1
|
Prithiviraj S, Garcia Garcia A, Linderfalk K, Yiguang B, Ferveur S, Falck LN, Subramaniam A, Mohlin S, Hidalgo Gil D, Dupard SJ, Zacharaki D, Raina DB, Bourgine PE. Compositional editing of extracellular matrices by CRISPR/Cas9 engineering of human mesenchymal stem cell lines. eLife 2025; 13:RP96941. [PMID: 40152921 PMCID: PMC11952750 DOI: 10.7554/elife.96941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Tissue engineering strategies predominantly rely on the production of living substitutes, whereby implanted cells actively participate in the regenerative process. Beyond cost and delayed graft availability, the patient-specific performance of engineered tissues poses serious concerns on their clinical translation ability. A more exciting paradigm consists in exploiting cell-laid, engineered extracellular matrices (eECMs), which can be used as off-the-shelf materials. Here, the regenerative capacity solely relies on the preservation of the eECM structure and embedded signals to instruct an endogenous repair. We recently described the possibility to exploit custom human stem cell lines for eECM manufacturing. In addition to the conferred standardization, the availability of such cell lines opened avenues for the design of tailored eECMs by applying dedicated genetic tools. In this study, we demonstrated the exploitation of CRISPR/Cas9 as a high precision system for editing the composition and function of eECMs. Human mesenchymal stromal/stem cell (hMSC) lines were modified to knock out vascular endothelial growth factor (VEGF) and Runt-related transcription factor 2 (RUNX2) and assessed for their capacity to generate osteoinductive cartilage matrices. We report the successful editing of hMSCs, subsequently leading to targeted VEGF and RUNX2-knockout cartilage eECMs. Despite the absence of VEGF, eECMs retained full capacity to instruct ectopic endochondral ossification. Conversely, RUNX2-edited eECMs exhibited impaired hypertrophy, reduced ectopic ossification, and superior cartilage repair in a rat osteochondral defect. In summary, our approach can be harnessed to identify the necessary eECM factors driving endogenous repair. Our work paves the road toward the compositional eECMs editing and their exploitation in broad regenerative contexts.
Collapse
Affiliation(s)
- Sujeethkumar Prithiviraj
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Alejandro Garcia Garcia
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Karin Linderfalk
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Bai Yiguang
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
- Department of Orthopaedics, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College NanchongSichuanChina
| | - Sonia Ferveur
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Ludvig Nilsén Falck
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund UniversityLundSweden
| | | | - Sofie Mohlin
- Division of Pediatrics, Clinical Sciences, Translational Cancer Research, Lund University Cancer Center at Medicon VillageLundSweden
| | - David Hidalgo Gil
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Steven J Dupard
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Dimitra Zacharaki
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| | - Deepak Bushan Raina
- The Faculty of Medicine, Department of Clinical Sciences Lund, OrthopedicsLundSweden
| | - Paul E Bourgine
- Cell, Tissue & Organ Engineering Laboratory, BMC, Department of Clinical Sciences, Lund UniversityLundSweden
- Wallenberg Centre for Molecular Medicine, Lund Stem Cell Centre, Lund University Cancer Centre, Lund UniversityLundSweden
| |
Collapse
|
2
|
Zhao Q, Zhang X, Li Y, He Z, Qin K, Buhl EM, Mert Ü, Horst K, Hildebrand F, Balmayor ER, Greven J. Porcine Mandibular Bone Marrow-Derived Mesenchymal Stem Cell (BMSC)-Derived Extracellular Vesicles Can Promote the Osteogenic Differentiation Capacity of Porcine Tibial-Derived BMSCs. Pharmaceutics 2024; 16:279. [PMID: 38399333 PMCID: PMC10893405 DOI: 10.3390/pharmaceutics16020279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Existing research suggests that bone marrow-derived mesenchymal stem cells (BMSCs) may promote endogenous bone repair. This may be through the secretion of factors that stimulate repair processes or directly through differentiation into osteoblast-progenitor cells. However, the osteogenic potential of BMSCs varies among different tissue sources (e.g., mandibular versus long BMSCs). The main aim of this study was to investigate the difference in osteogenic differentiation capacity between mandibular BMSCs (mBMSCs) and tibial BMSCs (tBMSCs). MATERIALS AND METHODS Bioinformatics analysis of the GSE81430 dataset taken from the Gene Expression Omnibus (GEO) database was performed using GEO2R. BMSCs were isolated from mandibular and tibial bone marrow tissue samples. Healthy pigs (n = 3) (registered at the State Office for Nature, Environment, and Consumer Protection, North Rhine-Westphalia (LANUV) 81-02.04.2020.A215) were used for this purpose. Cell morphology and osteogenic differentiation were evaluated in mBMSCs and tBMSCs. The expression levels of toll-like receptor 4 (TLR4) and nuclear transcription factor κB (NF-κB) were analyzed using quantitative polymerase chain reaction (qPCR) and Western blot (WB), respectively. In addition, mBMSC-derived extracellular vesicles (mBMSC-EVs) were gained and used as osteogenic stimuli for tBMSCs. Cell morphology and osteogenic differentiation capacity were assessed after mBMSC-EV stimulation. RESULTS Bioinformatic analysis indicated that the difference in the activation of the TLR4/NF-κB pathway was more pronounced compared to all other examined genes. Specifically, this demonstrated significant downregulation, whereas only 5-7 upregulated genes displayed significant variances. The mBMSC group showed stronger osteogenic differentiation capacity compared to the tBMSC group, confirmed via ALP, ARS, and von Kossa staining. Furthermore, qPCR and WB analysis revealed a significant decrease in the expression of the TLR4/NF-κB pathway in the mBMSC group compared to the tBMSC group (TLR4 fold changes: mBMSCs vs. tBMSCs p < 0.05; NF-κB fold changes: mBMSCs vs. tBMSCs p < 0.05). The osteogenic differentiation capacity was enhanced, and qPCR and WB analysis revealed a significant decrease in the expression of TLR4 and NF-κB in the tBMSC group with mBMSC-EVs added compared to tBMSCs alone (TLR4 fold changes: p < 0.05; NF-κB fold changes: p < 0.05). CONCLUSION Our results indicate that mBMSC-EVs can promote the osteogenic differentiation of tBMSCs in vitro. The results also provide insights into the osteogenic mechanism of mBMSCs via TLR4/NF-κB signaling pathway activation. This discovery promises a fresh perspective on the treatment of bone fractures or malunions, potentially offering a novel therapeutic method.
Collapse
Affiliation(s)
- Qun Zhao
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Xing Zhang
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - You Li
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Zhizhen He
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Kang Qin
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Shoulder and Elbow Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology and Medical Clinic II, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Ümit Mert
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Klemens Horst
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Elizabeth R. Balmayor
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Johannes Greven
- Experimental Orthopedics and Trauma Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| |
Collapse
|
3
|
Ribeiro M, Santos KC, Macedo MR, de Souza GA, Neto FIDA, Araujo GHM, Cavalcante DR, Costa FF, de Sá Ferreira G, Peixoto LA, de Miranda Moraes J, Vulcani VAS. Use of adipose derived stem cells accelerates the healing process in third-degree burns. Burns 2024; 50:132-145. [PMID: 37741785 DOI: 10.1016/j.burns.2023.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/25/2023] [Accepted: 08/20/2023] [Indexed: 09/25/2023]
Abstract
INTRODUCTION Burns are defined as a traumatic injury, usually of thermal origin, that affects the epithelial and adjacent tissue and is classified according to the depth reached. Tissue repair involved in this type of injury is often a challenge both due to its severity and the multiplicity of complications. Regenerative medicine has focused on the use of low-level laser photobiomodulation therapy (LLLT) and adipose-derived stem cells (ADSC), especially in the early stages of the process, to promote better healing and shorten repair time. Therefore, aim of this study was to evaluate the action of LLLT (660 nm) and ADSC in the repair process of burned skin tissue and investigate the association of the techniques (LLLT and ADSC). MATERIALS AND METHODS An in vivo study was carried out using 96 rats (Wister) with a scald burn model at a temperature of 95ºC, exposing the animal's back for 14 s. Animals were randomized into seven groups and three periods, five, 14 and 21 days. The groups included GC: Control group, ADSC-: Group treated with CD49d negative cells, ADSC+ : Group treated with positive CD49d cells, CULT: Group treated with conventional isolation cells, LLLT: Group treated only with LLLT Low Power Laser, ADSC-LLLT: Group treated with CD49d negative cells and LLLT. ADSC+LLLT: Group treated with positive CD49d cells and LLLT. The groups treated with LLLT (660 nm; 5 J/cm2) received irradiation three times a week, on alternate days for five, 14 and 21 days, according to the time of biopsy. ADSC-treated groups received one to three applications of the cells in a total volume of 1000 μL starting soon after the surgical debridement of the burn. Photographic monitoring was carried out at 5, 14 and 21 days after the beginning of the experiment to assess the degree of lesion contraction. Macroscopic, morphometric and histopathological analyzes were performed. RESULTS We showed significant re-epithelialization as well as an improvement in the healing process in the ADSC+, LLLT and ADSC+LLLT groups. We observed effects in the reduction of the inflammatory phase, increase in angiogenesis, decrease in oedema, greater collagen deposition, and better organization of the extracellular matrix compared to the other treatments. Moreover, the immunomagnetic separation of ADSC cells through the expression of the CD49d protein proved to be a useful means to obtain a more homogeneous population of cells with a role in tissue regeneration compared to the ADSC- and CULT groups. CONCLUSION In conclusion, the association of ADSC+ with LLLT was effective in accelerating the burn repair process, stimulating cell proliferation and formation of more normal skin tissue.
Collapse
Affiliation(s)
- Maisa Ribeiro
- Medicine Course, University Center of Mineiros, Mineiros, Goiás, Brazil; School of Veterinary and Zootechnics, Federal University of Goiás, Goiânia, Goiás, Brazil.
| | | | - Mathias Rezende Macedo
- Medicine Course, Health Sciences Academic Unit, Federal University of Jataí, Jataí, Goiás, Brazil
| | | | | | | | | | - Flavia Ferreira Costa
- Medicine Course, Health Sciences Academic Unit, Federal University of Jataí, Jataí, Goiás, Brazil
| | - Gabriel de Sá Ferreira
- Medicine Course, Health Sciences Academic Unit, Federal University of Jataí, Jataí, Goiás, Brazil
| | - Larissa Alves Peixoto
- Medicine Course, Health Sciences Academic Unit, Federal University of Jataí, Jataí, Goiás, Brazil
| | - Júlia de Miranda Moraes
- Medicine Course, Health Sciences Academic Unit, Federal University of Jataí, Jataí, Goiás, Brazil
| | - Valcinir Aloísio Scalla Vulcani
- School of Veterinary and Zootechnics, Federal University of Goiás, Goiânia, Goiás, Brazil; Veterinary Medicine Course, Agricultural Sciences Academic Unit, Federal University of Jataí, Jataí, Goiás, Brazil
| |
Collapse
|
4
|
Bacevich BM, Smith RDJ, Reihl AM, Mazzocca AD, Hutchinson ID. Advances with Platelet-Rich Plasma for Bone Healing. Biologics 2024; 18:29-59. [PMID: 38299120 PMCID: PMC10827634 DOI: 10.2147/btt.s290341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/17/2024] [Indexed: 02/02/2024]
Abstract
Despite significant advances in the understanding and delivery of osteosynthesis, fracture non-union remains a challenging clinical problem in orthopaedic surgery. To bridge the gap, basic science characterization of fracture healing provides a platform to identify and target biological strategies to enhance fracture healing. Of immense interest, Platelet-rich plasma (PRP) is a point of care orthobiologic that has been extensively studied in bone and soft tissue healing given its relative ease of translation from the benchtop to the clinic. The aim of this narrative review is to describe and relate pre-clinical in-vitro and in-vivo findings to clinical observations investigating the efficacy of PRP to enhance bone healing for primary fracture management and non-union treatment. A particular emphasis is placed on the heterogeneity of PRP preparation techniques, composition, activation strategies, and delivery. In the context of existing data, the routine use of PRP to enhance primary fracture healing and non-union management cannot be supported. However, it is acknowledged that extensive heterogeneity of PRP treatments in clinical studies adds obscurity; ultimately, refinement (and consensus) of PRP treatments for specific clinical indications, including repetition studies are warranted.
