1
|
Trapana J, Weinerman J, Lee D, Sedani A, Constantinescu D, Best TM, Hornicek FJ, Hare JM. Cell-based therapy in the treatment of musculoskeletal diseases. Stem Cells Transl Med 2024; 13:959-978. [PMID: 39226104 PMCID: PMC11465182 DOI: 10.1093/stcltm/szae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/21/2024] [Indexed: 09/04/2024] Open
Abstract
A limited number of tissues can spontaneously regenerate following injury, and even fewer can regenerate to a state comparable to mature, healthy adult tissue. Mesenchymal stem cells (MSCs) were first described in the 1960s-1970s by Friedenstein et al as a small population of bone marrow cells with osteogenic potential and abilities to differentiate into chondrocytes. In 1991, Arnold Caplan coined the term "mesenchymal cells" after identifying these cells as a theoretical precursor to bone, cartilage, tendon, ligament, marrow stroma, adipocyte, dermis, muscle, and connective tissues. MSCs are derived from periosteum, fat, and muscle. Another attractive property of MSCs is their immunoregulatory and regenerative properties, which result from crosstalk with their microenvironment and components of the innate immune system. Collectively, these properties make MSCs potentially attractive for various therapeutic purposes. MSCs offer potential in sports medicine, aiding in muscle recovery, meniscal tears, and tendon and ligament injuries. In joint disease, MSCs have the potential for chondrogenesis and reversing the effects of osteoarthritis. MSCs have also demonstrated potential application to the treatment of degenerative disc disease of the cervical, thoracic, and lumbar spine.
Collapse
Affiliation(s)
- Justin Trapana
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Jonathan Weinerman
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Danny Lee
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Anil Sedani
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - David Constantinescu
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Thomas M Best
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Francis J Hornicek
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| |
Collapse
|
2
|
Hejazi F, Zare F, Asgari M. Enhanced differentiation of mesenchymal stem cells in coral-incorporated PCL/elastin scaffold enables 3D defect healing in osteoporosis rat model. Int J Biol Macromol 2024; 277:134483. [PMID: 39102909 DOI: 10.1016/j.ijbiomac.2024.134483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/14/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
In osteoporosis, bone quality adversely affects the tissue structural competence which increases the risk of a complicated fracture healing. In the present study highly potent scaffold containing natural coral particles was designed and considered for the healing of critical size bone defect in osteoporosis rat model. Scaffold morphological evaluation confirmed the porous nanofibrous structure. Water uptake of about 900 % was obtained for the fabricated scaffold as the result of its composition and three-dimensional structure. Mechanical analysis revealed the compressive modulus of about 50 kPa for the fabricated coral-incorporated nanofibrous structure. In vitro cellular assessments revealed that the designed scaffold induces no toxicity and provides the proper substrate for cell attachment together with increased and prolonged cell proliferation. In vivo experiments demonstrated that implantation of the fabricated scaffold in the femoral defects of osteoporotic rats significantly increased the number of osteocytes and osteoblasts, and enhanced the BTV, and BMP-2 expression compared with the control group. Furthermore, it was observed that seeding the scaffolds with MSCs prior to implantation, resulted in substantial improvements in mRNA expression of the BMP-2 and VEGF genes and considerable enhancement in stereological findings such as significantly higher number of osteoblasts, osteocytes, TVB, and BTV.
Collapse
Affiliation(s)
- Fatemeh Hejazi
- Faculty of advanced technologies, Shiraz University, Shiraz, Iran.
| | - Fatemeh Zare
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Asgari
- Department of Anatomy, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
3
|
Kang I, Koo M, Jun JH, Lee J. Effect of nicotinamide mononucleotide on osteogenesis in MC3T3-E1 cells against inflammation-induced by lipopolysaccharide. Clin Exp Reprod Med 2024; 51:236-246. [PMID: 38599888 PMCID: PMC11372314 DOI: 10.5653/cerm.2023.06744] [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: 12/07/2023] [Accepted: 01/06/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVE Nicotinamide mononucleotide (NMN) is extensively utilized as an anti-aging agent and possesses anti-inflammatory properties. Lipopolysaccharide (LPS) activates Toll-like receptor 4, a process modulated by intracellular signaling pathways such as the Wnt/β-catenin pathway. This study investigated the impact of NMN on osteogenesis in the presence of LPS. METHODS To elucidate the role of NMN in osteogenesis in the context of Gram-negative bacterial infection after LPS treatment, we cultured a mouse pre-osteoblast cell line (MC3T3-E1) and subsequently incubated it with NMN and/or LPS. We then evaluated osteogenic activity by measuring alkaline phosphatase activity, assessing gene expression and protein levels, and performing Alizarin Red S staining and immunocytochemistry. RESULTS MC3T3-E1 cells underwent successful differentiation into osteoblasts following treatment with osteogenic induction medium. LPS diminished features related to osteogenic differentiation, which were subsequently partially reversed by treatment with NMN. The restorative effects of NMN on LPS-exposed MC3T3-E1 cells were further substantiated by elucidating the role of Wnt/β-catenin signaling, as confirmed through immunocytochemistry. CONCLUSION This study showed that infection with Gram-negative bacteria disrupted the osteogenic differentiation of MC3T3-E1 cells. This adverse effect was partially reversed by administering a high-dose of NMN. Drawing on these results, we propose that NMN could serve as a viable therapeutic strategy to preserve bone homeostasis in elderly and immunocompromised patients.
Collapse
Affiliation(s)
- Inyoung Kang
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| | - Myoungjoo Koo
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| | - Jin Hyun Jun
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| | - Jaewang Lee
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| |
Collapse
|
4
|
Wang X, He W, Huang H, Han J, Wang R, Li H, Long Y, Wang G, Han X. Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy. Biomolecules 2024; 14:858. [PMID: 39062572 PMCID: PMC11274544 DOI: 10.3390/biom14070858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoarthritis (OA), a chronic joint disease affecting over 500 million individuals globally, is characterized by the destruction of articular cartilage and joint inflammation. Conventional treatments are insufficient for repairing damaged joint tissue, necessitating novel therapeutic approaches. Mesenchymal stem cells (MSCs), with their potential for differentiation and self-renewal, hold great promise as a treatment for OA. However, challenges such as MSC viability and apoptosis in the ischemic joint environment hinder their therapeutic effectiveness. Hydrogels with biocompatibility and degradability offer a three-dimensional scaffold that support cell viability and differentiation, making them ideal for MSC delivery in OA treatment. This review discusses the pathological features of OA, the properties of MSCs, the challenges associated with MSC therapy, and methods for hydrogel preparation and functionalization. Furthermore, it highlights the advantages of hydrogel-based MSC delivery systems while providing insights into future research directions and the clinical potential of this approach.
Collapse
Affiliation(s)
- Xiangjiang Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Wentao He
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Hao Huang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Collage of Physics and Optoelectronics Engineering, Shenzhen University, Shenzhen 518060, China;
| | - Jiali Han
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Ruren Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Hongyi Li
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Ying Long
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Guiqing Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Xianjing Han
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| |
Collapse
|
5
|
Wang S, Jia Z, Dai M, Feng X, Tang C, Liu L, Cao L. Advances in natural and synthetic macromolecules with stem cells and extracellular vesicles for orthopedic disease treatment. Int J Biol Macromol 2024; 268:131874. [PMID: 38692547 DOI: 10.1016/j.ijbiomac.2024.131874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Serious orthopedic disorders resulting from myriad diseases and impairments continue to pose a considerable challenge to contemporary clinical care. Owing to its limited regenerative capacity, achieving complete bone tissue regeneration and complete functional restoration has proven challenging with existing treatments. By virtue of cellular regenerative and paracrine pathways, stem cells are extensively utilized in the restoration and regeneration of bone tissue; however, low survival and retention after transplantation severely limit their therapeutic effect. Meanwhile, biomolecule materials provide a delivery platform that improves stem cell survival, increases retention, and enhances therapeutic efficacy. In this review, we present the basic concepts of stem cells and extracellular vesicles from different sources, emphasizing the importance of using appropriate expansion methods and modification strategies. We then review different types of biomolecule materials, focusing on their design strategies. Moreover, we summarize several forms of biomaterial preparation and application strategies as well as current research on biomacromolecule materials loaded with stem cells and extracellular vesicles. Finally, we present the challenges currently impeding their clinical application for the treatment of orthopedic diseases. The article aims to provide researchers with new insights for subsequent investigations.
