, Turku, Finland
| | - H T Aro
- Orthopedic Research Unit, Department of Orthopedic Surgery and Traumatology, University of Turku, Turku, Finland
| | - T A Hentunen
- Department of Cell Biology and Anatomy, Kiinamyllynkatu 10 C3, 20520 Turku, Finland
| | - T J Heino
- Department of Cell Biology and Anatomy, Kiinamyllynkatu 10 C3, 20520 Turku, Finland; Orthopedic Research Unit, Department of Orthopedic Surgery and Traumatology, University of Turku, Turku, Finland
| |
Collapse
|
36
|
Zigdon-Giladi H, Rudich U, Michaeli Geller G, Evron A. Recent advances in bone regeneration using adult stem cells. World J Stem Cells 2015; 7:630-640. [PMID: 25914769 PMCID: PMC4404397 DOI: 10.4252/wjsc.v7.i3.630] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/30/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023] Open
Abstract
Bone is a highly vascularized tissue reliant on the close spatial and temporal association between blood vessels and bone cells. Therefore, cells that participate in vasculogenesis and osteogenesis play a pivotal role in bone formation during prenatal and postnatal periods. Nevertheless, spontaneous healing of bone fracture is occasionally impaired due to insufficient blood and cellular supply to the site of injury. In these cases, bone regeneration process is interrupted, which might result in delayed union or even nonunion of the fracture. Nonunion fracture is difficult to treat and have a high financial impact. In the last decade, numerous technological advancements in bone tissue engineering and cell-therapy opened new horizon in the field of bone regeneration. This review starts with presentation of the biological processes involved in bone development, bone remodeling, fracture healing process and the microenvironment at bone healing sites. Then, we discuss the rationale for using adult stem cells and listed the characteristics of the available cells for bone regeneration. The mechanism of action and epigenetic regulations for osteogenic differentiation are also described. Finally, we review the literature for translational and clinical trials that investigated the use of adult stem cells (mesenchymal stem cells, endothelial progenitor cells and CD34+ blood progenitors) for bone regeneration.
Collapse
|
37
|
Schmidt-Bleek K, Kwee BJ, Mooney DJ, Duda GN. Boon and Bane of Inflammation in Bone Tissue Regeneration and Its Link with Angiogenesis. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:354-64. [PMID: 25742724 DOI: 10.1089/ten.teb.2014.0677] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Delayed healing or nonhealing of bone is an important clinical concern. Although bone, one of the two tissues with scar-free healing capacity, heals in most cases, healing is delayed in more than 10% of clinical cases. Treatment of such delayed healing condition is often painful, risky, time consuming, and expensive. Tissue healing is a multistage regenerative process involving complex and well-orchestrated steps, which are initiated in response to injury. At best, these steps lead to scar-free tissue formation. At the onset of healing, during the inflammatory phase, stationary and attracted macrophages and other immune cells at the fracture site release cytokines in response to injury. This initial reaction to injury is followed by the recruitment, proliferation, and differentiation of mesenchymal stromal cells, synthesis of extracellular matrix proteins, angiogenesis, and finally tissue remodeling. Failure to heal is often associated with poor revascularization. Since blood vessels mediate the transport of circulating cells, oxygen, nutrients, and waste products, they appear essential for successful healing. The strategy of endogenous regeneration in a tissue such as bone is interesting to analyze since it may represent a blueprint of successful tissue formation. This review highlights the interdependency of the time cascades of inflammation, angiogenesis, and tissue regeneration. A better understanding of these inter-relations is mandatory to early identify patients at risk as well as to overcome critical clinical conditions that limit healing. Instead of purely tolerating the inflammatory phase, modulations of inflammation (immunomodulation) might represent a valid therapeutic strategy to enhance angiogenesis and foster later phases of tissue regeneration.