Collapse
Affiliation(s)
- Blake M Bacevich
- Division of Sports Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Brigham, Boston, MA, USA
| | - Richard David James Smith
- Division of Sports Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Brigham, Boston, MA, USA
| | - Alec M Reihl
- Division of Sports Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Brigham, Boston, MA, USA
| | - Augustus D Mazzocca
- Division of Sports Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Brigham, Boston, MA, USA
- Medical Director, Division of Sports Medicine, Department of Orthopaedic Surgery, Massachusetts General Brigham, Boston, MA, USA
| | - Ian D Hutchinson
- Division of Sports Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Brigham, Boston, MA, USA
| |
Collapse
|
5
|
Tantuway V, Jeyaraman M, Nallakumarasamy A, Prikh MB, Sharma AK, Sharma R. Functional Outcome Analysis of Autologous Stromal Vascular Fraction (SVF) (Sahaj Therapy ®) Using Direct Sonication in Osteonecrosis of the Femoral Head (ONFH): A 6-Year Follow-Up Study. Indian J Orthop 2024; 58:68-78. [PMID: 38161400 PMCID: PMC10754810 DOI: 10.1007/s43465-023-01041-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/28/2023] [Indexed: 01/03/2024]
Abstract
Introduction We investigated the safety, efficacy, functional, and clinical outcomes of intra-osseous implantation of mechanically isolated, autologous stromal vascular fraction (SVF), an Australian patented direct ultrasonication technology (Sahaj Therapy®) in osteonecrosis of the femoral head (ONFH). Materials and Methods A total of 32 cases of ONFH were enrolled in the study after confirming with an MRI of the affected hip. All cases were treated with an intra-osseous autologous SVF implantation [4-5 cc with the cellular dosage of 8.0 × 107 cells with a viability of > 85% SVF cells] on the same surgical sitting. All the cases were followed up clinically, functionally, and radiologically at regular intervals. A comparison of mean HOOS scores at different follow-ups was done using Paired 't'-test. A P value of < 0.05 was considered significant. Results In our study, male preponderance was seen (53.1%). According to the modified Ficat and Arlet classification, the most common grade of ONFH was grade 2 [right: 25 hips and left: 25 hips]. There was a statistically significant improvement in the mean HOOS score of the right hip (n = 10) and left hip (n = 9) from preoperative time till 72 months (P < 0.05). The follow-up MRI of the affected hips shows improved osteogenesis without any further worsening of the contour of the femoral head. No adverse effects were seen in any of the study participants. Conclusion For individuals with ONFH, treated with intra-osseous autologous SVF implantation in the same surgical procedure is an innovative and promising treatment modality. Even after 6 years of follow-up, the study participants with ONFH have shown good clinical and functional outcomes with autologous SVF.
Collapse
Affiliation(s)
- Vinay Tantuway
- Department of Orthopaedics and Traumatology, Index Medical College Hospital and Research Centre, Indore, Madhya Pradesh India
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| | - Madhan Jeyaraman
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX USA
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
| | - Mittal B. Prikh
- Department of Orthopaedics, Navjivan Hospital, Ahmedabad, Gujarat India
| | - Aashish K. Sharma
- Department of Orthopaedics and Joint Replacement, CK Birla Hospitals, Jaipur, Rajasthan India
| | - Raj Sharma
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| |
Collapse
|
6
|
Wu L, Liu Z, Xiao L, Ai M, Cao Y, Mao J, Song K. The Role of Gli1 + Mesenchymal Stem Cells in Osteogenesis of Craniofacial Bone. Biomolecules 2023; 13:1351. [PMID: 37759749 PMCID: PMC10526808 DOI: 10.3390/biom13091351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Glioma-associated oncogene homolog 1 (Gli1) is a transcriptional activator of hedgehog (Hh) signaling that regulates target gene expression and several cellular biological processes. Cell lineage tracing techniques have highlighted Gli1 as an ideal marker for mesenchymal stem cells (MSCs) in vivo. Gli1+ MSCs are critical for the osteogenesis of the craniofacial bone; however, the regulatory mechanism by which Gli1+ MSCs mediate the bone development and tissue regeneration of craniofacial bone has not been systematically outlined. This review comprehensively elucidates the specific roles of Gli1+ MSCs in craniofacial bone osteogenesis. In addition to governing craniofacial bone development, Gli1+ MSCs are associated with the tissue repair of craniofacial bone under pathological conditions. Gli1+ MSCs promote intramembranous and endochondral ossification of the craniofacial bones, and assist the osteogenesis of the craniofacial bone by improving angiopoiesis. This review summarizes the novel role of Gli1+ MSCs in bone development and tissue repair in craniofacial bones, which offers new insights into bone regeneration therapy.
Collapse
Affiliation(s)
- Laidi Wu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| | - Zhixin Liu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| | - Li Xiao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| | - Mi Ai
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| | - Jing Mao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regen-Eration, Wuhan 430022, China
| |
Collapse
|
7
|
Tsai MH, Megat Abdul Wahab R, Zainal Ariffin SH, Azmi F, Yazid F. Enhanced Osteogenesis Potential of MG-63 Cells through Sustained Delivery of VEGF via Liposomal Hydrogel. Gels 2023; 9:562. [PMID: 37504441 PMCID: PMC10378863 DOI: 10.3390/gels9070562] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/08/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023] Open
Abstract
The challenges of using VEGF to promote osteoblastic differentiation include a short half-life and a narrow therapeutic window. A carrier system combining hydrogel and liposomes may improve the therapeutic efficacy of VEGF for bone regeneration. This study aimed to investigate the effects of delivery of VEGF via liposomal hydrogel on the osteogenesis of MG-63 cells. Liposomal hydrogel scaffold was fabricated and then characterized in terms of the morphological and chemical properties using FESEM and FTIR. In 2.5D analysis, the MG-63 cells were cultured on liposomal hydrogel + VEGF as the test group. The osteogenic effects of VEGF were compared with the control groups, i.e., hydrogel without liposomes + VEGF, osteogenic medium (OM) supplemented with a bolus of VEGF, and OM without VEGF. Cell morphology, viability, and differentiation and mineralization potential were investigated using FESEM, MTT assay, ALP activity, and Alizarin red staining. The characterization of scaffold showed no significant differences in the morphological and chemical properties between hydrogel with and without liposomes (p > 0.05). The final 2.5D culture demonstrated that cell proliferation, differentiation, and mineralization were significantly enhanced in the liposomal hydrogel + VEGF group compared with the control groups (p < 0.05). In conclusion, liposomal hydrogel can be used to deliver VEGF in a sustained manner in order to enhance the osteogenesis of MG-63 cells.
Collapse
Affiliation(s)
- Milton Hongli Tsai
- Discipline of Orthodontics, Department of Family Oral Health, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Rohaya Megat Abdul Wahab
- Discipline of Orthodontics, Department of Family Oral Health, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Shahrul Hisham Zainal Ariffin
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Fazren Azmi
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Farinawati Yazid
- Discipline of Pediatric Dentistry, Department of Family Oral Health, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
8
|
Zhang Y, Chen X, Yang X, Huang L, Qiu X. Mesenchymal Stem Cell-Derived from Dental Tissues-Related lncRNAs: A New Regulator in Osteogenic Differentiation. J Tissue Eng Regen Med 2023; 2023:4622584. [PMID: 40226409 PMCID: PMC11919082 DOI: 10.1155/2023/4622584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/12/2023] [Accepted: 06/22/2023] [Indexed: 04/15/2025]
Abstract
Odontogenic stem cells are mesenchymal stem cells (MSCs) with multipotential differentiation potential from different dental tissues. Their osteogenic differentiation is of great significance in bone tissue engineering. In recent years, it has been found that long noncoding RNAs (lncRNAs) participate in regulating the osteoblastic differentiation of stem cells at the epigenetic level, transcriptional level, and posttranscriptional level. We reviewed the existing lncRNA related to the osteogenic differentiation of odontogenic stem cells and emphasized the critical mechanism of lncRNA in the osteogenic differentiation of odontogenic stem cells. These findings are expected to be an important target for promoting osteoblastic differentiation of odontogenic stem cells in bone regeneration therapy with lncRNA.
Collapse
Affiliation(s)
- Yinchun Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - Xuan Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - XiaoXia Yang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - Lei Huang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - Xiaoling Qiu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| |
Collapse
|
9
|
Bian Y, Hu T, Lv Z, Xu Y, Wang Y, Wang H, Zhu W, Feng B, Liang R, Tan C, Weng X. Bone tissue engineering for treating osteonecrosis of the femoral head. EXPLORATION (BEIJING, CHINA) 2023; 3:20210105. [PMID: 37324030 PMCID: PMC10190954 DOI: 10.1002/exp.20210105] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/12/2022] [Indexed: 06/16/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating and complicated disease with an unclear etiology. Femoral head-preserving surgeries have been devoted to delaying and hindering the collapse of the femoral head since their introduction in the last century. However, the isolated femoral head-preserving surgeries cannot prevent the natural progression of ONFH, and the combination of autogenous or allogeneic bone grafting often leads to many undesired complications. To tackle this dilemma, bone tissue engineering has been widely developed to compensate for the deficiencies of these surgeries. During the last decades, great progress has been made in ingenious bone tissue engineering for ONFH treatment. Herein, we comprehensively summarize the state-of-the-art progress made in bone tissue engineering for ONFH treatment. The definition, classification, etiology, diagnosis, and current treatments of ONFH are first described. Then, the recent progress in the development of various bone-repairing biomaterials, including bioceramics, natural polymers, synthetic polymers, and metals, for treating ONFH is presented. Thereafter, regenerative therapies for ONFH treatment are also discussed. Finally, we give some personal insights on the current challenges of these therapeutic strategies in the clinic and the future development of bone tissue engineering for ONFH treatment.
Collapse
Affiliation(s)
- Yixin Bian
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Tingting Hu
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Zehui Lv
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yiming Xu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yingjie Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Han Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Wei Zhu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Bin Feng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Chaoliang Tan
- Department of ChemistryCity University of Hong KongKowloonHong Kong SARChina
| | - Xisheng Weng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
10
|
Yıldız S, Nursal AF, Yıgıt S, Tumer MK. Role of VEGF I/D variant in suspectibility to odontogenic cyst formation. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:308-316. [PMID: 36270022 DOI: 10.1080/15257770.2022.2136693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Odontogenic cysts, are located in the jawbones, filled with fluid surrounded by epithelial lining and fibrous connective tissue. Vascular endothelial growth factor (VEGF) can induce physiological and pathological angiogenesis and is an endothelial cell-specific mitogen. The aim of the present study was to investigate whether any possible association between the VEGF insertion/deletion (I/D) variant and odontogenic cyst in Turkish population. Clinical information and venous blood samples were collected from 62 odontogenic cyst patients and 98 healthy controls. DNA was isolated from peripheral blood leukocytes. Genotyping of the VEGF I/D variant was done by the polymerase chain reaction (PCR) method. There was a statistically differece in terms of VEGF I/D allele frequencies between patients and controls. VEGF I/D variant I allele frequency was more prevalant in patients compared to controls (p = 0.006411, OR: 2.08, 95%Cl: 1.322-3.272). A statistically significant association was observed when the patients were compared with the controls according to D/D + I/D versus I/I genotype (p = 0.0508, OR: 1.925, 95%Cl: 0.872-4.246). The genotype distribution of VEGF I/D was not statistically different between patients and controls (p > 0.05). For the first time, our results provided evidence supporting the odontogenic cyst formation associated with the I/D variant at the promoter region of the VEGF gene in a group of Turkish population. Although it was seen in our study that the I/D variant in the promoter region of the VEGF gene supports odontogenic cyst formation, large-scale studies are needed to elucidate the effect of this variant on odontogenic cysts.