Collapse
Affiliation(s)
- Supeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China; Ningxia Medical University, Ningxia 750004, China
| | - Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xujun Feng
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China.
| |
Collapse
|
6
|
Wu W, Zhao Z, Wang Y, Zhu G, Tan K, Liu M, Li L. Biomechanical Effects of Mechanical Stress on Cells Involved in Fracture Healing. Orthop Surg 2024; 16:811-820. [PMID: 38439564 PMCID: PMC10984830 DOI: 10.1111/os.14026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Fracture healing is a complex staged repair process in which the mechanical environment plays a key role. Bone tissue is very sensitive to mechanical stress stimuli, and the literature suggests that appropriate stress can promote fracture healing by altering cellular function. However, fracture healing is a coupled process involving multiple cell types that balance and limit each other to ensure proper fracture healing. The main cells that function during different stages of fracture healing are different, and the types and molecular mechanisms of stress required are also different. Most previous studies have used a single mechanical stimulus on individual mechanosensitive cells, and there is no relatively uniform standard for the size and frequency of the mechanical stress. Analyzing the mechanisms underlying the effects of mechanical stimulation on the metabolic regulation of signaling pathways in cells such as in bone marrow mesenchymal stem cells (BMSCs), osteoblasts, chondrocytes, and osteoclasts is currently a challenging research hotspot. Grasping how stress affects the function of different cells at the molecular biology level can contribute to the refined management of fracture healing. Therefore, in this review, we summarize the relevant literature and describe the effects of mechanical stress on cells associated with fracture healing, and their possible signaling pathways, for the treatment of fractures and the further development of regenerative medicine.
Collapse
Affiliation(s)
- Weiyong Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Gengbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
7
|
Yun HM, Kim E, Kwon YJ, Park KR. Vanillin Promotes Osteoblast Differentiation, Mineral Apposition, and Antioxidant Effects in Pre-Osteoblasts. Pharmaceutics 2024; 16:485. [PMID: 38675146 PMCID: PMC11054936 DOI: 10.3390/pharmaceutics16040485] [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: 02/27/2024] [Revised: 03/19/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Antioxidant vanillin (4-hydroxy-3-methoxybenzaldehyde) is used as a flavoring in foods, beverages, and pharmaceuticals. Vanillin possesses various biological effects, such as antioxidant, anti-inflammatory, antibacterial, and anticancer properties. This study aimed to investigate the biological activities of vanillin purified from Adenophora triphylla var. japonica Hara on bone-forming processes. Vanillin treatment induced mineralization as a marker for mature osteoblasts, after stimulating alkaline phosphatase (ALP) staining and activity. The bone-forming processes of vanillin are mainly mediated by the upregulation of the bone morphogenetic protein 2 (BMP2), phospho-Smad1/5/8, and runt-related transcription factor 2 (RUNX2) pathway during the differentiation of osteogenic cells. Moreover, vanillin promoted osteoblast-mediated bone-forming phenotypes by inducing migration and F-actin polymerization. Furthermore, we validated that vanillin-mediated bone-forming processes were attenuated by noggin and DKK1. Finally, we demonstrated that vanillin-mediated antioxidant effects prevent the death of osteoblasts during bone-forming processes. Overall, vanillin has bone-forming properties through the BMP2-mediated biological mechanism, indicating it as a bone-protective compound for bone health and bone diseases such as periodontitis and osteoporosis.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eonmi Kim
- National Development Institute of Korean Medicine, Gyeongsan 38540, Republic of Korea; (E.K.); (Y.-J.K.)
| | - Yoon-Ju Kwon
- National Development Institute of Korean Medicine, Gyeongsan 38540, Republic of Korea; (E.K.); (Y.-J.K.)
| | - Kyung-Ran Park
- Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
8
|
Zhang X, Deng C, Qi S. Periosteum Containing Implicit Stem Cells: A Progressive Source of Inspiration for Bone Tissue Regeneration. Int J Mol Sci 2024; 25:2162. [PMID: 38396834 PMCID: PMC10889827 DOI: 10.3390/ijms25042162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The periosteum is known as the thin connective tissue covering most bone surfaces. Its extrusive bone regeneration capacity was confirmed from the very first century-old studies. Recently, pluripotent stem cells in the periosteum with unique physiological properties were unveiled. Existing in dynamic contexts and regulated by complex molecular networks, periosteal stem cells emerge as having strong capabilities of proliferation and multipotential differentiation. Through continuous exploration of studies, we are now starting to acquire more insight into the great potential of the periosteum in bone formation and repair in situ or ectopically. It is undeniable that the periosteum is developing further into a more promising strategy to be harnessed in bone tissue regeneration. Here, we summarized the development and structure of the periosteum, cell markers, and the biological features of periosteal stem cells. Then, we reviewed their pivotal role in bone repair and the underlying molecular regulation. The understanding of periosteum-related cellular and molecular content will help enhance future research efforts and application transformation of the periosteum.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| |
Collapse
|
9
|
Khaledi M, Zandi B, Mohsenipour Z. The Effect of Mesenchymal Stem Cells on the Wound Infection. Curr Stem Cell Res Ther 2024; 19:1084-1092. [PMID: 37815189 DOI: 10.2174/011574888x252482230926104342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/10/2023] [Accepted: 08/17/2023] [Indexed: 10/11/2023]
Abstract
Wound infection often requires a long period of care and an onerous treatment process. Also, the rich environment makes the wound an ideal niche for microbial growth. Stable structures, like biofilm, and drug-resistant strains cause a delay in the healing process, which has become one of the important challenges in wound treatment. Many studies have focused on alternative methods to deal the wound infections. One of the novel and highly potential ways is mesenchymal stromal cells (MSCs). MSCs are mesoderm-derived pluripotent adult stem cells with the capacity for self-renewal, multidirectional differentiation, and immunological control. Also, MSCs have anti-inflammatory and antiapoptotic effects. MScs, as pluripotent stromal cells, differentiate into many mature cells. Also, MSCs produce antimicrobial compounds, such as antimicrobial peptides (AMP), as well as secrete immune modulators, which are two basic features considered in wound healing. Despite the advantages, preserving the structure and activity of MSCs is considered one of the most important points in the treatment. MSCs' antimicrobial effects on microorganisms involved in wound infection have been confirmed in various studies. In this review, we aimed to discuss the antimicrobial and therapeutic applications of MSCs in the infected wound healing processes.
Collapse
Affiliation(s)
- Mansoor Khaledi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Bita Zandi
- Department of Microbiology, Faculty of advanced science and technology, Tehran medical science, Islamic Azad University, Tehran, Iran
| | - Zeinab Mohsenipour
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Li Y, Ding L, Zhang Y, Zhou B, Wai Man GC, Wang M, Li J, Liu Y, Lin W, Wang H, Lin S, Xu L. Histone demethylase Kdm5c regulates osteogenesis and bone formation via PI3K/Akt/HIF1α and Wnt/β-catenin signaling pathways. Genes Dis 2024; 11:64-66. [PMID: 37588198 PMCID: PMC10425781 DOI: 10.1016/j.gendis.2023.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 04/07/2023] Open
Affiliation(s)
- Ying Li
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Lingli Ding
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Yage Zhang
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Bingyu Zhou
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Gene Chi Wai Man
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Min Wang
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Jinglan Li
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Yamei Liu
- Departments of Diagnostics of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Weiping Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Haibin Wang
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
- Orthopaedic Research Lab, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, China
| |
Collapse
|
11
|
Diaz-Solano D, Sadri B, Peshkova M, Shpichka A, Smirnova O, Shams R, Timashev P, Vosough M. Advanced Therapeutic Medicinal Products in Bone and Cartilage Defects. Curr Rev Clin Exp Pharmacol 2024; 19:355-369. [PMID: 38275042 DOI: 10.2174/0127724328274436231207062008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 01/27/2024]
Abstract
The number of patients with functional loss of bone and cartilage tissue has shown an increasing trend. Insufficient or inappropriate conventional treatments applied for trauma, orthopedic diseases, or other bone and cartilage-related disorders can lead to bone and cartilage damage. This represents a worldwide public health issue and a significant economic burden. Advanced therapeutic medicinal products (ATMPs) proposed promising alternative therapeutic modalities by application of cell-based and tissue engineering approaches. Recently, several ATMPs have been developed to promote bone and cartilage tissue regeneration. Fifteen ATMPs, two related to bone and 13 related to cartilage, have received regulatory approval and marketing authorization. However, four ATMPs were withdrawn from the market for various reasons. However, ATMPs that are still on the market have demonstrated positive results, their broad application faced limitations. The development and standardization of methodologies will be a major challenge in the coming decades. Currently, the number of ATMPs in clinical trials using mesenchymal stromal cells or chondrocytes indicates a growing recognition that current ATMPs can be improved. Research on bone and cartilage tissue regeneration continues to expand. Cell-based therapies are likely to be clinically supported by the new ATMPs, innovative fabrication processes, and enhanced surgical approaches. In this study, we highlighted the available ATMPs that have been used in bone and cartilage defects and discussed their advantages and disadvantages in clinical applications.