Collapse
Affiliation(s)
- Katharina Schmidt-Bleek
- 1 Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Brian J Kwee
- 3 The Mooney Lab: Laboratory for Cell and Tissue Engineering, Harvard-School of Engineering and Applied Sciences , Cambridge, Massachusetts
| | - David J Mooney
- 3 The Mooney Lab: Laboratory for Cell and Tissue Engineering, Harvard-School of Engineering and Applied Sciences , Cambridge, Massachusetts
| | - Georg N Duda
- 1 Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| |
Collapse
|
38
|
Kittaka M, Kajiya M, Shiba H, Takewaki M, Takeshita K, Khung R, Fujita T, Iwata T, Nguyen TQ, Ouhara K, Takeda K, Fujita T, Kurihara H. Clumps of a mesenchymal stromal cell/extracellular matrix complex can be a novel tissue engineering therapy for bone regeneration. Cytotherapy 2015; 17:860-73. [PMID: 25743634 DOI: 10.1016/j.jcyt.2015.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 01/17/2015] [Accepted: 01/27/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND AIMS The transplantation of mesenchymal stromal cells (MSCs) to damaged tissue has attracted attention in scientific and medical fields as an effective regenerative therapy. Nevertheless, additional studies are required to develop an MSC transplant method for bone regeneration because the use of an artificial scaffold restricts the number of transplanted cells and their function. Furthermore, regulating the degree of cell differentiation in vitro is desirable for a more effective regenerative therapy. To address these unresolved issues, with the use of a self-produced extracellular matrix (ECM), we developed clumps of an MSC/ECM complex (C-MSCs). METHODS MSCs isolated from rat femur were cultured in growth medium supplemented with 50 μg/mL of ascorbic acid for 7 days. To obtain C-MSCs, confluent cells were scratched with the use of a micropipette tip to roll up the cellular sheet, which consisted of ECM produced by the MSCs. The biological properties of C-MSCs were assessed in vitro and their bone regenerative activity was tested by use of a rat calvarial defect model. RESULTS Immunofluorescent confocal microscopic analysis revealed that type I collagen formed C-MSCs. Osteopontin messenger RNA expression and amount of calcium content were higher in C-MSCs cultured in osteo-inductive medium than those of untreated C-MSCs. The transplantation of osteogenic-differentiated C-MSCs led to rapid bone regeneration in the rat calvarial defect model. CONCLUSIONS These results suggest that the use of C-MSCs refined by self-produced ECM, which contain no artificial scaffold and can be processed in vitro, may represent a novel tissue engineering therapy.
Collapse
Affiliation(s)
- Mizuho Kittaka
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Shiba
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Manabu Takewaki
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kei Takeshita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rathvisal Khung
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takako Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Truong Quoc Nguyen
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Katsuhiro Takeda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tsuyoshi Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
39
|
Grayson WL, Bunnell BA, Martin E, Frazier T, Hung BP, Gimble JM. Stromal cells and stem cells in clinical bone regeneration. Nat Rev Endocrinol 2015; 11:140-50. [PMID: 25560703 PMCID: PMC4338988 DOI: 10.1038/nrendo.2014.234] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stem-cell-mediated bone repair has been used in clinical trials for the regeneration of large craniomaxillofacial defects, to slow the process of bone degeneration in patients with osteonecrosis of the femoral head and for prophylactic treatment of distal tibial fractures. Successful regenerative outcomes in these investigations have provided a solid foundation for wider use of stromal cells in skeletal repair therapy. However, employing stromal cells to facilitate or enhance bone repair is far from being adopted into clinical practice. Scientific, technical, practical and regulatory obstacles prevent the widespread therapeutic use of stromal cells. Ironically, one of the major challenges lies in the limited understanding of the mechanisms via which transplanted cells mediate regeneration. Animal models have been used to provide insight, but these models largely fail to reproduce the nuances of human diseases and bone defects. Consequently, the development of targeted approaches to optimize cell-mediated outcomes is difficult. In this Review, we highlight the successes and challenges reported in several clinical trials that involved the use of bone-marrow-derived mesenchymal or adipose-tissue-derived stromal cells. We identify several obstacles blocking the mainstream use of stromal cells to enhance skeletal repair and highlight technological innovations or areas in which novel techniques might be particularly fruitful in continuing to advance the field of skeletal regenerative medicine.