Collapse
Affiliation(s)
- Serkan Yıldız
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Istanbul, Turkey
| | - Ayse Feyda Nursal
- Department of Medical Genetics, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Serbulent Yıgıt
- Department of Veterinary Genetics, Faculty of Veterinary, Ondokuz Mayıs University, Samsun, Turkey
| | - Mehmet Kemal Tumer
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Alanya Alaaddin Keykubat University, Alanya, Turkey
| |
Collapse
|
11
|
Diri D, Alasaad H, Muhammed H, Ibrahim J. Case report: adipose-derived mesenchymal stem cells combined with core decompression in the treatment of early-stage avascular necrosis of the femoral head. Int J Surg Case Rep 2022; 102:107861. [PMID: 36603496 PMCID: PMC9826853 DOI: 10.1016/j.ijscr.2022.107861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/25/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Core decompression is a well-known modality for treating the early stages of avascular necrosis of the femoral head (AVN), however, several methods have been suggested to augment this procedure and improve the outcomes. CASE REPORT A 52 male was diagnosed with a stage I AVN of the femoral head and treated with core decompression (CD) and injection of adipose-derived mesenchymal stem cells (AD-MSCs). The MRI showed full healing of the lesion after 3 months with significant clinical and functional improvement. DISCUSSION AD-MSCs could have the same capabilities as bone marrow-derived stem cells with many advantages, implantation of AD-MSCs in orthopedics and as an augmentation of core decompression has been tried before, but no clear guidelines nor methods of application are well established in the literature. CONCLUSION Implantation of AD-MSCs with Core decompression could be an effective modality to treat osteonecrosis of the femoral head in pre-collapse stages, however, we need bigger clinical studies to determine the actual effectiveness of this method.
Collapse
|
12
|
Mahapatra C, Kumar P, Paul MK, Kumar A. Angiogenic stimulation strategies in bone tissue regeneration. Tissue Cell 2022; 79:101908. [DOI: 10.1016/j.tice.2022.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
|
13
|
Sadeghifar A, Sheibani M, Joukar S, Dabiri S, Alavi S, Azari O, Vosoghi D, Zeynali Y, Zeynali Y, Shahraki M, Torghabe A, Rostamzadeh F, Nasri A. The Effect of Waterpipe Tobacco Smoking on Bone Healing Following Femoral Fractures in Male Rats. Front Surg 2021; 8:722446. [PMID: 34671637 PMCID: PMC8520932 DOI: 10.3389/fsurg.2021.722446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Given the increasing use of waterpipe tobacco smoking in the world and its unknown effects on bone healing, this study investigated the repairing of femoral bone fractures in rats exposed to waterpipe tobacco smoking (WTS). Main Methods: This study involved 40 male Wistar rats that were divided into two groups, including the femoral fracture (Fx) and the Fx + WTS groups. Each group was divided into two subgroups that were evaluated for bone healing 28 and 42 days after femoral fracture. After fixing the fractured femur, the healing process was evaluated by radiography, pathological indicators, and a measurement of the blood levels of vascular endothelial growth factor (VEGF), parathyroid hormone (PTH), Ca ++, transforming growth factor-beta (TGF-β), and insulin-like growth factor 1 (IGF-1). Additionally, the density of VEGF and CD34 in fracture tissue was investigated by immunohistochemistry. Key Findings: Radiographic findings showed that factors related to the earlier stages of bone healing had higher scores in the Fx + WTS28 and 42 subgroups in comparison to the Fx groups. The density of VEGF and CD34 showed that the angiogenesis processes were different in the bone fracture area and callus tissue in the Fx +WTS subgroups. The serum levels of VEGF, TGF-β, and IGF-1 were significantly lower in the Fx +WTS42 group, and PTH in the Fx +WTS28 group was higher than that in the other groups. Significance: The findings showed the disturbance and delay in the femoral fracture union in rats exposed to hookah smoke. This is partly due to the reduction of molecular stimuli of bone synthesis and the attenuation of quantitative angiogenesis.
Collapse
Affiliation(s)
- Amirreza Sadeghifar
- Orthopedic Department, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohamad Sheibani
- Orthopedic Department, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Siyavash Joukar
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Pathology Department and Stem Cell Research Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Samanehsadat Alavi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Omid Azari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Darioush Vosoghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Yas Zeynali
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasman Zeynali
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohamad Shahraki
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Amirhesam Torghabe
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Farzaneh Rostamzadeh
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Nasri
- Pathology Department and Stem Cell Research Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Le Q, Madhu V, Hart JM, Farber CR, Zunder ER, Dighe AS, Cui Q. Current evidence on potential of adipose derived stem cells to enhance bone regeneration and future projection. World J Stem Cells 2021; 13:1248-1277. [PMID: 34630861 PMCID: PMC8474721 DOI: 10.4252/wjsc.v13.i9.1248] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/22/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Injuries to the postnatal skeleton are naturally repaired through successive steps involving specific cell types in a process collectively termed “bone regeneration”. Although complex, bone regeneration occurs through a series of well-orchestrated stages wherein endogenous bone stem cells play a central role. In most situations, bone regeneration is successful; however, there are instances when it fails and creates non-healing injuries or fracture nonunion requiring surgical or therapeutic interventions. Transplantation of adult or mesenchymal stem cells (MSCs) defined by the International Society for Cell and Gene Therapy (ISCT) as CD105+CD90+CD73+CD45-CD34-CD14orCD11b-CD79αorCD19-HLA-DR- is being investigated as an attractive therapy for bone regeneration throughout the world. MSCs isolated from adipose tissue, adipose-derived stem cells (ADSCs), are gaining increasing attention since this is the most abundant source of adult stem cells and the isolation process for ADSCs is straightforward. Currently, there is not a single Food and Drug Administration (FDA) approved ADSCs product for bone regeneration. Although the safety of ADSCs is established from their usage in numerous clinical trials, the bone-forming potential of ADSCs and MSCs, in general, is highly controversial. Growing evidence suggests that the ISCT defined phenotype may not represent bona fide osteoprogenitors. Transplantation of both ADSCs and the CD105- sub-population of ADSCs has been reported to induce bone regeneration. Most notably, cells expressing other markers such as CD146, AlphaV, CD200, PDPN, CD164, CXCR4, and PDGFRα have been shown to represent osteogenic sub-population within ADSCs. Amongst other strategies to improve the bone-forming ability of ADSCs, modulation of VEGF, TGF-β1 and BMP signaling pathways of ADSCs has shown promising results. The U.S. FDA reveals that 73% of Investigational New Drug applications for stem cell-based products rely on CD105 expression as the “positive” marker for adult stem cells. A concerted effort involving the scientific community, clinicians, industries, and regulatory bodies to redefine ADSCs using powerful selection markers and strategies to modulate signaling pathways of ADSCs will speed up the therapeutic use of ADSCs for bone regeneration.
Collapse
Affiliation(s)
- Quang Le
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Vedavathi Madhu
- Orthopaedic Surgery Research, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Joseph M Hart
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, United States
- Departments of Public Health Sciences and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, United States
| | - Eli R Zunder
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, United States
| | - Abhijit S Dighe
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Quanjun Cui
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| |
Collapse
|
15
|
Burn-induced heterotopic ossification from incidence to therapy: key signaling pathways underlying ectopic bone formation. Cell Mol Biol Lett 2021; 26:34. [PMID: 34315404 PMCID: PMC8313878 DOI: 10.1186/s11658-021-00277-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/20/2021] [Indexed: 01/02/2023] Open
Abstract
Burn injury is one of the potential causes of heterotopic ossification (HO), which is a rare but debilitating condition. The incidence ranges from 3.5 to 5.6 depending on body area. Burns that cover a larger percentage of the total body surface area (TBSA), require skin graft surgeries, or necessitate pulmonary intensive care are well-researched risk factors for HO. Since burns initiate such complex pathophysiological processes with a variety of molecular signal changes, it is essential to focus on HO in the specific context of burn injury to define best practices for its treatment. There are numerous key players in the pathways of burn-induced HO, including neutrophils, monocytes, transforming growth factor-β1-expressing macrophages and the adaptive immune system. The increased inflammation associated with burn injuries is also associated with pathway activation. Neurological and calcium-related contributions are also known. Endothelial-to-mesenchymal transition (EMT) and vascularization are known to play key roles in burn-induced HO, with hypoxia-inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) as potential initiators. Currently, non-steroidal anti-inflammatory drugs (NSAIDs) and radiotherapy are effective prophylaxes for HO. Limited joint motion, ankylosis and intolerable pain caused by burn-induced HO can be effectively tackled via surgery. Effective biomarkers for monitoring burn-induced HO occurrence and bio-prophylactic and bio-therapeutic strategies should be actively developed in the future.
Collapse
|
16
|
Yang F, Ji Z, Peng L, Fu T, Liu K, Dou W, Li J, Li Y, Long Y, Zhang W. Efficacy, safety and complications of autologous fat grafting to the eyelids and periorbital area: A systematic review and meta-analysis. PLoS One 2021; 16:e0248505. [PMID: 33793573 PMCID: PMC8016360 DOI: 10.1371/journal.pone.0248505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
Background In recent years, autologous fat grafting (AFG), also known as fat transfer or lipofilling, has been widely performed for periorbital rejuvenation and defect correction, although the evidence regarding its efficacy and safety is still lacking. Besides, with respect to the periorbital region, it is invariably the earliest appearance area of the facial aging phenomenon. Therefore, a systematic review and meta-analysis is needed to evaluate the efficacy and safety of this technique. Methods A literature search was performed in PubMed, Embase, and the Cochrane library databases on November 20, 2020, adhering to the PRISMA guidelines, to identify all relevant articles. Then, a data extraction and standardization process was performed to assess all outcome data. Ultimately, the data were assessed using a random effects regression model with comprehensive meta-analysis software. Results Thirty-nine studies consisting of 3 cohorts and 36 case series with a total of 4046 cases were included. Meta-analysis revealed a relatively high satisfaction rate of 90.9% (95% CI, 86.4%–94.0%). Frequent complications in 4046 patients receiving AFG were edema, chemosis, and contour irregularity, with an overall complication rate of 7.9% (95% CI, 4.8%–12.8%). Conclusion This systematic review and meta-analysis showed that AFG for rejuvenation of eyelids and periorbital area provided a high satisfaction rate and did not result in severe complications. Therefore, AFG might be performed safely for periorbital rejuvenation and reconstruction.