Collapse
Affiliation(s)
- Dylana Diaz-Solano
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Venezuela
| | - Bahareh Sadri
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Smirnova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
12
|
Yun HM, Cho MH, Jeong H, Kim SH, Jeong YH, Park KR. Osteogenic Activities of Trifolirhizin as a Bioactive Compound for the Differentiation of Osteogenic Cells. Int J Mol Sci 2023; 24:17103. [PMID: 38069425 PMCID: PMC10706948 DOI: 10.3390/ijms242317103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
Plant extracts are widely used as traditional medicines. Sophora flavescens Aiton-derived natural compounds exert various beneficial effects, such as anti-inflammatory, anticancer, antioxidant, and antiregenerative activities, through their bioactive compounds, including flavonoids and alkaloids. In the present study, we investigated the biological effects of an S. flavescens-derived flavonoid, trifolirhizin (trifol), on the stimulation of osteogenic processes during osteoblast differentiation. Trifol (>98% purity) was successfully isolated from the root of S. flavescens and characterized. Trifol did not exhibit cellular toxicity in osteogenic cells, but promoted alkaline phosphatase (ALP) staining and activity, with enhanced expression of the osteoblast differentiation markers, including Alp, ColI, and Bsp. Trifol induced nuclear runt-related transcription factor 2 (RUNX2) expression during the differentiation of osteogenic cells, and concomitantly stimulated the major osteogenic signaling proteins, including GSK3β, β-catenin, and Smad1/5/8. Among the mitogen-activated protein kinases (MAPKs), Trifol activated JNK, but not ERK1/2 and p38. Trifol also increased the osteoblast-mediated bone-forming phenotypes, including transmigration, F-actin polymerization, and mineral apposition, during osteoblast differentiation. Overall, trifol exhibits bioactive activities related to osteogenic processes via differentiation, migration, and mineralization. Collectively, these results suggest that trifol may serve as an effective phytomedicine for bone diseases such as osteoporosis.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mi Hyeon Cho
- Korea Basic Science Institute (KBSI), Seoul 02841, Republic of Korea; (M.H.C.); (H.J.)
| | - Hoibin Jeong
- Korea Basic Science Institute (KBSI), Seoul 02841, Republic of Korea; (M.H.C.); (H.J.)
| | - Soo Hyun Kim
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (S.H.K.); (Y.H.J.)
| | - Yun Hee Jeong
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (S.H.K.); (Y.H.J.)
| | - Kyung-Ran Park
- Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
13
|
Mahajan A, Bhattacharyya S. Immunomodulation by mesenchymal stem cells during osteogenic differentiation: Clinical implications during bone regeneration. Mol Immunol 2023; 164:143-152. [PMID: 38011783 DOI: 10.1016/j.molimm.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
Critical bone defects resulting in delayed and non-union are a major concern in the field of orthopedics. Over the past decade, mesenchymal stem cells (MSCs) have become a promising frontier for bone repair and regeneration owing to their high expansion rate and osteogenic differentiation potential ex vivo. MSCs have also long been associated with their ability to modulate immune response in the recipients. These can even skew the immune response towards pro-inflammatory or anti-inflammatory type by sensing their local microenvironment. MSCs adopt anti-inflammatory phenotype at bone injury site and secrete various immunomodulatory factors such as IDO, NO, TGFβ1 and PGE-2 which have redundant role in osteoblast differentiation and bone formation. As such, several studies have also sought to decipher the immunomodulatory effects of osteogenically differentiated MSCs. The present review discusses the immunomodulatory status of MSCs during their osteogenic differentiation and summarizes few mechanisms that cause immunosuppression by osteogenically differentiated MSCs and its implication during bone healing.
Collapse
Affiliation(s)
- Aditi Mahajan
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
14
|
Shiroud Heidari B, Lopez EM, Chen P, Ruan R, Vahabli E, Davachi SM, Granero-Moltó F, De-Juan-Pardo EM, Zheng M, Doyle B. Silane-modified hydroxyapatite nanoparticles incorporated into polydioxanone/poly(lactide- co-caprolactone) creates a novel toughened nanocomposite with improved material properties and in vivo inflammatory responses. Mater Today Bio 2023; 22:100778. [PMID: 37664796 PMCID: PMC10474235 DOI: 10.1016/j.mtbio.2023.100778] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/13/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023] Open
Abstract
The interface tissue between bone and soft tissues, such as tendon and ligament (TL), is highly prone to injury. Although different biomaterials have been developed for TL regeneration, few address the challenges of the TL-bone interface. Here, we aim to develop novel hybrid nanocomposites based on poly(p-dioxanone) (PDO), poly(lactide-co-caprolactone) (LCL), and hydroxyapatite (HA) nanoparticles suitable for TL-bone interface repair. Nanocomposites, containing 3-10% of both unmodified and chemically modified hydroxyapatite (mHA) with a silane coupling agent. We then explored biocompatibility through in vitro and in vivo studies using a subcutaneous mouse model. Through different characterisation tests, we found that mHA increases tensile properties, creates rougher surfaces, and reduces crystallinity and hydrophilicity. Morphological observations indicate that mHA nanoparticles are attracted by PDO rather than LCL phase, resulting in a higher degradation rate for mHA group. We found that adding the 5% of nanoparticles gives a balance between the properties. In vitro experiments show that osteoblasts' activities are more affected by increasing the nanoparticle content compared with fibroblasts. Animal studies indicate that both HA and mHA nanoparticles (10%) can reduce the expression of pro-inflammatory cytokines after six weeks of implantation. In summary, this work highlights the potential of PDO/LCL/HA nanocomposites as an excellent biomaterial for TL-bone interface tissue engineering applications.
Collapse
Affiliation(s)
- Behzad Shiroud Heidari
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Emma Muinos Lopez
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Peilin Chen
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
- School of Medicine, Monash University, VIC, Melbourne, Australia
| | - Rui Ruan
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
| | - Ebrahim Vahabli
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
| | - Seyed Mohammad Davachi
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX, USA
| | - Froilán Granero-Moltó
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Elena M. De-Juan-Pardo
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
| | - Minghao Zheng
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Barry Doyle
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
15
|
Liu C, Lou Y, Sun Z, Ma H, Sun M, Li S, You D, Wu J, Ying B, Ding W, Yu M, Wang H. 4D Printing of Personalized-Tunable Biomimetic Periosteum with Anisotropic Microstructure for Accelerated Vascularization and Bone Healing. Adv Healthc Mater 2023; 12:e2202868. [PMID: 37171209 DOI: 10.1002/adhm.202202868] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/12/2023] [Indexed: 05/13/2023]
Abstract
An ideal biomimetic periosteum is expected to wrap various bone surfaces to orchestrate an optimal microenvironment for bone regeneration, including facilitating local vascularization, recruiting osteoblasts, and mineralizing the extracellular matrix (ECM). To mimic the role of the natural periosteum in promoting bone repair, a 4D printing technique to inlay aligned cell sheets on shape-shifting hydrogel is used, containing biophysical signals and spatially adjustable physical properties, for the first time. The outer hydrogel layer endows the biomimetic periosteum with the ability to digitally coordinate its 3D geometry to match the specific macroscopic bone shape to maintain a bone healing microenvironment. The inner aligned human mesenchymal stem cells (hMSCs) layer not only promotes the migration and angiogenesis of co-cultured cells but also exhibits excellent osteogenic differentiation properties. In vivo experiments show that apart from morphing preset shapes as physical barriers, the aligned biomimetic periosteum can actively facilitate local angiogenesis and early-stage osteogenesis. Altogether, this present work provides a novel route to construct a personalized biomimetic periosteum with anisotropic microstructure by introducing a tunable shape to maintain the bone reconstruction microenvironment and this strategy can be extended to repair sophisticated bone defects.
Collapse
Affiliation(s)
- Chao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Yiting Lou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Zheyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Haiying Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Shengjie Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
- Department of Stomatology, The First Affiliated Hospital of Ningbo University, 59 Liuting street, Ningbo, 315000, China
| | - Dongqi You
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Junjie Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Binbin Ying
- Department of Stomatology, The First Affiliated Hospital of Ningbo University, 59 Liuting street, Ningbo, 315000, China
| | - Wanghui Ding
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, 310000, China
| |
Collapse
|
16
|
Rinderknecht H, Mayer A, Histing T, Ehnert S, Nüssler A. Herbal Extracts of Ginseng and Maqui Berry Show Only Minimal Effects on an In Vitro Model of Early Fracture Repair of Smokers. Foods 2023; 12:2960. [PMID: 37569229 PMCID: PMC10419284 DOI: 10.3390/foods12152960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Smoking is a major risk factor for delayed fracture healing, affecting several aspects of early fracture repair, including inflammation, osteogenesis, and angiogenesis. Panax ginseng (GE) and maqui berry extract (MBE) were shown in our previous studies to reduce smoke-induced cellular damage in late bone-healing in vitro models. We aimed here to analyze their effects on the early fracture repair of smokers in a 3D co-culture model of fracture hematomas and endothelial cells. Both extracts did not alter the cellular viability at concentrations of up to 100 µg/mL. In early fracture repair in vitro, they were unable to reduce smoking-induced inflammation and induce osteo- or chondrogenicity. Regarding angiogenesis, smoking-induced stress in HUVECs could not be counteracted by both extracts. Furthermore, smoking-impaired tube formation was not restored by GE but was harmed by MBE. However, GE promoted angiogenesis initiation under smoking conditions via the Angpt/Tie2 axis. To summarize, cigarette smoking strikingly affected early fracture healing processes in vitro, but herbal extracts at the applied doses had only a limited effect. Since both extracts were shown before to be very effective in later stages of fracture healing, our data suggest that their early use immediately after fracture does not appear to negatively impact later beneficial effects.