Collapse
Affiliation(s)
- Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Bruce A Bunnell
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Elizabeth Martin
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Trivia Frazier
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Ben P Hung
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Jeffrey M Gimble
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| |
Collapse
|
40
|
Paul AJ, Momier D, Boukhechba F, Michiels JF, Lagadec P, Rochet N. Effect of G-CSF on the osteoinductive property of a BCP/blood clot composite. J Biomed Mater Res A 2015; 103:2830-8. [DOI: 10.1002/jbm.a.35424] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 01/19/2015] [Accepted: 02/04/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Adrien J. Paul
- Université Nice Sophia Antipolis, iBV, UMR7277; Nice 06100 France
- CNRS, iBV, UMR7277; Nice 06100 France
- Inserm, iBV, U1091; Nice 06100 France
- Université Nice Sophia Antipolis, UFR odontologie; Nice 06000 France
- Centre Hospitalier Universitaire, Pôle d'odontologie; Nice 06000 France
| | - David Momier
- Université Nice Sophia Antipolis, iBV, UMR7277; Nice 06100 France
- CNRS, iBV, UMR7277; Nice 06100 France
- Inserm, iBV, U1091; Nice 06100 France
| | - Florian Boukhechba
- Université Nice Sophia Antipolis, iBV, UMR7277; Nice 06100 France
- CNRS, iBV, UMR7277; Nice 06100 France
- Inserm, iBV, U1091; Nice 06100 France
- Graftys, 13854 Aix En Provence; France
| | | | - Patricia Lagadec
- Université Nice Sophia Antipolis, iBV, UMR7277; Nice 06100 France
- CNRS, iBV, UMR7277; Nice 06100 France
- Inserm, iBV, U1091; Nice 06100 France
| | - Nathalie Rochet
- Université Nice Sophia Antipolis, iBV, UMR7277; Nice 06100 France
- CNRS, iBV, UMR7277; Nice 06100 France
- Inserm, iBV, U1091; Nice 06100 France
| |
Collapse
|
41
|
Park SS, Bauer G, Abedi M, Pontow S, Panorgias A, Jonnal R, Zawadzki RJ, Werner JS, Nolta J. Intravitreal autologous bone marrow CD34+ cell therapy for ischemic and degenerative retinal disorders: preliminary phase 1 clinical trial findings. Invest Ophthalmol Vis Sci 2014; 56:81-9. [PMID: 25491299 DOI: 10.1167/iovs.14-15415] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Because human bone marrow (BM) CD34+ stem cells home into damaged tissue and may play an important role in tissue repair, this pilot clinical trial explored the safety and feasibility of intravitreal autologous CD34+ BM cells as potential therapy for ischemic or degenerative retinal conditions. METHODS This prospective study enrolled six subjects (six eyes) with irreversible vision loss from retinal vascular occlusion, hereditary or nonexudative age-related macular degeneration, or retinitis pigmentosa. CD34+ cells were isolated under Good Manufacturing Practice conditions from the mononuclear cellular fraction of the BM aspirate using a CliniMACs magnetic cell sorter. After intravitreal CD34+ cell injection, serial ophthalmic examinations, microperimetry/perimetry, fluorescein angiography, electroretinography (ERG), optical coherence tomography (OCT), and adaptive optics OCT were performed during the 6-month follow-up. RESULTS A mean of 3.4 million (range, 1-7 million) CD34+ cells were isolated and injected per eye. The therapy was well tolerated with no intraocular inflammation or hyperproliferation. Best-corrected visual acuity and full-field ERG showed no worsening after 6 months. Clinical examination also showed no worsening during follow-up except among age-related macular degeneration subjects in whom mild progression of geographic atrophy was noted in both the study eye and contralateral eye at 6-month follow-up, concurrent with some possible decline on multifocal ERG and microperimetry. Cellular in vivo imaging using adaptive optics OCT showed changes suggestive of new cellular incorporation into the macula of the hereditary macular degeneration study eye. CONCLUSIONS Intravitreal autologous BM CD34+ cell therapy appears feasible and well tolerated in eyes with ischemic or degenerative retinal conditions and merits further exploration. (ClinicalTrials.gov number, NCT01736059.).