Collapse
Affiliation(s)
- Fan Yang
- Department of Plastic Surgery and Burns, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhaohua Ji
- Department of Epidemiology, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Liwei Peng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Ting Fu
- Department of Epidemiology, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Kun Liu
- Department of Epidemiology, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, The Fourth Military Medical University, Xi’an, China
| | - Wenjie Dou
- Department of Plastic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jing Li
- Department of Plastic Surgery and Burns, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yuejun Li
- Department of Plastic Surgery and Burns, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- * E-mail: (WZ); (YL); (YL)
| | - Yong Long
- Department of Epidemiology, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, The Fourth Military Medical University, Xi’an, China
- * E-mail: (WZ); (YL); (YL)
| | - Weilu Zhang
- Department of Epidemiology, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, The Fourth Military Medical University, Xi’an, China
- * E-mail: (WZ); (YL); (YL)
| |
Collapse
|
17
|
Extracellular Vesicle-Derived microRNAs of Human Wharton's Jelly Mesenchymal Stromal Cells May Activate Endogenous VEGF-A to Promote Angiogenesis. Int J Mol Sci 2021; 22:ijms22042045. [PMID: 33669517 PMCID: PMC7922033 DOI: 10.3390/ijms22042045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
Despite low levels of vascular endothelial growth factor (VEGF)-A, the secretome of human Wharton’s jelly (WJ) mesenchymal stromal cells (MSCs) effectively promoted proangiogenic responses in vitro, which were impaired upon the depletion of small (~140 nm) extracellular vesicles (EVs). The isolated EVs shared the low VEGF-A profile of the secretome and expressed five microRNAs, which were upregulated compared to fetal dermal MSC-derived EVs. These upregulated microRNAs exclusively targeted the VEGF-A gene within 54 Gene Ontology (GO) biological processes, 18 of which are associated with angiogenesis. Moreover, 15 microRNAs of WJ-MSC-derived EVs were highly expressed (Ct value ≤ 26) and exclusively targeted the thrombospondin 1 (THBS1) gene within 75 GO biological processes, 30 of which are associated with the regulation of tissue repair. The relationship between predicted microRNA target genes and WJ-MSC-derived EVs was shown by treating human umbilical-vein endothelial cells (HUVECs) with appropriate doses of EVs. The exposure of HUVECs to EVs for 72 h significantly enhanced the release of VEGF-A and THBS1 protein expression compared to untreated control cells. Finally, WJ-MSC-derived EVs stimulated in vitro tube formation along with the migration and proliferation of HUVECs. Our findings can contribute to a better understanding of the molecular mechanisms underlying the proangiogenic responses induced by human umbilical cord-derived MSCs, suggesting a key regulatory role for microRNAs delivered by EVs.
Collapse
|
18
|
Jang CH, Kim W, Kim G. Effects of fibrous collagen/CDHA/hUCS biocomposites on bone tissue regeneration. Int J Biol Macromol 2021; 176:479-489. [PMID: 33571590 DOI: 10.1016/j.ijbiomac.2021.02.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 12/17/2022]
Abstract
Collagen- and bioceramic-based composites have been widely used in hard tissue engineering because they are analogous to the organic/inorganic constituents of native bones. However, biocomposites based on collagen and bioceramics show low mechanical stiffness and limited osteogenic activities. To elevate the low biophysical and biological activities, we have introduced a new biocomposite structure. Herein, we propose a biocomposite mimicking not only the physical structure of the extracellular matrix (ECM) structure but also the biochemical components of native bone tissues. Several components including fibrillated collagen, calcium-deficient hydroxyapatite (CDHA) obtained from α-tricalcium phosphate hydrolysis, and human umbilical cord serum (hUCS) were used to generate a unique structure of the biocomposite. The 3D-printed composites were topographically similar to the nanofibrous ECM and exhibited a mechanically stable structure. We also evaluated the in vitro biocompatibilities of the biocomposite using human adipose stem cells and found that the collagen/hUCS/CDHA scaffold accelerated the in vitro osteogenic differentiation of human adipose-derived stem cells and in vivo osteogenesis in a mastoid obliterated rat model.
Collapse
Affiliation(s)
- Chul Ho Jang
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - WonJin Kim
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon 16419, South Korea
| | - GeunHyung Kim
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon 16419, South Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
19
|
Kim HD, Park J, Amirthalingam S, Jayakumar R, Hwang NS. Bioinspired inorganic nanoparticles and vascular factor microenvironment directed neo-bone formation. Biomater Sci 2021; 8:2627-2637. [PMID: 32242197 DOI: 10.1039/d0bm00041h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Various strategies have been explored to stimulate new bone formation. These strategies include using angiogenic stimulants in combination with inorganic biomaterials. Neovascularization during the neo-bone formation provides nutrients along with bone-forming minerals. Therefore, it is crucial to design a bone stimulating microenvironment composed of both pro-angiogenic and osteogenic factors. In this respect, human vascular endothelial growth factor (hVEGF) has been shown to promote blood vessel formation and bone formation. Furthermore, in recent years, whitlockite (WH), a novel phase of magnesium-containing calcium phosphate derivatives that exist in our bone tissue, has been synthesized and applied in bone tissue engineering. In this study, our aim is to explore the potential use of hVEGF and WH for bone tissue engineering. Our study demonstrated that hVEGF and a WH microenvironment synergistically stimulated osteogenic commitment of mesenchymal stem cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Hwan D Kim
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea.
| | - Jungha Park
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea.
| | - Sivashanmugam Amirthalingam
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - R Jayakumar
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea. and Interdisciplinary Program in Bioengineering, Seoul National University, 151-742, Seoul, Republic of Korea and The BioMax/N-Bio Institute of Seoul National University, Seoul, 151-742, Republic of Korea
| |
Collapse
|
20
|
Dreyer CH, Kjaergaard K, Ding M, Qin L. Vascular endothelial growth factor for in vivo bone formation: A systematic review. J Orthop Translat 2020; 24:46-57. [PMID: 32642428 PMCID: PMC7334443 DOI: 10.1016/j.jot.2020.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To achieve optimal bone formation one of the most influential parameters has been mentioned to be adequate blood supply. Vascular endothelial growth factor (VEGF) is hereby of particular interest in bone regeneration, because of its primary ability to induce neovascularization and chemokine affection for endothelial cells (EC), and is considered to be the main regulator of vascular formation. However, the growth factor has yet to be implemented in a clinical setting in orthopaedic intervention surgery. We hypothesised that the development of VEGF in vivo for bone formation in the last decade had progressed towards clinical application since the latest systematic review from 2008. OBJECTIVE This systematic review recapped the last 13 years of in vivo bone regeneration using vascular endothelial growth factor (VEGF). METHOD A total of 1374 articles were identified using the PubMed search string (vegf or "vascular endothelial growth factor") and (osteogen∗ or "bone formation" or "bone regeneration"). By 3 selection phases 24 published articles were included by the criteria of being in vivo, using only VEGF for bone formation, published after 2007 and written in English. Articles in vitro, written in different languages than English and older than 2007 was excluded. The most recent systematic review on this subject was published in 2008, with the latest included study from 01 to 11-2007. All included studies were classified based on animal, type of defect, scaffold, control group, type of VEGF, release rate, dosage of VEGF, time of evaluation and results. Each study was evaluated for risk of bias by modified CAMARADES quality assessment for the use in experimental animal studies. The score was calculated by peer review journal publication, use of control group, randomisation of groups, justified VEGF dosage, blinding of results, details on animal model, sample size calculation, comply with ethics and no conflict of interest. RESULTS No clinical trials or human application studies were obtained from our search. Experimentally, 11 articles using solely VEGF for bone formation had a group or a timepoint significantly better than the corresponding control group. 18 articles revealed no significant difference of VEGF compared to the control group and 1 article reported a significant decreased bone growth using VEGF compared to control. CONCLUSION Based on these results no clinical studies have yet been performed. However, indications in the best use of VEGF from experimental studies could be made towards that the optimal release is within the first three weeks, in defect models, with the best effect before eight weeks. Future designs should incorporate this with standardised and reproducible models for verification towards clinical practice. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This systematic review aims to assess the existing literature to focus on methodologies and outcomes that can provide future knowledge regarding the solitary use of VEGF for bone regeneration in a clinical setting.
Collapse
Affiliation(s)
- Chris H. Dreyer
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
- Musculoskeletal Research Laboratory, Department of Orthopaedic Surgery & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
- Acute Medicine, Department of Emergency Medicine, Slagelse Hospital, Slagelse, Denmark
| | - Kristian Kjaergaard
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
| | - Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedic Surgery & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
21
|
Combined Delivery of Two Different Bioactive Factors Incorporated in Hydroxyapatite Microcarrier for Bone Regeneration. Tissue Eng Regen Med 2020; 17:607-624. [PMID: 32803541 DOI: 10.1007/s13770-020-00257-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/03/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The delivery of growth factors using a carrier system presents a promising and innovative tool in tissue engineering and dentistry today. Two of the foremost bioactive factors, bone morphogenetic protein-2 and vascular endothelial growth factor (VEGF), are widely applied using a ceramic scaffold. The aim of this study was to determine the use of hydroxyapatite microcarrier (MC) for dual delivery of osteogenic and angiogenic factors to accelerate hard tissue regeneration during the regenerative process. METHODS Two MCs of different sizes were fabricated by emulsification of gelatin and alpha-tricalcium phosphate (α-TCP). The experimental group was divided based on the combination of MC size and growth factors. For investigating the in vitro properties, rat mesenchymal stem cells (rMSCs) were harvested from bone marrow of the femur and tibia. For in vivo experiments, MC with/without growth factors was applied into the standardized, 5-mm diameter defects, which were made bilaterally on the parietal bone of the rat. The animals were allowed to heal for 8 weeks, and samples were harvested and analyzed by micro-computed tomography and histology. RESULTS Improved proliferation of rat mesenchymal stem cells was observed with VEGF loaded MC. For osteogenic differentiation, dual growth factors delivered by MC showed higher osteogenic gene expression, alkaline phosphatse production and calcium deposition. The in vivo results revealed statistically significant increase in new bone formation when dual growth factors were delivered by MC. Dual growth factors administered on a calcium phosphate matrix showed significantly enhanced osteogenic potential. CONCLUSION We propose this system has potential clinical utility in providing solutions for craniofacial bone defects, with the added benefit of early availability.
Collapse
|
22
|
Zakrzewski W, Dobrzynski M, Rybak Z, Szymonowicz M, Wiglusz RJ. Selected Nanomaterials' Application Enhanced with the Use of Stem Cells in Acceleration of Alveolar Bone Regeneration during Augmentation Process. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1216. [PMID: 32580409 PMCID: PMC7353104 DOI: 10.3390/nano10061216] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 01/15/2023]
Abstract
Regenerative properties are different in every human tissue. Nowadays, with the increasing popularity of dental implants, bone regenerative procedures called augmentations are sometimes crucial in order to perform a successful dental procedure. Tissue engineering allows for controlled growth of alveolar and periodontal tissues, with use of scaffolds, cells, and signalling molecules. By modulating the patient's tissues, it can positively influence poor integration and healing, resulting in repeated implant surgeries. Application of nanomaterials and stem cells in tissue regeneration is a newly developing field, with great potential for maxillofacial bony defects. Nanostructured scaffolds provide a closer structural support with natural bone, while stem cells allow bony tissue regeneration in places when a certain volume of bone is crucial to perform a successful implantation. Several types of selected nanomaterials and stem cells were discussed in this study. Their use has a high impact on the efficacy of the current and future procedures, which are still challenging for medicine. There are many factors that can influence the regenerative process, while its general complexity makes the whole process even harder to control. The aim of this study was to evaluate the effectiveness and advantage of both stem cells and nanomaterials in order to better understand their function in regeneration of bone tissue in oral cavity.