Collapse
Affiliation(s)
| | | | | | | | - Andreas Nüssler
- Siegfried-Weller Institute for Trauma Research, BG Trauma Center, University of Tuebingen, Schnarrenbergstrasse 95, 72070 Tuebingen, Germany; (H.R.); (A.M.); (T.H.); (S.E.)
| |
Collapse
|
17
|
Jin Y, Li S, Yu Q, Chen T, Liu D. Application of stem cells in regeneration medicine. MedComm (Beijing) 2023; 4:e291. [PMID: 37337579 PMCID: PMC10276889 DOI: 10.1002/mco2.291] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/21/2023] Open
Abstract
Regeneration is a complex process affected by many elements independent or combined, including inflammation, proliferation, and tissue remodeling. Stem cells is a class of primitive cells with the potentiality of differentiation, regenerate with self-replication, multidirectional differentiation, and immunomodulatory functions. Stem cells and their cytokines not only inextricably linked to the regeneration of ectodermal and skin tissues, but also can be used for the treatment of a variety of chronic wounds. Stem cells can produce exosomes in a paracrine manner. Stem cell exosomes play an important role in tissue regeneration, repair, and accelerated wound healing, the biological properties of which are similar with stem cells, while stem cell exosomes are safer and more effective. Skin and bone tissues are critical organs in the body, which are essential for sustaining life activities. The weak repairing ability leads a pronounced impact on the quality of life of patients, which could be alleviated by stem cell exosomes treatment. However, there are obstacles that stem cells and stem cells exosomes trough skin for improved bioavailability. This paper summarizes the applications and mechanisms of stem cells and stem cells exosomes for skin and bone healing. We also propose new ways of utilizing stem cells and their exosomes through different nanoformulations, liposomes and nanoliposomes, polymer micelles, microspheres, hydrogels, and scaffold microneedles, to improve their use in tissue healing and regeneration.
Collapse
Affiliation(s)
- Ye Jin
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Shuangyang Li
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Qixuan Yu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Tianli Chen
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Da Liu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| |
Collapse
|
18
|
Toya M, Zhang N, Tsubosaka M, Kushioka J, Gao Q, Li X, Chow SKH, Goodman SB. CCL2 promotes osteogenesis by facilitating macrophage migration during acute inflammation. Front Cell Dev Biol 2023; 11:1213641. [PMID: 37457301 PMCID: PMC10348816 DOI: 10.3389/fcell.2023.1213641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Novel minimally invasive strategies are needed to obtain robust bone healing in complex fractures and bone defects in the elderly population. Local cell therapy is one potential option for future treatment. Mesenchymal stromal cells (MSCs) are not only involved in osteogenesis but also help direct the recruitment of macrophages during bone regeneration via MSC-macrophage crosstalk. The C-C motif chemokine ligand 2 (CCL2) is an inflammatory chemokine that is associated with the migration of macrophages and MSCs during inflammation. This study investigated the use of CCL2 as a therapeutic target for local cell therapy. MSCs and macrophages were isolated from 10 to 12 week-old BALB/c male mice. Genetically modified CCL2 over-expressing MSCs were produced using murine CCL2-secreting pCDH-CMV-mCCL2-copGFP expressing lentivirus vector. Osteogenic differentiation assays were performed using MSCs with or without macrophages in co-culture. Cell migration assays were also performed. MSCs transfected with murine CCL2-secreting pCDH-CMV-mCCL2-copGFP expressing lentivirus vector showed higher levels of CCL2 secretion compared to unaltered MSCs (p < 0.05). Genetic manipulation did not affect cell proliferation. CCL2 did not affect the osteogenic ability of MSCs alone. However, acute (1 day) but not sustained (7 days) stimulation with CCL2 increased the alizarin red-positive area when MSCs were co-cultured with macrophages (p < 0.001). Both recombinant CCL2 (p < 0.05) and CCL2 released from MSCs (p < 0.05) facilitated macrophage migration. We demonstrated that acute CCL2 stimulation promoted subsequent osteogenesis in co-culture of MSCs and macrophages. Acute CCL2 stimulation potentially facilitates osteogenesis during the acute inflammatory phase of bone healing by directing local macrophage migration, fostering macrophage-MSC crosstalk, and subsequently, by activating or licensing of MSCs by macrophage pro-inflammatory cytokines. The combination of CCL2, MSCs, and macrophages could be a potential strategy for local cell therapy in compromised bone healing.
Collapse
Affiliation(s)
- Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
19
|
Ferrie L, Premnath P, Olsen A, Larijani L, Besler BA, Rancourt DE, Duncan NA, Underhill TM, Krawetz RJ. Exogenously delivered iPSCs disrupt the natural repair response of endogenous MPCs after bone injury. Sci Rep 2023; 13:9378. [PMID: 37296277 PMCID: PMC10256810 DOI: 10.1038/s41598-023-36609-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 06/07/2023] [Indexed: 06/12/2023] Open
Abstract
Promoting bone healing including fracture non-unions are promising targets for bone tissue engineering due to the limited success of current clinical treatment methods. There has been significant research on the use of stem cells with and without biomaterial scaffolds to treat bone fractures due to their promising regenerative capabilities. However, the relative roles of exogenous vs. endogenous stem cells and their overall contribution to in vivo fracture repair is not well understood. The purpose of this study was to determine the interaction between exogenous and endogenous stem cells during bone healing. This study was conducted using a standardized burr-hole bone injury model in a mesenchymal progenitor cell (MPC) lineage-tracing mouse under normal homeostatic and osteoporotic conditions. Burr-hole injuries were treated with a collagen-I biomaterial loaded with and without labelled induced pluripotent stem cells (iPSCs). Using lineage-tracing, the roles of exogenous and endogenous stem cells during bone healing were examined. It was observed that treatment with iPSCs resulted in muted healing compared to untreated controls in intact mice post-injury. When the cell populations were examined histologically, iPSC-treated burr-hole defects presented with a dramatic reduction in endogenous MPCs and cell proliferation throughout the injury site. However, when the ovaries were removed and an osteoporotic-like phenotype induced in the mice, iPSCs treatment resulted in increased bone formation relative to untreated controls. In the absence of iPSCs, endogenous MPCs demonstrated robust proliferative and osteogenic capacity to undertake repair and this behaviour was disrupted in the presence of iPSCs which instead took on an osteoblast fate but with little proliferation. This study clearly demonstrates that exogenously delivered cell populations can impact the normal function of endogenous stem/progenitor populations during the normal healing cascade. These interactions need to be better understood to inform cell and biomaterial therapies to treat fractures.
Collapse
Affiliation(s)
- Leah Ferrie
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Priyatha Premnath
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- College of Engineering and Applied Science, University of Wisconsin Milwaukee, Milwaukee, WI, USA
| | - Alexandra Olsen
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Leila Larijani
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryce A Besler
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Derrick E Rancourt
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Neil A Duncan
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Civil Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada.
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
20
|
Abduljauwad SN, Habib T, Ur-Rehman H. Clay microparticles for the enhancement of bone regeneration: in vitro studies. Histochem Cell Biol 2023:10.1007/s00418-023-02189-2. [PMID: 37029830 PMCID: PMC10082439 DOI: 10.1007/s00418-023-02189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2023] [Indexed: 04/09/2023]
Abstract
Humans develop osteoporosis as they age, a disease characterized by the slow and consistent reduction in bone mass and the subsequent risk of fractures. Due to aging, the mesenchymal stem cells within the bone marrow niche, show a shift in differentiation from osteogenesis to adipogenesis. The challenge of osteoporosis treatment is being met with advances in nanotechnology and tissue engineering. In this study , we evaluated the potential of palygorskite clay mineral microparticles for the promotion of the osteogenic differentiation in human mesenchymal stem cells (hMSCs) in vitro. Alkaline phosphatase (ALP) activity and Alizarin red staining showed that osteogenic differentiation of hMSCs is enhanced in the presence of palygorskite clay. Although, gene expression analysis did not reveal upregulation of several osteogenic markers in the presence of the clay microparticles, another interaction mechanism resulted in the enhanced osteogenic differentiation of hMSCs. The charged surfaces of the palygorskite clay particles interact with the stem cells using their high adhesion characteristics, leading to complete bridging, adherence, and enveloping of the stem cells' cadherins and integrins with their environment. This restoration of the adhesion among the stem cells and their environment most probably promotes/restores the osteogenic differentiation of hMSCs. Therefore, palygorskite clay microparticles are a promising candidate for further in vivo studies on bone regeneration.