Collapse
Affiliation(s)
- Susanna S Park
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Gerhard Bauer
- Institute for Regenerative Cures, University of California-Davis School of Medicine, Sacramento, California, United States
| | - Mehrdad Abedi
- Division of Hematology and Oncology, University of California-Davis Cancer Center, Sacramento, California, United States
| | - Suzanne Pontow
- Institute for Regenerative Cures, University of California-Davis School of Medicine, Sacramento, California, United States
| | - Athanasios Panorgias
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Ravi Jonnal
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Robert J Zawadzki
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - John S Werner
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Jan Nolta
- Institute for Regenerative Cures, University of California-Davis School of Medicine, Sacramento, California, United States
| |
Collapse
|
42
|
CD34/CD133 enriched bone marrow progenitor cells promote neovascularization of tissue engineered constructs in vivo. Stem Cell Res 2014; 13:465-77. [DOI: 10.1016/j.scr.2014.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 08/22/2014] [Accepted: 10/13/2014] [Indexed: 12/12/2022] Open
|
43
|
Imazu M, Takahama H, Asanuma H, Funada A, Sugano Y, Ohara T, Hasegawa T, Asakura M, Kanzaki H, Anzai T, Kitakaze M. Pathophysiological impact of serum fibroblast growth factor 23 in patients with nonischemic cardiac disease and early chronic kidney disease. Am J Physiol Heart Circ Physiol 2014; 307:H1504-11. [PMID: 25217649 DOI: 10.1152/ajpheart.00331.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Although the important role of fibroblast growth factor (FGF)23 on cardiac remodeling has been suggested in advanced chronic kidney disease (CKD), little is known about serum (s)FGF23 levels in patients with heart failure (HF) due to nonischemic cardiac disease (NICD) and early CKD. The present study aimed to investigate sFGF23 levels in NICD patients and identify the responsible factors for the elevation of sFGF23 levels. We prospectively measured sFGF23 levels in consecutive hospitalized NICD patients with early CKD (estimated glomerular filtration rate ≥ 40 ml·min(-1)·1.73 m(-2)) and analyzed the data of both echocardiography and right heart catheterization. Of the 156 NICD patients (estimated glomerular filtration rate range: 41-128 ml·min(-1)·1.73 m(-2)), the most severe HF symptom (New York Heart Association class III-IV, 53% vs. 33%, P = 0.015) was found in the above median sFGF23 (39.1 pg/ml) group compared with the below median sFGF23 group. sFGF23 levels were higher in patients with HF hospitalization history compared with those without HF [median: 46.8 (interquartile range: 38.8-62.7) vs. 34.7 (interquartile range: 29.6-42.4) pg/ml, P < 0.0001]. In the multivariate analysis, HF hospitalization was independently related to elevated sFGF23 levels (P = 0.022). Both systolic dysfunction and high plasma aldosterone concentration were identified as predictors of high sFGF23 levels (P < 0.05). Among the neurohormonal parameters, elevated sFGF23 levels were the only factor to predict a declining left ventricular ejection fraction (P = 0.001). These findings suggest that the progression of HF per se contributes to the elevation of sFGF23 levels even in the early stages of CKD, which leads to further myocardial dysfunction, potentially creating a vicious cycle.
Collapse
Affiliation(s)
- Miki Imazu
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hiroyuki Takahama
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hiroshi Asanuma
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan; and
| | - Akira Funada
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yasuo Sugano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Takahiro Ohara
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Takuya Hasegawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Masanori Asakura
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan; Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hideaki Kanzaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Masafumi Kitakaze
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan; Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
44
|
Ritz U, Spies V, Mehling I, Gruszka D, Rommens PM, Hofmann A. Mobilization of CD34+-progenitor cells in patients with severe trauma. PLoS One 2014; 9:e97369. [PMID: 24826895 PMCID: PMC4020858 DOI: 10.1371/journal.pone.0097369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/17/2014] [Indexed: 01/31/2023] Open
Abstract
Circulating CD34+ progenitor cells () gained importance in the field of regenerative medicine due to their potential to home in on injury sites and differentiate into cells of both endothelial and osteogenic lineages. In this study, we analyzed the mobilization kinetics and the numbers of CD34+, CD31+, CD45+, and CD133+ cells in twenty polytrauma patients (n = 13 male, n = 7 female, mean age 46.5±17.2 years, mean injury severity score (ISS) 35.8±12.5 points). In addition, the endothelial differentiation capacity of enriched CD34+cells was assessed by analyzing DiI-ac-LDL/lectin uptake, the expression of endothelial markers, and the morphological characteristics of these cells in Matrigel and spheroid cultures. We found that on days 1, 3, and 7 after a major trauma, the number of CD34+cells increased from 6- up to 12-fold (p<0.0001) over the number of CD34+cells from a control population of healthy, age-matched volunteers. The numbers of CD31+ cells were consistently higher on days 1 (1.4-fold, p<0.01) and 7 (1.3-fold, p<0.01), whereas the numbers of CD133+ cell did not change during the time course of investigation. Expression of endothelial marker molecules in CD34+cells was significantly induced in the polytrauma patients. In addition, we show that the CD34+ cell levels in severely injured patients were not correlated with clinical parameters, such as the ISS score, the acute physiology and chronic health evaluation II score (APACHE II), as well as the sequential organ failure assessment score (SOFA-2). Our results clearly indicate that pro-angiogenic cells are systemically mobilized after polytrauma and that their numbers are sufficient for the development of novel therapeutic models in regenerative medicine.
Collapse
Affiliation(s)
- Ulrike Ritz
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Volker Spies
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Isabella Mehling
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Dominik Gruszka
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Pol Maria Rommens
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Alexander Hofmann
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
- * E-mail:
| |
Collapse
|