Collapse
Affiliation(s)
- Wojciech Zakrzewski
- Department of Experimental Surgery and Biomaterial Research, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland; (W.Z.); (Z.R.); (M.S.)
| | - Maciej Dobrzynski
- Department of Conservative Dentistry and Pedodontics, Wroclaw Medical University, Krakowska 26, 50-425 Wroclaw, Poland;
| | - Zbigniew Rybak
- Department of Experimental Surgery and Biomaterial Research, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland; (W.Z.); (Z.R.); (M.S.)
| | - Maria Szymonowicz
- Department of Experimental Surgery and Biomaterial Research, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland; (W.Z.); (Z.R.); (M.S.)
| | - Rafal J. Wiglusz
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, 50-422 Wroclaw, Poland
| |
Collapse
|
23
|
Yu T, You X, Zhou H, He W, Li Z, Li B, Xia J, Zhu H, Zhao Y, Yu G, Xiong Y, Yang Y. MiR-16-5p regulates postmenopausal osteoporosis by directly targeting VEGFA. Aging (Albany NY) 2020; 12:9500-9514. [PMID: 32427128 PMCID: PMC7288956 DOI: 10.18632/aging.103223] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/31/2020] [Indexed: 05/13/2023]
Abstract
In this study, we used bioinformatics tools, and experiments with patient tissues and human mesenchymal stem cells (hMSCs) to identify differentially regulated genes (DEGs) and microRNAs (miRNAs) that promote postmenopausal osteoporosis. By analyzing the GSE56815 dataset from the NCBI GEO database, we identified 638 DEGs, including 371 upregulated and 267 downregulated genes, in postmenopausal women with low bone density. Enrichment and protein-protein interaction network analyses showed that TP53, RPS27A, and VEGFA were the top three hub genes with the highest degree of betweenness and closeness centrality. TargetScanHuman and DIANA software analyses and dual luciferase reporter assays confirmed that miR-16a-5p directly targets the 3'UTR of VEGFA. Postmenopausal patients with osteoporosis showed higher miR-16-5p and lower VEGFA levels than those without osteoporosis (n=10 each). VEGFA levels were higher in miR-16-5p knockdown hMSCs and were reduced in miR-16-5p-overexpressing hMSCs. mRNA expression of osteogenic markers, ALP, OCN, and RUNX2, as well as calcium deposition based on Alizarin red staining, correlated inversely with miR-16-5p levels and correlated positively with VEGFA levels. These findings suggest that miR-16-5p suppresses osteogenesis by inhibiting VEGFA expression and is a promising target for postmenopausal osteoporosis therapy.
Collapse
Affiliation(s)
- Tao Yu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Xiaomeng You
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haichao Zhou
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Wenbao He
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Zihua Li
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Bing Li
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jiang Xia
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Hui Zhu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Youguang Zhao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Guangrong Yu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yunfeng Yang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
24
|
Forghani A, Koduru SV, Chen C, Leberfinger AN, Ravnic DJ, Hayes DJ. Differentiation of Adipose Tissue-Derived CD34+/CD31- Cells into Endothelial Cells In Vitro. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020; 6:101-110. [PMID: 33344757 PMCID: PMC7747864 DOI: 10.1007/s40883-019-00093-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 02/07/2019] [Indexed: 12/18/2022]
Abstract
In this study, CD34+/CD31- progenitor cells were isolated from the stromal vascular fraction (SVF) of adipose tissue using magnetic activated cell sorting. The endothelial differentiation capability of these cells in vitro was evaluated by culturing them in vascular endothelial growth factor (VEGF) induced medium for 14 days. Viability, proliferation, differentiation and tube formation of these cells were evaluated. Cell viability study revealed that both undifferentiated and endothelial differentiated cells remained healthy for 14 days. However, the proliferation rate was higher in undifferentiated cells compared to endothelial differentiated ones. Upregulation of endothelial characteristic genes (Von Willebrand Factor (vWF) and VE Cadherin) was observed in 2D culture. However, PECAM (CD31) was only found to be upregulated after the cells had formed tube-like structures in 3D Matrigel culture. These results indicate that adipose derived CD34+/CD31- cells when cultured in VEGF induced medium, are capable differentiation into endothelial-like lineages. Tube formation of the cells started 3h after seeding the cells on Matrigel and formed more stable and connected network 24 h post seeding in presence of VEGF.
Collapse
Affiliation(s)
- Anoosha Forghani
- Department of Biomedical Engineering, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Srinivas V Koduru
- Department of Surgery, College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Cong Chen
- Department of Biomedical Engineering, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ashley N Leberfinger
- Department of Surgery, College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Dino J Ravnic
- Department of Surgery, College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
- Materials Research Institute, Materials Characterization Lab, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institute of the Life Sciences, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
25
|
Kuterbekov M, Jonas AM, Glinel K, Picart C. Osteogenic Differentiation of Adipose-Derived Stromal Cells: From Bench to Clinics. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:461-474. [PMID: 32098603 DOI: 10.1089/ten.teb.2019.0225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In addition to mesenchymal stem cells, adipose-derived stem/stromal cells (ASCs) are an attractive source for a large variety of cell-based therapies. One of their most important potential applications is related to the regeneration of bone tissue thanks to their capacity to differentiate in bone cells. However, this requires a proper control of their osteogenic differentiation, which depends not only on the initial characteristics of harvested cells but also on the conditions used for their culture. In this review, we first briefly describe the preclinical and clinical trials using ASCs for bone regeneration and present the quantitative parameters used to characterize the osteogenic differentiation of ASCs. We then focus on the soluble factors influencing the osteogenic differentiation of ACS, including the steroid hormones and various growth factors, notably the most osteoinductive ones, the bone morphogenetic proteins (BMPs). Impact statement Adipose-derived stromal/stem cells are reviewed for their use in bone regeneration.
Collapse
Affiliation(s)
- Mirasbek Kuterbekov
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium.,Grenoble Institute of Technology, University Grenoble Alpes, LMGP, Grenoble, France
| | - Alain M Jonas
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Karine Glinel
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Catherine Picart
- Grenoble Institute of Technology, University Grenoble Alpes, LMGP, Grenoble, France.,Biomimetism and Regenerative Medicine Lab, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Université Grenoble-Alpes/CEA/CNRS, Grenoble, France
| |
Collapse
|
26
|
Wang N, Liu X, Shi L, Liu Y, Guo S, Liu W, Li X, Meng J, Ma X, Guo Z. Identification of a prolonged action molecular GLP-1R agonist for the treatment of femoral defects. Biomater Sci 2020; 8:1604-1614. [PMID: 31967113 DOI: 10.1039/c9bm01426h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Poly-GLP-1 promotes angiogenesis to accelerate bone formationviaBMSC differentiation and M2 polarization.
Collapse
|
27
|
Zhou Y, Liu C, He J, Dong L, Zhu H, Zhang B, Feng X, Weng W, Cheng K, Yu M, Wang H. KLF2 + stemness maintains human mesenchymal stem cells in bone regeneration. Stem Cells 2019; 38:395-409. [PMID: 31721356 DOI: 10.1002/stem.3120] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 09/01/2019] [Indexed: 01/04/2023]
Abstract
Mesenchymal stem cells (MSCs), which are undifferentiated stem cells with the property of stemness and the potential to differentiate into multiple lineages, including osteoblasts, have attracted a great deal of attention in bone tissue engineering. Consistent with the heterogeneity of MSCs, various surface markers have been used. However, it is still unclear which markers of MSCs are best for cell amplification in vitro and later bone regeneration in vivo. Krüppel-like Factor 2 (KLF2) is an important indicator of the stemness of human MSCs (hMSCs) and as early vascularization is also critical for bone regeneration, we used KLF2 as a novel in vitro marker for MSCs and investigated the angiogenesis and osteogenesis between KLF2+ MSCs and endothelial cells (ECs). We found a synergistic interaction between hMSCs and human umbilical vein ECs (HUVECs) in that KLF2+ stemness-maintained hMSCs initially promoted the angiogenesis of HUVECs, which in turn more efficiently stimulated the osteogenesis of hMSCs. In fact, KLF2+ hMSCs secreted angiogenic factors initially, with some of the cells then differentiating into pericytes through the PDGF-BB/PDGFR-β signaling pathway, which improved blood vessel formation. The matured HUVECs in turn synergistically enhanced the osteogenesis of KLF2+ hMSCs through upregulated vascular endothelial growth factor. A three-dimensional coculture model using cell-laden gelatin methacrylate (GelMA) hydrogel further confirmed these results. This study provides insight into the stemness-directed synergistic interaction between hMSCs and HUVECs, and our results will have a profound impact on further strategies involving the application of KLF2+ hMSC/HUVEC-laden GelMA hydrogel in vascular network bioengineering and bone regeneration.
Collapse
Affiliation(s)
- Ying Zhou
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Chao Liu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jianxiang He
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Lingqing Dong
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- State Key Laboratory of Silicon Materials, Cyrus Tang Center for Sensor Materials and Applications, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Huiyong Zhu
- The First Affiliated Hospital of Medical College, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Bin Zhang
- The State Key Laboratory of Fluid Power Transmission and Control, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoxia Feng
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Wenjian Weng
- State Key Laboratory of Silicon Materials, Cyrus Tang Center for Sensor Materials and Applications, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Kui Cheng
- State Key Laboratory of Silicon Materials, Cyrus Tang Center for Sensor Materials and Applications, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- The First Affiliated Hospital of Medical College, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Huiming Wang
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- The First Affiliated Hospital of Medical College, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
28
|
Wagner JM, Schmidt SV, Dadras M, Huber J, Wallner C, Dittfeld S, Becerikli M, Jaurich H, Reinkemeier F, Drysch M, Lehnhardt M, Behr B. Inflammatory processes and elevated osteoclast activity chaperon atrophic non-union establishment in a murine model. J Transl Med 2019; 17:416. [PMID: 31831031 PMCID: PMC6909450 DOI: 10.1186/s12967-019-02171-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/05/2019] [Indexed: 01/09/2023] Open
Abstract
Background Delayed bone healing, especially in long bones poses one of the biggest problems in orthopeadic and reconstructive surgery and causes tremendous costs every year. There is a need for exploring the causes in order to find an adequate therapy. Earlier investigations of human scaphoid non-union revealed an elevated osteoclast activity, accompanied by upregulated levels of TGF-beta and RANKL. Interestingly, scaphoid non-union seemed to be well vascularized. Methods In the current study, we used a murine femur-defect model to study atrophic non unions over a time-course of 10 weeks. Different time points were chosen, to gather insights into the dynamic processes of non-union establishment. Results Histological analyses as well as western blots and qRT-PCR indicated enhanced osteoclast activity throughout the observation period, paralleled by elevated levels of TGF-beta, TNF-alpha, MMP9, MMP13 and RANKL, especially during the early phases of non-union establishment. Interestingly, elevated levels of these mediators decreased markedly over a period of 10 weeks, as inflammatory reaction during non-union establishment seemed to wear out. To our surprise, osteoblastogenesis seemed to be unaffected during early stages of non-union establishment. Conclusion Taken together, we gained first insights into the establishment process of atrophic non unions, in which inflammatory processes accompanied by highly elevated osteoclast activity seem to play a leading role.