Collapse
Affiliation(s)
- Sahel N Abduljauwad
- Civil, Environmental Engineering Department, King Fahd University of Petroleum, Minerals (KFUPM), Dhahran, Saudi Arabia
| | - Taimur Habib
- School of Medicine, Royal College of Surgeons in Ireland (RCSI) Medical University, Busaiteen, Bahrain
| | - Habib Ur-Rehman
- Visiting Faculty, Civil, Environmental Engineering Department, King Fahd University of Petroleum, Minerals (KFUPM), Dhahran, Saudi Arabia.
- National University of Computer and Emerging Sciences (NUCES), Lahore, Pakistan.
| |
Collapse
|
21
|
Novel hybrid biocomposites for tendon grafts: The addition of silk to polydioxanone and poly(lactide-co-caprolactone) enhances material properties, in vitro and in vivo biocompatibility. Bioact Mater 2023; 25:291-306. [PMID: 36844365 PMCID: PMC9945711 DOI: 10.1016/j.bioactmat.2023.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Biopolymers play a critical role as scaffolds used in tendon and ligament (TL) regeneration. Although advanced biopolymer materials have been proposed with optimised mechanical properties, biocompatibility, degradation, and processability, it is still challenging to find the right balance between these properties. Here, we aim to develop novel hybrid biocomposites based on poly(p-dioxanone) (PDO), poly(lactide-co-caprolactone) (LCL) and silk to produce high-performance grafts suitable for TL tissue repair. Biocomposites containing 1-15% of silk were studied through a range of characterisation techniques. We then explored biocompatibility through in vitro and in vivo studies using a mouse model. We found that adding up to 5% silk increases the tensile properties, degradation rate and miscibility between PDO and LCL phases without agglomeration of silk inside the composites. Furthermore, addition of silk increases surface roughness and hydrophilicity. In vitro experiments show that the silk improved attachment of tendon-derived stem cells and proliferation over 72 h, while in vivo studies indicate that the silk can reduce the expression of pro-inflammatory cytokines after six weeks of implantation. Finally, we selected a promising biocomposite and created a prototype TL graft based on extruded fibres. We found that the tensile properties of both individual fibres and braided grafts could be suitable for anterior cruciate ligament (ACL) repair applications.
Collapse
|
22
|
Esposito A, Klüppel M, Wilson BM, Meka SRK, Spagnoli A. CXCR4 mediates the effects of IGF-1R signaling in rodent bone homeostasis and fracture repair. Bone 2023; 166:116600. [PMID: 36368465 PMCID: PMC10057209 DOI: 10.1016/j.bone.2022.116600] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/10/2022]
Abstract
Non-union fractures have considerable clinical and economic burdens and yet the underlying pathogenesis remains largely undetermined. The fracture healing process involves cellular differentiation, callus formation and remodeling, and implies the recruitment and differentiation of mesenchymal stem cells that are not fully characterized. C-X-C chemokine receptor 4 (CXCR4) and Insulin-like growth factor 1 receptor (IGF-1R) are expressed in the fracture callus, but their interactions still remain elusive. We hypothesized that the regulation of CXCR4 by IGF-1R signaling is essential to maintain the bone homeostasis and to promote fracture repair. By using a combination of in vivo and in vitro approaches, we found that conditional ablation of IGF-1R in osteochondroprogenitors led to defects in bone formation and mineralization that associated with altered expression of CXCR4 by a discrete population of endosteal cells. These defects were corrected by AMD3100 (a CXCR4 antagonist). Furthermore, we found that the inducible ablation of IGF-1R in osteochondroprogenitors led to fracture healing failure, that associated with an altered expression of CXCR4. In vivo AMD3100 treatment improved fracture healing and normalized CXCR4 expression. Moreover, we determined that these effects were mediated through the IGF-1R/Insulin receptor substrate 1 (IRS-1) signaling pathway. Taken together, our studies identified a novel population of endosteal cells that is functionally regulated through the modulation of CXCR4 by IGF-1R signaling, and such control is essential in bone homeostasis and fracture healing. Knowledge gained from these studies has the potential to accelerate the development of novel therapeutic interventions by targeting CXCR4 signaling to treat non-unions.
Collapse
Affiliation(s)
- Alessandra Esposito
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Michael Klüppel
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Brittany M Wilson
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Sai R K Meka
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Anna Spagnoli
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA; Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
23
|
Yun H, Kim B, Jeong YH, Hong JT, Park K. Suffruticosol A elevates osteoblast differentiation targeting BMP2-Smad/1/5/8-RUNX2 in pre-osteoblasts. Biofactors 2023; 49:127-139. [PMID: 35852295 PMCID: PMC10947220 DOI: 10.1002/biof.1878] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/03/2022] [Indexed: 11/11/2022]
Abstract
The Paeonia suffruticosa ANDR. (P. suffruticosa) is commonly used in traditional medicine for various purposes. Suffruticosol A (Suf-A), isolated from P. suffruticosa, is a beneficial compound with antibiofilm, antivirulence, and anti-inflammatory properties. The aim of the present study was to investigate the biological effects of Suf-A on osteogenic processes in pre-osteoblasts. It was determined here in that Suf-A (>98.02%), isolated from P. suffruticosa, showed no cytotoxicity at 0.1-30 μM; however, it induced cytotoxicity at 50-100 μM in pre-osteoblasts. Suf-A increased osteogenic alkaline phosphatase activity and expression levels of noncollagenous proteins. Adhesion and trans-migration on the extracellular matrix were potentiated by Suf-A, but not by wound-healing migration. Suf-A did not affect autophagy or necroptosis during osteoblast differentiation. We found that Suf-A increased runt-related transcription factor 2 (RUNX2) levels and mineralized matrix formation. RUNX2 expression was mediated by Suf-A-induced BMP2-Smad1/5/8 and mitogen-activated protein kinase signaling, as demonstrated by Noggin, a BMP2 inhibitor. These results suggest that Suf-A is a potential natural osteogenic compound.
Collapse
Affiliation(s)
- Hyung‐Mun Yun
- Department of Oral and Maxillofacial PathologySchool of Dentistry, Kyung Hee UniversitySeoulRepublic of Korea
| | - Bomi Kim
- National Development Institute of Korean MedicineGyeongsanRepublic of Korea
| | - Yun Hee Jeong
- National Development Institute of Korean MedicineGyeongsanRepublic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National UniversityChungbukRepublic of Korea
| | - Kyung‐Ran Park
- Gwangju CenterKorea Basic Science Institute (KBSI)GwangjuRepublic of Korea
| |
Collapse
|
24
|
Trifloroside Induces Bioactive Effects on Differentiation, Adhesion, Migration, and Mineralization in Pre-Osteoblast MC3T3E-1 Cells. Cells 2022; 11:cells11233887. [PMID: 36497145 PMCID: PMC9738977 DOI: 10.3390/cells11233887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Gentianae Scabrae Radix is used in traditional medicine and is known to possess bioactive compounds, including secoiridoid glycosides, flavonoids, lignans, and triterpenes. Trifloroside (TriFs) is a secoiridoid glycoside known for its antioxidant activity; however, its other effects have not been studied. In the present study, we investigated the biological effects of TriFs isolated from the roots of Gentianae Scabrae Radix using pre-osteoblast MC3T3E-1 cells. No cellular toxicity was observed with 1 μM TriFs, whereas 5-100 μM TriFs showed a gradual increase in cell viability. Alkaline phosphatase staining and microscopic observations revealed that 1-10 μM TriFs stimulated osteogenic activity during early osteoblast differentiation. Trifloroside also increased mineral apposition during osteoblast maturation. Biochemical analyses revealed that TriFs promoted nuclear RUNX2 expression and localization by stimulating the major osteogenic BMP2-Smad1/5/8-RUNX2 pathway. Trifloroside also increased p-GSK3β, β-catenin, p-JNK, and p-p38, but not Wnt3a, p-AKT, and p-ERK. Moreover, TriFs increased the MMP13 levels and promoted cell migration and adhesion. In contrast, TriFs-induced osteoblast differentiation and maturation had negligible effects on autophagy and necrosis. Our findings suggest that TriFs induces osteogenic effects through differentiation, adhesion, migration, and mineral apposition. Therefore, TriFs is suggested as a potential drug target in osteoblast-mediated bone diseases.
Collapse
|
25
|
Yun HM, Lee JY, Kim B, Park KR. Suffruticosol B Is an Osteogenic Inducer through Osteoblast Differentiation, Autophagy, Adhesion, and Migration. Int J Mol Sci 2022; 23:ijms232113559. [PMID: 36362346 PMCID: PMC9658763 DOI: 10.3390/ijms232113559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Suffruticosol B (Suf-B) is a stilbene found in Paeonia suffruticosa ANDR., which has been traditionally used in medicine. Stilbenes and their derivatives possess various pharmacological effects, such as anticancer, anti-inflammatory, and anti-osteoporotic activities. This study aimed to explore the bone-forming activities and mechanisms of Suf-B in pre-osteoblasts. Herein, >99.9% pure Suf-B was isolated from P. suffruticosa methanolic extracts. High concentrations of Suf-B were cytotoxic, whereas low concentrations did not affect cytotoxicity in pre-osteoblasts. Under zero levels of cytotoxicity, Suf-B exhibited bone-forming abilities by enhancing alkaline phosphatase enzyme activities, bone matrix calcification, and expression levels with non-collagenous proteins. Suf-B induces intracellular signal transduction, leading to nuclear RUNX2 expression. Suf-B-stimulated differentiation showed increases in autophagy proteins and autophagosomes, as well as enhancement of osteoblast adhesion and transmigration on the ECM. These results indicate that Suf-B has osteogenic qualities related to differentiation, autophagy, adhesion, and migration. This also suggests that Suf-B could have a therapeutic effect as a phytomedicine in skeletal disorders.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea
| | - Joon Yeop Lee
- National Development Institute for Korean Medicine, Gyeongsan 38540, Korea
| | - Bomi Kim
- National Development Institute for Korean Medicine, Gyeongsan 38540, Korea
| | - Kyung-Ran Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Korea
- Correspondence: ; Tel.: +82-62-712-4412; Fax: +82-62-372-4102
| |
Collapse
|
26
|
Rahyussalim AJ, Nasser MK, Al As'ady FM, Kurniawati T. Umbilical cord-derived mesenchymal stem cells implantation on Hemivertebra defect with three-year follow-up: Biological approach in congenital scoliosis treatment - A case report. Int J Surg Case Rep 2022; 99:107602. [PMID: 36116304 PMCID: PMC9568731 DOI: 10.1016/j.ijscr.2022.107602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction and importance Congenital scoliosis is abnormal vertebral column growth and development during embryogenesis. The most common type of congenital scoliosis is failure of growth which is called as hemivertebra. However, the recent surgical treatment of hemivertebra has several complications especially in young patient. The mesenchymal stem cells (MSCs) have been used to treat several bone problems including bone defect and may be have potential to treat the defect in hemiverterbra. We reported a hemivertebra treated by umbilical cord-derived MSCs (UC-MSCs). Case presentation A two-year-old boy presented with scoliosis deformity. The mother noticed the patient's deformity when he was 10th month of age as he learned to stand and progressed since then. There were no growth and development problems. On physical examination, the patient appeared to have scoliosis at lumbar level with bending to the right and asymmetry of waist fold with left shoulder depression. Based on X-ray and CT-Scan investigations, the patient was diagnosed with single fully segmented hemivertebra at 3rd lumbar level. 20 × 106 UC-MSCs were implanted into the bone defect of hemivertebra. Clinical discussion At three-year follow-up, the X-ray and MRI investigations showed a decrease of Cobb angle and increase of hemivertebra ratio. These findings may be due to improvement of the bone defect, which is consistent with several studies that MSCs have abilities to promote bone formation by maintaining the osteoblast cells and improving vascularization. Conclusion We found that MSCs therapy for hemivertebra represent a potential therapy to correct scoliosis curvature and prevent further curvature. Further clinical studies are required to investigate the efficacy of this therapy in hemivertebra. Hemivertebrae is due to the failure of development of vertebrae. Mesenchymal stem cells induce new bone formation. Symmetrical growth of the spine is required for prevent spinal deformity. Regenerative therapy using mesenchymal stem cells as treatment strategy for hemivertebra Implantation of mesenchymal stem cells in hemivertebra is safe procedure.
Collapse
Affiliation(s)
- Ahmad Jabir Rahyussalim
- Department of Orthopaedic & Traumatology, Cipto Mangunkusumo National General Hospital and Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia; Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia; Stem Cells and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.
| | - Mochammad Kamal Nasser
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia; Post Graduate Medical Doctor, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Faiz Muhammad Al As'ady
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Tri Kurniawati
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia; Stem Cells and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
27
|
Wang T, Huang S, He C. Senescent cells: A therapeutic target for osteoporosis. Cell Prolif 2022; 55:e13323. [DOI: 10.1111/cpr.13323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
- Institute of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | - Shishu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital and West China School of Medicine Sichuan University Chengdu Sichuan China
| | - Chengqi He
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
- Institute of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| |
Collapse
|
28
|
Yao H, Zhang L, Yan S, He Y, Zhu H, Li Y, Wang D, Yang K. Low-intensity pulsed ultrasound/nanomechanical force generators enhance osteogenesis of BMSCs through microfilaments and TRPM7. J Nanobiotechnology 2022; 20:378. [PMID: 35964037 PMCID: PMC9375242 DOI: 10.1186/s12951-022-01587-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Low-intensity pulsed ultrasound (LIPUS) has been reported to accelerate fracture healing, but the mechanism is unclear and its efficacy needs to be further optimized. Ultrasound in combination with functionalized microbubbles has been shown to induce local shear forces and controllable mechanical stress in cells, amplifying the mechanical effects of LIPUS. Nanoscale lipid bubbles (nanobubbles) have high stability and good biosafety. However, the effect of LIPUS combined with functionalized nanobubbles on osteogenesis has rarely been studied. RESULTS In this study, we report cyclic arginine-glycine-aspartic acid-modified nanobubbles (cRGD-NBs), with a particle size of ~ 500 nm, able to actively target bone marrow mesenchymal stem cells (BMSCs) via integrin receptors. cRGD-NBs can act as nanomechanical force generators on the cell membrane, and further enhance the BMSCs osteogenesis and bone formation promoted by LIPUS. The polymerization of actin microfilaments and the mechanosensitive transient receptor potential melastatin 7 (TRPM7) ion channel play important roles in BMSCs osteogenesis promoted by LIPUS/cRGD-NBs. Moreover, the mutual regulation of TRPM7 and actin microfilaments promote the effect of LIPUS/cRGD-NBs. The extracellular Ca2 + influx, controlled partly by TRPM7, could participated in the effect of LIPUS/cRGD-NBs on BMSCs. CONCLUSIONS The nanomechanical force generators cRGD-NBs could promote osteogenesis of BMSCs and bone formation induced by LIPUS, through regulation TRPM7, actin cytoskeleton, and intracellular calcium oscillations. This study provides new directions for optimizing the efficacy of LIPUS for fracture healing, and a theoretical basis for the further application and development of LIPUS in clinical practice.
Collapse
Affiliation(s)
- Huan Yao
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, 400014, China.,Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shujin Yan
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiman He
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hui Zhu
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yasha Li
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, 400014, China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ke Yang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, 400014, China.
| |
Collapse
|
29
|
Jing Z, Liang Z, Yang L, Du W, Yu T, Tang H, Li C, Wei W. Bone formation and bone repair: The roles and crosstalk of osteoinductive signaling pathways. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Lin Y, Zhou HC, Chen N, Ren Y, Gao R, Li Q, Deng Y, Han X, Zhang X, Xiang AP, Guo B, Liu C, Ren J. Unveiling the improved targeting migration of mesenchymal stem cells with CXC chemokine receptor 3-modification using intravital NIR-II photoacoustic imaging. J Nanobiotechnology 2022; 20:307. [PMID: 35764961 PMCID: PMC9238014 DOI: 10.1186/s12951-022-01513-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/14/2022] [Indexed: 12/13/2022] Open
Abstract
Background Therapy with genetically modified mesenchymal stem cells (MSCs) has clinical translation promise. Optimizing the targeting migratory ability of MSCs relies on accurate imaging of the distribution and extravasation kinetics of MSCs, and the corresponding imaging results could be used to predict therapeutic outcomes and guide the optimization of the treatment program. Among the different imaging modalities, second near-infrared (NIR-II) optical-resolution photoacoustic microscopy (OR-PAM) has merits, including a fine resolution, a deep penetration, a high sensitivity, and a large signal-to-background ratio. It would be an ideal candidate for precise monitoring of MSCs, although it has not been tested for this purpose so far. Results Penetrating peptide-decorated conjugated polymer nanoparticles (TAT-CPNPs) with strong NIR-II absorbance were used to label chemokine-receptor genetically modified MSCs, which were subsequently evaluated under intravital NIR-II OR-PAM regarding their targeting migratory ability. Based on the upregulation of chemokine (C-X-C motif) ligand 10 in the inflamed ears of contact hypersensitivity mice, MSCs with overexpression of corresponding receptor, chemokine (C-X-C motif) receptor 3 (Cxcr3) were successfully generated (MSCCxcr3). TAT-CPNPs labeling enabled NIR-II photoacoustic imaging to discern MSCCxcr3 covered by 1.2 cm of chicken breast tissue. Longitudinal OR-PAM imaging revealed enhanced inflammation-targeting migration of MSCCxcr3 over time attributed to Cxcr3 gene modification, which was further validated by histological analysis. Conclusions TAT-CPNPs-assisted NIR-II PA imaging is promising for monitoring distribution and extravasation kinetics of MSCs, which would greatly facilitate optimizing MSC-based therapy. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01513-7.