Collapse
Affiliation(s)
- Johannes M Wagner
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany.
| | - Sonja V Schmidt
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Mehran Dadras
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Julika Huber
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Christoph Wallner
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Stephanie Dittfeld
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Mustafa Becerikli
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Henriette Jaurich
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Felix Reinkemeier
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Marius Drysch
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Marcus Lehnhardt
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Björn Behr
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| |
Collapse
|
29
|
Nadine S, Patrício SG, Correia CR, Mano JF. Dynamic microfactories co-encapsulating osteoblastic and adipose-derived stromal cells for the biofabrication of bone units. Biofabrication 2019; 12:015005. [DOI: 10.1088/1758-5090/ab3e16] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
30
|
Bone allografts combined with adipose-derived stem cells in an optimized cell/volume ratio showed enhanced osteogenesis and angiogenesis in a murine femur defect model. J Mol Med (Berl) 2019; 97:1439-1450. [PMID: 31367858 DOI: 10.1007/s00109-019-01822-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 07/02/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Critical sized defects, especially in long bones, pose one of the biggest problems in orthopedic surgery. By definition, these defects do not heal without further treatment. Different therapeutic options range from autologous bone grafts, for example, free vascularized bone grafts, to commercially available bone allografts. Disadvantages of these bone allografts are related to reduced osteogenesis, since they are solely composed of cell-free bone matrix. The purpose of this study was to investigate the cell seeding efficiency of human adipose-derived stem cells (hASCs) on human bone allografts in vitro and furthermore analyze these optimized seeded allografts in a critical sized defect model in vivo. Cancellous human bone allografts were colonized with human ASCs in vitro. Cell seeding efficiency was evaluated by Cell Counting Kit-8 assay. Thereafter, optimized hASC-seeded bone scaffolds were examined in a murine femur defect model, stabilized with the MouseExFix system. Subsequently, x-ray analysis and histology were performed. Examination of cell seeding efficiency revealed an optimum starting population of 84,600 cells per 100 mm3 scaffold. In addition, scaffolds seeded with hASCs showed increased osteogenesis compared with controls. Histological analysis revealed increased remodeling and elevated new bone formation within hASC-seeded scaffolds. Moreover, immunohistochemical stainings revealed increased proliferation, osteogenesis, and angiogenesis. In this study, we systemically optimized cell/volume ratio of two promising components of tissue engineering: hASCs and human bone allografts. These findings may serve as a basis for future translational studies. KEY MESSAGES: Bone tissue engineering. Mesenchymal stem cells derived from human adipose tissue (hASCs). Optimal cell/volume ratio of cell-seeded scaffolds. Increased osteogenesis and angiogenesis in vivo.
Collapse
|
31
|
Ma X, Fan C, Wang Y, Du Y, Zhu Y, Liu H, Lv L, Liu Y, Zhou Y. MiR-137 knockdown promotes the osteogenic differentiation of human adipose-derived stem cells via the LSD1/BMP2/SMAD4 signaling network. J Cell Physiol 2019; 235:909-919. [PMID: 31241766 DOI: 10.1002/jcp.29006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
MicroRNAs are a group of endogenous regulators that participate in several cellular physiological processes. However, the role of miR-137 in the osteogenic differentiation of human adipose-derived stem cells (hASCs) has not been reported. This study verified a general downward trend in miR-137 expression during the osteogenic differentiation of hASCs. MiR-137 knockdown promoted the osteogenesis of hASCs in vitro and in vivo. Mechanistically, inhibition of miR-137 activated the bone morphogenetic protein 2 (BMP2)-mothers against the decapentaplegic homolog 4 (SMAD4) pathway, whereas repressed lysine-specific histone demethylase 1 (LSD1), which was confirmed as a negative regulator of osteogenesis in our previous studies. Furthermore, LSD1 knockdown enhanced the expression of BMP2 and SMAD4, suggesting the coordination of LSD1 in the osteogenic regulation of miR-137. This study indicated that miR-137 negatively regulated the osteogenic differentiation of hASCs via the LSD1/BMP2/SMAD4 signaling network, revealing a new potential therapeutic target of hASC-based bone tissue engineering.
Collapse
Affiliation(s)
- Xiaohan Ma
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Cong Fan
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuejun Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yangge Du
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
32
|
Wagner JM, Reinkemeier F, Wallner C, Dadras M, Huber J, Schmidt SV, Drysch M, Dittfeld S, Jaurich H, Becerikli M, Becker K, Rauch N, Duhan V, Lehnhardt M, Behr B. Adipose-Derived Stromal Cells Are Capable of Restoring Bone Regeneration After Post-Traumatic Osteomyelitis and Modulate B-Cell Response. Stem Cells Transl Med 2019; 8:1084-1091. [PMID: 31179644 PMCID: PMC6766598 DOI: 10.1002/sctm.18-0266] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/10/2019] [Indexed: 12/17/2022] Open
Abstract
Bone infections are a frequent cause for large bony defects with a reduced healing capacity. In previous findings, we could already show diminished healing capacity after bone infections, despite the absence of the causing agent, Staphylococcus aureus. Moreover, these bony defects showed reduced osteoblastogenesis and increased osteoclastogenesis, meaning elevated bone resorption ongoing with an elevated B‐cell activity. To overcome the negative effects of this postinfectious inflammatory state, we tried to use the regenerative capacity of mesenchymal stem cells derived from adipose tissue (adipose‐derived stem cells [ASCs]) to improve bone regeneration and moreover were curious about immunomodulation of applicated stem cells in this setting. Therefore, we used our established murine animal model and applicated ASCs locally after sufficient debridement of infected bones. Bone regeneration and resorption as well as immunological markers were investigated via histology, immunohistochemistry, Western blot, and fluorescence‐activated cell scanning (FACS) analysis and μ‐computed tomography (CT) analysis. Interestingly, ASCs were able to restore bone healing via elevation of osteoblastogenesis and downregulation of osteoclasts. Surprisingly, stem cells showed an impact on the innate immune system, downregulating B‐cell population. In summary, these data provide a fascinating new and innovative approach, supporting bone healing after bacterial infections and moreover gain insights into the complex ceremony of stem cell interaction in terms of bone infection and regeneration. stem cells translational medicine2019;8:1084–1091
Collapse
Affiliation(s)
| | - Felix Reinkemeier
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Christoph Wallner
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Mehran Dadras
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Julika Huber
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Sonja Verena Schmidt
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Marius Drysch
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Stephanie Dittfeld
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Henriette Jaurich
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Mustafa Becerikli
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Kathrin Becker
- Poliklinik für Kieferorthopädie, University Hospital Düsseldorf, Poliklinik für Kieferorthopädie, Düsseldorf, Germany
| | - Nicole Rauch
- Poliklinik für Kieferorthopädie, University Hospital Düsseldorf, Poliklinik für Kieferorthopädie, Düsseldorf, Germany
| | - Vikas Duhan
- Institute of Immunology, University Hospital Essen, Essen, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, University Hospital BG Bergmannsheil Bochum, Bochum, Germany
| |
Collapse
|
33
|
Kim I, Lee SS, Kim SHL, Bae S, Lee H, Hwang NS. Osteogenic Effects of VEGF-Overexpressed Human Adipose-Derived Stem Cells with Whitlockite Reinforced Cryogel for Bone Regeneration. Macromol Biosci 2019; 19:e1800460. [PMID: 30821921 DOI: 10.1002/mabi.201800460] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Bone is a vascularized tissue that is comprised of collagen fibers and calcium phosphate crystals such as hydroxyapatite (HAp) and whitlockite (WH). HAp and WH are known to elicit bone regeneration by stimulating osteoblast activities and osteogenic commitment of stem cells. In addition, vascular endothelial growth factor (VEGF) is shown to promote osteogenesis and angiogenesis which is considered as an essential process in bone repair by providing nutrients. In this study, VEGF-secreting human adipose-derived stem cells (VEGF-ADSCs) are developed by transducing ADSCs with VEGF-encoded lentivirus. Additionally, WH-reinforced gelatin/heparin cryogels (WH-C) are fabricated by loading WH into gelatin/heparin cryogels. VEGF-ADSC secrete tenfold more VEGF than ADSC and show increased VEGF secretion with cell growth. Also, incorporation of WH into cryogels provides a mineralized environment with ions secreted from WH. When the VEGF-ADSCs are seeded on WH-C, sustained release of VEGF is observed due to the specific affinity of VEGF to heparin. Finally, the synergistic effect of VEGF-ADSC and WH on osteogenesis is successfully confirmed by alkaline phosphatase and real-time polymerase chain reaction analysis. In vivo bone formation is demonstrated via implantation of VEGF-ADSC seeded WH-C into mouse calvarial bone defect model, resulted in enhanced bone development with the highest bone volume/total volume.
Collapse
Affiliation(s)
- Inseon Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seunghun S Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hyun L Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sunghoon Bae
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoyon Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.,Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.,BioMax Institute of Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
34
|
Wallner C, Huber J, Drysch M, Schmidt SV, Wagner JM, Dadras M, Dittfeld S, Becerikli M, Jaurich H, Lehnhardt M, Behr B. Activin Receptor 2 Antagonization Impairs Adipogenic and Enhances Osteogenic Differentiation in Mouse Adipose-Derived Stem Cells and Mouse Bone Marrow-Derived Stem Cells In Vitro and In Vivo. Stem Cells Dev 2019; 28:384-397. [PMID: 30654712 DOI: 10.1089/scd.2018.0155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tumors, traumata, burn injuries or surgeries can lead to critical-sized bony defects which need to be reconstructed. Mesenchymal stem cells (MSCs) have the ability to differentiate into multiple cell lineages and thus present a promising alternative for use in tissue engineering and reconstruction. However, there is an ongoing debate whether all MSCs are equivalent in their differentiation and proliferation ability. The goal of this study was to assess osteogenic and adipogenic characteristic changes of adipose-derived stem cells (ASCs) and bone marrow-derived stem cells (BMSCs) upon Myostatin inhibition with Follistatin in vitro and in vivo. We harvested ASCs from mice inguinal fat pads and BMSCs from tibiae of mice. By means of histology, real-time cell analysis, immunohistochemistry, and PCR osteogenic and adipogenic proliferation and differentiation in the presence or absence of Follistatin were analyzed. In vivo, osteogenic capacity was investigated in a tibial defect model of wild-type (WT) mice treated with mASCs and mBMSCs of Myo-/- and WT origin. In vitro, we were able to show that inhibition of Myostatin leads to markedly reduced proliferative capacity in mBMSCs and mASCs in adipogenic differentiation and reduced proliferation in osteogenic differentiation in mASCs, whereas proliferation in mBMSCs in osteogenic differentiation was increased. Adipogenic differentiation was inhibited in mASCs and mBMSCs upon Follistatin treatment, whereas osteogenic differentiation was increased in both cell lineages. In vivo, we could demonstrate increased osteoid formation in WT mice treated with mASCs and mBMSCs of Myo-/- origin and enhanced osteogenic differentiation and proliferation of mASCs of Myo-/- origin. We could demonstrate that the osteogenic potential of mASCs could be raised to a level comparable to mBMSCs upon inhibition of Myostatin. Moreover, Follistatin treatment led to inhibition of adipogenesis in both lineages.