Collapse
Affiliation(s)
- Yuejun Lin
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Hui-Chao Zhou
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Ningbo Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yaguang Ren
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Rongkang Gao
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qiaojia Li
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yiwen Deng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xuejiao Han
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen, 518055, China.
| | - Chengbo Liu
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Jie Ren
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
31
|
Shen H, Jiang W, Yu Y, Feng Y, Zhang T, Liu Y, Guo L, Zhou N, Huang X. microRNA-146a mediates distraction osteogenesis via bone mesenchymal stem cell inflammatory response. Acta Histochem 2022; 124:151913. [PMID: 35759812 DOI: 10.1016/j.acthis.2022.151913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/09/2022] [Accepted: 05/24/2022] [Indexed: 11/20/2022]
Abstract
Distraction osteogenesis (DO) is a widely used surgical technique to repair bone defects, partly owing to its high efficiency in inducing osteogenesis; however, the process of osteogenesis is complex, and the precise mechanism is still unclear. Among the factors identified for an effective DO procedure, well-controlled inflammation is essential. We aimed to explore how microRNA(miR)-146a, a negative regulator of inflammation, influences osteogenesis in DO. First, we established canine right mandibular DO and bone fracture models to evaluate the expression level of miR-146a in response to these procedures. Second, bone marrow mesenchymal stem cells (BMSCs) were isolated from healthy puppies and cultured with lipopolysaccharide (LPS) to observe how inflammation affects osteogenesis. Finally, the osteogenesis activity of BMSCs transfected with lentiviral vector either overexpressing (miR-146a-up) or inhibited for miR-146a expression was evaluated. miR-146a-up-transfected BMSCs were injected locally into the distraction gaps of the DO model canines. On days 42 and 56 post-surgery, the bone volume/tissue volume and bone mineral density values were evaluated via using micro-computed tomography, and newly formed tissues were harvested and evaluated via histological staining. The expression of miR-146a in both the DO canine model and LPS-stimulated BMSCs increased. Overexpression of miR-146a enhanced cell proliferation, migration, and osteogenic differentiation. Additionally, the newly formed callus was improved in canine mandibles injected with miR-146a-up-transfected BMSCs. In summary, miR-146a regulates mandibular DO by improving osteogenesis, and can serve as a potential target to shorten the therapy period of DO.
Collapse
Affiliation(s)
- Huijuan Shen
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Weidong Jiang
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Yangyang Yu
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Yuan Feng
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Tao Zhang
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Yan Liu
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Lina Guo
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China
| | - Nuo Zhou
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China.
| | - Xuanping Huang
- Departement of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning 530021, People's Republic of China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning 530021, People's Republic of China.
| |
Collapse
|
32
|
Sabbaghzadeh A, Bonakdar S, Gorji M, Gholipour M. Evaluation of the effect of preoperative hemoglobin level and proinflammatory factors on intertrochanteric fracture union. Wien Klin Wochenschr 2022; 134:458-462. [PMID: 35639200 DOI: 10.1007/s00508-022-02042-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Intertrochanteric fractures are associated with high mortality and morbidity, so these patients should undergo fracture fixation surgery immediately. Despite surgery, the possibility of fracture fusion may not occur due to the association with various causes. Therefore, our aim is to investigate these factors (TNF‑a, IL‑1, Hb) and their effect on fracture union after fixation. METHODS From 2018 to 2020, at our orthopedic trauma center, 163 patients older than 50 years with intertrochanteric fractures underwent DHS fixation surgery. Patients were divided into anemic and non-anemic groups in terms of preoperative hemoglobin level (standard hemoglobin 11 mg/dl). For 3 months, patients were assessed for union and failure fixation criteria, levels of proinflammation (TNF‑α, IL-1) and level of hemoglobin. RESULTS The results show that out of 163 patients with fractures, at the time of initial admission, 74 patients had less than 11 hemoglobin g/dl. Patients with union fractures had higher hemoglobin levels than patients with non-union (11.71 ± 1.51 versus 11.24 ± 1.96), which was statistically significant between hemoglobin and union level (p = 0.030). At the end of the third visit (third month), 44 (59.5%) anemic patients received union completly, while among the patients with normal hemoglobin level, 32 (36%) received union bread, which was statistically significant (p = 0.003). There were no statistically significant differences between proinflammatory factors before surgery and 3 months after surgery (p > 0.05). CONCLUSION Due to the effect of anemia and proinflammatory factors in the process of healing fractures and bone formation and creating musculoskeletal balance, low hemoglobin level before surgery has a significant effect on fracture union and failure of fixation. So it is recommended to correct this anemia in these patients before surgery and during follow-up.
Collapse
Affiliation(s)
- Amir Sabbaghzadeh
- Physiotherapy Research Center, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran.,Clinical Research Development Unit, Akhtar Hospital Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Sona Bonakdar
- Department of Foreign Languages, Urmia University, Urmia, Iran
| | - Mona Gorji
- Physiotherapy Research Center, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran.,Skin research center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Morteza Gholipour
- Physiotherapy Research Center, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran. .,Clinical Research Development Unit, Akhtar Hospital Shahid Beheshti University of Medical Science, Tehran, Iran.
| |
Collapse
|
33
|
Chen LS, Zhang M, Chen P, Xiong XF, Liu PQ, Wang HB, Wang JJ, Shen J. The m 6A demethylase FTO promotes the osteogenesis of mesenchymal stem cells by downregulating PPARG. Acta Pharmacol Sin 2022; 43:1311-1323. [PMID: 34462564 PMCID: PMC9061799 DOI: 10.1038/s41401-021-00756-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022] Open
Abstract
N6-methyladenosine (m6A) is the most abundant posttranscriptional methylation modification that occurs in mRNA and modulates the fine-tuning of various biological processes in mammalian development and human diseases. In this study we investigated the role of m6A modification in the osteogenesis of mesenchymal stem cells (MSCs), and the possible mechanisms by which m6A modification regulated the processes of osteoporosis and bone necrosis. We performed systematic analysis of the differential gene signatures in patients with osteoporosis and bone necrosis and conducted m6A-RNA immunoprecipitation (m6A-RIP) sequencing to identify the potential regulatory genes involved in osteogenesis. We showed that fat mass and obesity (FTO), a primary m6A demethylase, was significantly downregulated in patients with osteoporosis and osteonecrosis. During the differentiation of human MSCs into osteoblasts, FTO was markedly upregulated. Both depletion of FTO and application of the FTO inhibitor FB23 or FB23-2 impaired osteogenic differentiation of human MSCs. Knockout of FTO in mice resulted in decreased bone mineral density and impaired bone formation. PPARG, a biomarker for osteoporosis, was identified as a critical downstream target of FTO. We further revealed that FTO mediated m6A demethylation in the 3'UTR of PPARG mRNA, and reduced PPARG mRNA stability in an YTHDF1-dependent manner. Overexpression of PPARG alleviated FTO-mediated osteogenic differentiation of MSCs, whereas knockdown of PPARG promoted FTO-induced expression of the osteoblast biomarkers ALPL and OPN during osteogenic differentiation. Taken together, this study demonstrates the functional significance of the FTO-PPARG axis in promoting the osteogenesis of human MSCs and sheds light on the role of m6A modification in mediating osteoporosis and osteonecrosis.