Collapse
Affiliation(s)
- Christoph Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Julika Huber
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Marius Drysch
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Sonja Verena Schmidt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Johannes Maximilian Wagner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Mehran Dadras
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Stephanie Dittfeld
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Mustafa Becerikli
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Henriette Jaurich
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
35
|
Huang P, Gu J, Wu J, Geng L, Hong Y, Wang S, Wang M. Microarray analysis of the molecular mechanisms associated with age and body mass index in human meniscal injury. Mol Med Rep 2018; 19:93-102. [PMID: 30483788 PMCID: PMC6297773 DOI: 10.3892/mmr.2018.9685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to identify genes and functional pathways associated with meniscal injuries affected by age or body mass index (BMI) using microarray analysis. The GSE45233 gene expression dataset with 12 injured meniscus samples associated with age and BMI and GSE66635 dataset with 12 injured and 12 normal meniscus samples were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified based on age or BMI in GSE45233. DEGs between injured and normal meniscus samples in GSE66635 were also identified. Common DEGs between GSE45233 and GSE66635 were identified as feature genes associated with age or BMI, followed by protein-protein interaction (PPI) network and functional pathway enrichment analyses for the feature genes. Finally, the GSE51588 genome-wide expression profile was then downloaded from the GEO database to validate the results. A total of 1,328 DEGs were identified. Of these, 28 age-associated and 20 BMI-associated meniscal injury genes were obtained. B-cell lymphoma-2 (Bcl-2) and matrix metalloproteinase-14 were identified as hub genes in the PPI networks. Functional pathway enrichment analysis revealed that vascular endothelial growth factor A (VEGFA), transferrin (TF) and Bcl-2 were involved in the hypoxia-inducible factor 1 signaling pathway. TF was involved in the mineral absorption function pathway associated with BMI. Additionally, TF and VEGFA were identified to be overlapping candidate genes of GSE45233 and GSE66635, and DEGs in GSE51588. Therefore, VEGFA, TF, and Bcl-2 may be important genes for human meniscal injuries. Additional evaluations of these results are required.
Collapse
Affiliation(s)
- Peiyan Huang
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Jun Gu
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Junguo Wu
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Lei Geng
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Yang Hong
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Siqun Wang
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Minghai Wang
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
36
|
Angiogenic and Osteogenic Synergy of Human Mesenchymal Stem Cells and Human Umbilical Vein Endothelial Cells Cocultured on a Nanomatrix. Sci Rep 2018; 8:15749. [PMID: 30356078 PMCID: PMC6200728 DOI: 10.1038/s41598-018-34033-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 10/08/2018] [Indexed: 11/12/2022] Open
Abstract
To date, bone tissue regeneration strategies lack an approach that effectively provides an osteogenic and angiogenic environment conducive to bone growth. In the current study, we evaluated the osteogenic and angiogenic response of human mesenchymal stem cells (hMSCs) and green fluorescent protein-expressing human umbilical vein endothelial cells (GFP-HUVECs) cocultured on a self-assembled, peptide amphiphile nanomatrix functionalized with the cell adhesive ligand RGDS (PA-RGDS). Analysis of alkaline phosphatase activity, von Kossa staining, Alizarin Red quantification, and osteogenic gene expression, indicates a significant synergistic effect between the PA-RGDS nanomatrix and coculture that promoted hMSC osteogenesis. In addition, coculturing on PA-RGDS resulted in enhanced HUVEC network formation and upregulated vascular endothelial growth factor gene and protein expression. Though PA-RGDS and coculturing hMSCs with HUVECs were each previously reported to individually enhance hMSC osteogenesis, this study is the first to demonstrate a synergistic promotion of HUVEC angiogenesis and hMSC osteogenesis by integrating coculturing with the PA-RGDS nanomatrix. We believe that using the combination of hMSC/HUVEC coculture and PA-RGDS substrate is an efficient method for promoting osteogenesis and angiogenesis, which has immense potential as an efficacious, engineered platform for bone tissue regeneration.
Collapse
|
37
|
Quarto N, Shailendra S, Meyer NP, Menon S, Renda A, Longaker MT. Twist1-Haploinsufficiency Selectively Enhances the Osteoskeletal Capacity of Mesoderm-Derived Parietal Bone Through Downregulation of Fgf23. Front Physiol 2018; 9:1426. [PMID: 30374308 PMCID: PMC6196243 DOI: 10.3389/fphys.2018.01426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/19/2018] [Indexed: 02/05/2023] Open
Abstract
Craniofacial development is a program exquisitely orchestrated by tissue contributions and regulation of genes expression. The basic helix–loop–helix (bHLH) transcription factor Twist1 expressed in the skeletal mesenchyme is a key regulator of craniofacial development playing an important role during osteoskeletogenesis. This study investigates the postnatal impact of Twist1 haploinsufficiency on the osteoskeletal ability and regeneration on two calvarial bones arising from tissues of different embryonic origin: the neural crest-derived frontal and the mesoderm-derived parietal bones. We show that Twist1 haplonsufficiency as well Twist1-sh-mediated silencing selectively enhanced osteogenic and tissue regeneration ability of mesoderm-derived bones. Transcriptomic profiling, gain-and loss-of-function experiments revealed that Twist1 haplonsufficiency triggers its selective activity on mesoderm-derived bone through a sharp downregulation of the bone-derived hormone Fgf23 that is upregulated exclusively in wild-type parietal bone.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States.,Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Siny Shailendra
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Nathaniel P Meyer
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| |
Collapse
|
38
|
Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC, Deng WP. Revisiting the Advances in Isolation, Characterization and Secretome of Adipose-Derived Stromal/Stem Cells. Int J Mol Sci 2018; 19:ijms19082200. [PMID: 30060511 PMCID: PMC6121360 DOI: 10.3390/ijms19082200] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) seems to be a promising regenerative therapeutic agent due to the minimally invasive approach of their harvest and multi-lineage differentiation potential. The harvested adipose tissues are further digested to extract stromal vascular fraction (SVF), which is cultured, and the anchorage-dependent cells are isolated in order to characterize their stemness, surface markers, and multi-differentiation potential. The differentiation potential of ASCs is directed through manipulating culture medium composition with an introduction of growth factors to obtain the desired cell type. ASCs have been widely studied for its regenerative therapeutic solution to neurologic, skin, wound, muscle, bone, and other disorders. These therapeutic outcomes of ASCs are achieved possibly via autocrine and paracrine effects of their secretome comprising of cytokines, extracellular proteins and RNAs. Therefore, secretome-derivatives might offer huge advantages over cells through their synthesis and storage for long-term use. When considering the therapeutic significance and future prospects of ASCs, this review summarizes the recent developments made in harvesting, isolation, and characterization. Furthermore, this article also provides a deeper insight into secretome of ASCs mediating regenerative efficacy.
Collapse
Affiliation(s)
- Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Viraj Krishna Mishra
- Applied Biotech Engineering Centre (ABEC), Department of Biotechnology, Ambala College of Engineering and Applied Research, Ambala 133101, India.
| | - Rajni Dubey
- Graduate Institute Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Yue-Hua Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Life Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Feng-Chou Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Win-Ping Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Basic medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan.
| |
Collapse
|
39
|
Wang YH, Wu JY, Kong SC, Chiang MH, Ho ML, Yeh ML, Chen CH. Low power laser irradiation and human adipose-derived stem cell treatments promote bone regeneration in critical-sized calvarial defects in rats. PLoS One 2018; 13:e0195337. [PMID: 29621288 PMCID: PMC5886537 DOI: 10.1371/journal.pone.0195337] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/20/2018] [Indexed: 01/18/2023] Open
Abstract
Both stem cell therapy and physical treatments have been shown to be beneficial in accelerating bone healing. However, the efficacy of combined treatment with stem cells and physical stimuli for large bone defects remains uncertain. The aim of this study was to evaluate the bone regeneration effects of low-power laser irradiation (LPLI) and human adipose-derived stem cell (ADSC) treatments during fracture repair using a comparative rat calvarial defect model. We evaluated the viability of human ADSCs, which were cultured on a porous PLGA scaffold using an MTS assay. The critical-sized calvarial bone defect rats were divided into 4 groups: control group, LPLI group, ADSC group, and ADSC+LPLI group. Bone formation was evaluated using micro-CT. New bone formation areas and osteogenic factor expression levels were then examined by histomorphological analysis and immunohistochemical staining. Our data showed that PLGA had no cytotoxic effect on human ADSCs. Micro-CT analyses revealed that both the LPLI and ADSC groups showed improved calvarial bone defect healing compared to the control group. In addition, the ADSC+LPLI group showed significantly increased bone volume at 16 weeks after surgery. The area of new bone formation ranked as follows: control group < LPLI group < ADSC group < ADSC+LPLI group. There were significant differences between the groups. In addition, both ADSC and ADSC+LPLI groups showed strong signals of vWF expression. ADSC and LPLI treatments improved fracture repair in critical-sized calvarial defects in rats. Importantly, the combined treatment of ADSCs and LPLI further enhances the bone healing process.
Collapse
Affiliation(s)
- Yan-Hsiung Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jyun-Yi Wu
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Fresenius Kabi Taiwan Ltd, Taipei, Taiwan
| | - Su Chii Kong
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Min-Hsuan Chiang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ming-Long Yeh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hsin Chen
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
40
|
Sun Z, Nair LS, Laurencin CT. The Paracrine Effect of Adipose-Derived Stem Cells Inhibits IL-1β-induced Inflammation in Chondrogenic Cells through the Wnt/β-Catenin Signaling Pathway. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0047-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Sharma S, Sapkota D, Xue Y, Rajthala S, Yassin MA, Finne-Wistrand A, Mustafa K. Delivery of VEGFA in bone marrow stromal cells seeded in copolymer scaffold enhances angiogenesis, but is inadequate for osteogenesis as compared with the dual delivery of VEGFA and BMP2 in a subcutaneous mouse model. Stem Cell Res Ther 2018; 9:23. [PMID: 29386057 PMCID: PMC5793460 DOI: 10.1186/s13287-018-0778-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background In bone tissue engineering (BTE), extensive research into vascular endothelial growth factor A (VEGFA)-mediated angiogenesis has yielded inconsistent results. The aim of this study was to investigate the influence on angio- and osteogenesis of adenoviral-mediated delivery of VEGFA alone or in combination with bone morphogenetic protein 2 (BMP2) in bone marrow stromal cells (BMSC) seeded onto a recently developed poly(LLA-co-CL) scaffold. Methods Human BMSC were engineered to express VEGFA alone or in combination with BMP2 and seeded onto poly(LLA-co-CL) scaffolds. Changes in angiogenic and osteogenic gene and protein levels were examined by quantitative reverse-transcription polymerase chain reaction (RT-PCR), PCR array, and alkaline phosphatase assay. An in vivo subcutaneous mouse model was used to investigate the effect on angio- and osteogenesis of VEGFA alone or in combination with BMP2, using microcomputed tomography (μCT), histology, immunohistochemistry, and immunofluorescence. Results Combined delivery of a lower ratio (1:3) of VEGFA and BMP2 (ad-BMP2 + VEGFA) led to upregulation of osteogenic and angiogenic genes in vitro at 3 and 14 days, compared with mono-delivery of VEGFA (ad-VEGFA) and other controls. In vivo, in a subcutaneous mouse model, both ad-VEGFA and ad-BMP2 + VEGFA scaffold explants exhibited increased angiogenesis at 2 weeks. Enhanced angiogenesis was largely related to the recruitment and differentiation of mouse progenitor cells to the endothelial lineage and, to a lesser extent, to endothelial differentiation of the implanted BMSC. μCT and histological analyses revealed enhanced de novo bone formation only in the ad-BMP2 + VEGFA group, corresponding at the molecular level to the upregulation of genes related to osteogenesis, such as ALPL, RUNX2, and SPP1. Conclusions Although BMSC expressing VEGFA alone or in combination with BMP2 significantly induced angiogenesis, VEGFA alone failed to demonstrate osteogenic activity both in vitro and in vivo. These results not only call into question the use of VEGFA alone in bone regeneration, but also highlight the importance in BTE of appropriately formulated combined delivery of VEGFA and BMP2. Electronic supplementary material The online version of this article (10.1186/s13287-018-0778-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sunita Sharma
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, 5020, Bergen, Norway.
| | - Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, 0316, Oslo, Norway
| | - Ying Xue
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, 5020, Bergen, Norway
| | - Saroj Rajthala
- The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Mohammed A Yassin
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, 10044, Stockholm, Sweden
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, 10044, Stockholm, Sweden
| | - Kamal Mustafa
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, 5020, Bergen, Norway.
| |
Collapse
|
42
|
Lin DPL, Dass CR. Transdifferentiation of adipocytes to osteoblasts: potential for orthopaedic treatment. ACTA ACUST UNITED AC 2018; 70:307-319. [PMID: 29365349 DOI: 10.1111/jphp.12862] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVES As both adipocytes and osteoblasts originate from the same pool of mesenchymal stem cells, increasing clinical evidence has emerged of the plasticity between the two lineages. For instance, the downregulation of osteoblast differentiation and upregulation of adipogenesis are common features of conditions such as multiple myeloma, obesity and drug-induced bone loss in diabetes mellitus. However, despite in-vitro and in-vivo observations of adipocyte transdifferentiation into osteoblasts, little is known of the underlying mechanisms. KEY FINDINGS This review summarises the current knowledge of this particular transdifferentiation process whereby the Wnt/β-catenin signalling pathway and Runx2 overexpression have been postulated to play a critical role. SUMMARY Furthermore, due to the possibility of a novel therapy in the treatment of bone conditions, a number of agents with the potential to induce adipo-to-osteoblast transdifferentiation have been investigated such as all-trans retinoic acid, bone morphogenetic protein-9 and vascular endothelial growth factor.
Collapse
Affiliation(s)
- Daphne P L Lin
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA, Australia.,Curtin Biosciences Research Precinct, Bentley, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA, Australia.,Curtin Biosciences Research Precinct, Bentley, Perth, WA, Australia
| |
Collapse
|
43
|
Murphy MP, Quarto N, Longaker MT, Wan DC. * Calvarial Defects: Cell-Based Reconstructive Strategies in the Murine Model. Tissue Eng Part C Methods 2017; 23:971-981. [PMID: 28825366 DOI: 10.1089/ten.tec.2017.0230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calvarial defects pose a continued clinical dilemma for reconstruction. Advancements within the fields of stem cell biology and tissue engineering have enabled researchers to develop reconstructive strategies using animal models. We review the utility of various animal models and focus on the mouse, which has aided investigators in understanding cranial development and calvarial bone healing. The murine model has also been used to study regenerative approaches to critical-sized calvarial defects, and we discuss the application of stem cells such as bone marrow-derived mesenchymal stromal cells, adipose-derived stromal cells, muscle-derived stem cells, and pluripotent stem cells to address deficient bone in this animal. Finally, we highlight strategies to manipulate stem cells using various growth factors and inhibitors and ultimately how these factors may prove crucial in future advancements within calvarial reconstruction using native skeletal stem cells.
Collapse
Affiliation(s)
- Matthew P Murphy
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Natalina Quarto
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| |
Collapse
|
44
|
Inhibition of GDF8 (Myostatin) accelerates bone regeneration in diabetes mellitus type 2. Sci Rep 2017; 7:9878. [PMID: 28852138 PMCID: PMC5575348 DOI: 10.1038/s41598-017-10404-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/08/2017] [Indexed: 12/21/2022] Open
Abstract
Metabolic diseases like diabetes mellitus cause bone healing deficiencies. We found significant impairment of bone regeneration, osteogenic differentiation and proliferation in diabetic bone. Moreover recent studies suggest a highly underestimated importance of GDF8 (Myostatin) in bone metabolism. Our goal was to analyze the role of GDF8 as a regulator of osteogenic differentiation, proliferation and bone regeneration. We used a murine tibial defect model in diabetic (Leprdb-/-) mice. Myostatin-Inhibitor Follistatin was administered in tibial bony defects of diabetic mice. By means of histology, immunohistochemistry and QRT-PC osteogenesis, differentiation and proliferation were analyzed. Application of Myostatin-inhibitor showed a significant improvement in diabetic bone regeneration compared to the control group (6.5 fold, p < 0.001). Immunohistochemistry revealed a significantly higher proliferation (7.7 fold, p = 0.009), osteogenic differentiation (Runx-2: 3.7 fold, p = 0.011, ALP: 9.3 fold, p < 0.001) and calcification (4.9 fold, p = 0.024) in Follistatin treated diabetic animals. Therapeutical application of Follistatin, known for the importance in muscle diseases, plays an important role in bone metabolism. Diabetic bone revealed an overexpression of the catabolic protein Myostatin. Antagonization of Myostatin in diabetic animals leads to a restoration of the impaired bone regeneration and represents a promising therapeutic option.
Collapse
|
45
|
Combined Effects of Vascular Endothelial Growth Factor and Bone Morphogenetic Protein 2 on Odonto/Osteogenic Differentiation of Human Dental Pulp Stem Cells In Vitro. J Endod 2017; 43:930-935. [PMID: 28457634 DOI: 10.1016/j.joen.2017.01.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/21/2017] [Accepted: 01/25/2017] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The purpose of this study was to investigate whether combined and concerted delivery of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2) enhances odonto/osteogenic differentiation of human dental pulp stem cells (DPSCs) in vitro. METHODS Various concentrations of VEGF and/or BMP-2 with or without the presence of odonto/osteogenic medium (OM) were added into DPSC cultures for 21 days. The mineral formation in cultures was evaluated using alizarin red stain (ARS). Optimal concentrations of VEGF and BMP-2 were codelivered to DPSCs for total of 21 days with the following experimental groups: (1) group 1: OM only, (2) group 2: OM + VEGF, (3) group 3: OM + BMP-2, and (4) group 4: OM + VEGF + BMP-2 (subgroup 4a: VEGF present the first 7 days, 4b: BMP-2 present the last 14 days, and 4c, both present for 21 days). Cultures were then subjected to quantitative ARS analysis or harvested for quantitative polymerase chain reaction analysis for the expression of core-binding factor alpha 1 (CBFA1), alkaline phosphatase (ALP), and dentin matrix protein 1 (DMP-1). RESULTS No mineral formation was detected by ARS when VEGF and/or BMP-2 were used without OM. OM + VEGF, but not OM + BMP-2, formed more mineralization than OM (P < .05). In the codelivery groups, the highest mineralization was observed in OM + VEGF and subgroup 4a compared with OM or the other groups (P < .05). Quantitative polymerase chain reaction analysis showed that CBFA1, ALP, and DMP-1 levels were higher in groups 2, 3, and 4a compared with 4b and 4c (P < .05). CBFA1 expressed higher in groups 2, 3, and 4a compared with OM (P < .05). For ALP expression, only subgroup 4a expressed higher than OM (P < .05). No difference was detected between groups 2 and 3 (P > .05) in the expression of the 3 genes. CONCLUSIONS VEGF addition in the early phase rather than a continuous presence of both VEGF and BMP-2 enhances odonto/osteogenic differentiation of DPSCs.
Collapse
|
46
|
Barba M, Di Taranto G, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther 2017; 17:677-689. [PMID: 28374644 DOI: 10.1080/14712598.2017.1315403] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell-based therapies exploit the heterogeneous and self-sufficient biological environment of stem cells to restore, maintain and improve tissue functions. Adipose-derived stem cells (ASCs) are, to this aim, promising cell types thanks to advantageous isolation procedures, growth kinetics, plasticity and trophic properties. Specifically, bone regeneration represents a suitable, though often challenging, target setting to test and apply ASC-based therapeutic strategies. Areas covered: ASCs are extremely plastic and secrete bioactive peptides that mediate paracrine functions, mediating their trophic actions in vivo. Numerous preclinical studies demonstrated that ASCs improve bone healing. Clinical trials are ongoing to validate the clinical feasibility of these approaches. This review is intended to define the state-of-the-art on ASCs, encompassing the biological features that make them suitable for bone regenerative strategies, and to provide an update on existing preclinical and clinical applications. Expert opinion: ASCs offer numerous advantages over other stem cells in terms of feasibility of clinical translation. Data obtained from in vivo experimentation are encouraging, and clinical trials are ongoing. More robust validations are thus expected to be achieved during the next few years, and will likely pave the way to optimized patient-tailored treatments for bone regeneration.
Collapse
Affiliation(s)
- Marta Barba
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Di Taranto
- b Department of Plastic, Reconstructive and Aesthetic Surgery , University of Rome "Sapienza" , Policlinico Umberto I, Rome , Italy
| | - Wanda Lattanzi
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
47
|
Yu W, Sun TW, Ding Z, Qi C, Zhao H, Chen F, Shi Z, Zhu YJ, Chen D, He Y. Copper-doped mesoporous hydroxyapatite microspheres synthesized by a microwave-hydrothermal method using creatine phosphate as an organic phosphorus source: application in drug delivery and enhanced bone regeneration. J Mater Chem B 2017; 5:1039-1052. [PMID: 32263882 DOI: 10.1039/c6tb02747d] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The development of multifunctional biomaterials with drug delivery ability, and pro-osteogenic and pro-angiogenic activities has garnered increasing interest in the field of regenerative medicine. In the present study, hypoxia-mimicking copper (Cu)-doped mesoporous hydroxyapatite (HAP) microspheres (Cu-MHMs) were successfully synthesized through a microwave-hydrothermal method by using creatine phosphate as an organic phosphorus source. The Cu-MHMs doped with 0.2, 0.5 and 1 mol% Cu were prepared. The Cu-MHMs consisting of HAP nanorods or nanosheets exhibited a hierarchically mesoporous hollow structure and a high specific surface area. Then the Cu-MHMs were investigated as a drug nanocarrier using doxorubicin hydrochloride (DOX) as a model drug. The Cu-MHMs showed a relatively high drug-loading capacity and a pH-responsive drug release behavior. Furthermore, the Cu-MHMs were incorporated into a chitosan (CS) matrix to construct a biomimetic scaffold optimized for bone regeneration. The Cu-MHM/CS composite scaffolds maintained high degrees of porosity and showed a sustained release of Cu ions. More importantly, the Cu-MHM/CS scaffolds not only enhanced the osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (rBMSCs) but also promoted the migration and tube formation of EA.hy926 cells. When implanted in rat critical-sized calvarial defects, the Cu-MHM/CS scaffolds significantly enhanced bone regeneration accompanied by more new blood vessel formation at 8 weeks post-operation compared with the MHM/CS scaffolds. These results suggest that the hypoxia-mimicking Cu-MHM/CS scaffolds could encourage bone regeneration by enhancing osteogenesis and angiogenesis simultaneously, which bodes well for the reconstruction of vascularized tissue-engineered bone.
Collapse
Affiliation(s)
- Weilin Yu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Comparison of Endothelial Differentiation Capacities of Human and Rat Adipose-Derived Stem Cells. Plast Reconstr Surg 2016; 138:1231-1241. [DOI: 10.1097/prs.0000000000002791] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
In vitroand long-term (2-year follow-up)in vivoosteogenic activities of human periosteum-derived osteoblasts seeded into growth factor-releasing polycaprolactone/pluronic F127 beads scaffolds. J Biomed Mater Res A 2016; 105:363-376. [DOI: 10.1002/jbm.a.35907] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/24/2016] [Accepted: 09/14/2016] [Indexed: 12/20/2022]
|
50
|
Amiri B, Ghollasi M, Shahrousvand M, Kamali M, Salimi A. Osteoblast differentiation of mesenchymal stem cells on modified PES-PEG electrospun fibrous composites loaded with Zn2SiO4 bioceramic nanoparticles. Differentiation 2016; 92:148-158. [DOI: 10.1016/j.diff.2016.08.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/28/2016] [Accepted: 08/03/2016] [Indexed: 12/25/2022]
|