Collapse
Affiliation(s)
- Liu-shan Chen
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006 China
| | - Meng Zhang
- grid.414011.10000 0004 1808 090XDepartment of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, 450003 China
| | - Peng Chen
- grid.411866.c0000 0000 8848 7685The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Xiao-feng Xiong
- grid.12981.330000 0001 2360 039XDepartment of Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Pei-qing Liu
- grid.12981.330000 0001 2360 039XDepartment of Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Hai-bin Wang
- grid.411866.c0000 0000 8848 7685The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Jun-jian Wang
- grid.12981.330000 0001 2360 039XDepartment of Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
34
|
Yahara Y, Nguyen T, Ishikawa K, Kamei K, Alman BA. The origins and roles of osteoclasts in bone development, homeostasis and repair. Development 2022; 149:275249. [PMID: 35502779 PMCID: PMC9124578 DOI: 10.1242/dev.199908] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying bone development, repair and regeneration are reliant on the interplay and communication between osteoclasts and other surrounding cells. Osteoclasts are multinucleated monocyte lineage cells with resorptive abilities, forming the bone marrow cavity during development. This marrow cavity, essential to hematopoiesis and osteoclast-osteoblast interactions, provides a setting to investigate the origin of osteoclasts and their multi-faceted roles. This Review examines recent developments in the embryonic understanding of osteoclast origin, as well as interactions within the immune environment to regulate normal and pathological bone development, homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Tuyet Nguyen
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Koji Ishikawa
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Orthopaedic Surgery, Showa University School of Medicine, Tokyo, 142-8666, Japan
| | - Katsuhiko Kamei
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
35
|
Park KR, Kwon YJ, Jeong YH, Hong JT, Yun HM. Thelephoric acid, p-terphenyl, induces bone-forming activities in pre-osteoblasts. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
36
|
Wang Y, Li J, Zhou J, Qiu Y, Song J. Low-intensity pulsed ultrasound enhances bone marrow-derived stem cells-based periodontal regenerative therapies. ULTRASONICS 2022; 121:106678. [PMID: 35051693 DOI: 10.1016/j.ultras.2021.106678] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/17/2021] [Accepted: 12/30/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Alveolar bone loss is one of the most common consequence for periodontitis, which is a major obstacle in periodontal regeneration. Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of various disease, which also contribute to the natural bone repair process. Low-intensity pulsed ultrasound (LIPUS) is a therapeutic ultrasound used in our previous studies to promotes alveolar bone regeneration. In addition, LIPUS was found to be a promising method to enhance mesenchymal stromal cell-based therapies. In the current study, we have investigated the effects of LIPUS combined with BMSCs therapies on BMSCs homing and its potential to promote alveolar bone regeneration. METHODS BMSCs were isolated from rat and characterized by multilineages differentiation assay. Then these cells were labeled with luciferase and green fluorescent protein (GFP) by lentivirus in vitro. Periodontal bone defect was made on the mesial area of the maxillary first molar in rats. A total of 1 × 106 Luc-GFP labeled BMSCs were injected into rat tail vein. Bioluminescence imaging was utilized to track BMSCs in vivo. The rats were sacrificed eight weeks after surgery and the samples were harvested. Micro-computed tomography (Micro-CT) was performed to evaluate alveolar bone regeneration. Paraffin sections were made and subject to hematoxylin-eosin staining, masson staining and immunohistochemistry staining. RESULTS BMSCs display a fibroblast-like morphology and can differentiate into adipocytes or osteoblasts under appropriate condition. The transfected BMSCs are strongly positive for GFP express. Bioluminescence imaging showed that most of BMSCs were trapped in the lung. A small portion BMSCs were homed to the alveolar bone defect area in BMSCs group, while more cells were observed in BMSCs/LIPUS group compare to other groups on day 3 and 7. Micro-CT results showed that BMSCs/LIPUS group resulted in more new bone formation than other groups. Immunohistochemical results showed higher expression of COL-I and osteopontin in BMSCs/LIPUS group compared with the other groups. CONCLUSIONS These results suggested that LIPUS can enhance BMSCs-based periodontal alveolar bone regeneration. This study provides new insights into how LIPUS might provide therapeutic benefits by promoting BMSCs homing.
Collapse
Affiliation(s)
- Yunji Wang
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jianpin Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ye Qiu
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
37
|
Hirata H, Zhang N, Ueno M, Barati D, Kushioka J, Shen H, Tsubosaka M, Toya M, Lin T, Huang E, Yao Z, Wu JY, Zwingenberger S, Yang F, Goodman SB. Ageing attenuates bone healing by mesenchymal stem cells in a microribbon hydrogel with a murine long bone critical-size defect model. Immun Ageing 2022; 19:14. [PMID: 35279175 PMCID: PMC8917642 DOI: 10.1186/s12979-022-00272-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/24/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite the high incidence of fractures and pseudoarthrosis in the aged population, a potential role for the use of mesenchymal stem cells (MSCs) in the treatment of bone defects in elderly patients has not been elucidated. Inflammation and the innate immune system, including macrophages, play crucial roles in the differentiation and activation of MSCs. We have developed lentivirus-transduced interleukin 4 (IL4) over-expressing MSCs (IL4-MSCs) to polarize macrophages to an M2 phenotype to promote bone healing in an established young murine critical size bone defect model. In the current study, we explore the potential of IL4-MSCs in aged mice. METHODS A 2 mm femoral diaphyseal bone defect was created and fixed with an external fixation device in 15- to 17-month-old male and female BALB/c mice. Microribbon (µRB) scaffolds (Sc) with or without encapsulation of MSCs were implanted in the defect sites. Accordingly, the mice were divided into three treatment groups: Sc-only, Sc + MSCs, and Sc + IL4-MSCs. Mice were euthanized six weeks after the surgery; subsequently, MicroCT (µCT), histochemical and immunohistochemical analyses were performed. RESULTS µCT analysis revealed that bone formation was markedly enhanced in the IL4-MSC group. Compared with the Sc-only, the amount of new bone increased in the Sc + MSCs and Sc + IL4-MSC groups. However, no bridging of bone was observed in all groups. H&E staining showed fibrous tissue within the defect in all groups. Alkaline phosphatase (ALP) staining was increased in the Sc + IL4-MSC group. The Sc + IL4-MSCs group showed a decrease in the number of M1 macrophages and an increase in the number of M2 macrophages, with a significant increase in the M2/M1 ratio. DISCUSSION IL4 promotes macrophage polarization to an M2 phenotype, facilitating osteogenesis and vasculogenesis. The addition of IL4-MSCs in the µRB scaffold polarized macrophages to an M2 phenotype and increased bone formation; however, complete bone bridging was not observed in any specimens. These results suggest that IL4-MSCs are insufficient to heal a critical size bone defect in aged mice, as opposed to younger animals. Additional therapeutic strategies are needed in this challenging clinical scenario.
Collapse
Affiliation(s)
- Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA.,Department of Orthopaedic Surgery, Saga University, Saga, Japan
| | - Danial Barati
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Joy Y Wu
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Stefan Zwingenberger
- University Center for Orthopaedics, Traumatology, and Plastic Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA.,Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA. .,Department of Bioengineering, Stanford University, Stanford, California, USA.
| |
Collapse
|
38
|
Mineralizing Gelatin Microparticles as Cell Carrier and Drug Delivery System for siRNA for Bone Tissue Engineering. Pharmaceutics 2022; 14:pharmaceutics14030548. [PMID: 35335924 PMCID: PMC8949427 DOI: 10.3390/pharmaceutics14030548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/19/2022] Open
Abstract
The local release of complexed siRNA from biomaterials opens precisely targeted therapeutic options. In this study, complexed siRNA was loaded to gelatin microparticles cross-linked (cGM) with an anhydride-containing oligomer (oPNMA). We aggregated these siRNA-loaded cGM with human mesenchymal stem cells (hMSC) to microtissues and stimulated them with osteogenic supplements. An efficient knockdown of chordin, a BMP-2 antagonist, caused a remarkably increased alkaline phosphatase (ALP) activity in the microtissues. cGM, as a component of microtissues, mineralized in a differentiation medium within 8–9 days, both in the presence and in the absence of cells. In order to investigate the effects of our pre-differentiated and chordin-silenced microtissues on bone homeostasis, we simulated in vivo conditions in an unstimulated co-culture system of hMSC and human peripheral blood mononuclear cells (hPBMC). We found enhanced ALP activity and osteoprotegerin (OPG) secretion in the model system compared to control microtissues. Our results suggest osteoanabolic effects of pre-differentiated and chordin-silenced microtissues.
Collapse
|
39
|
He W, Shi X, Guo Z, Wang H, Kang M, Lv Z. Circ_0019693 promotes osteogenic differentiation of bone marrow mesenchymal stem cell and enhances osteogenesis-coupled angiogenesis via regulating microRNA-942-5p-targeted purkinje cell protein 4 in the development of osteoporosis. Bioengineered 2022; 13:2181-2193. [PMID: 35030971 PMCID: PMC8973649 DOI: 10.1080/21655979.2021.2023982] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Circular RNA (circRNA) is a crucial regulator in multiple human diseases, including osteoporosis (OP). However, the function of numerous circRNAs remains unclear. This study aimed to explore the role and mechanism of circ_0019693 in bone marrow mesenchymal stem cell (BMSC) osteogenic differentiation and osteogenesis-coupled angiogenesis. The expression of circ_0019693, miR-942-5p and purkinje cell protein 4 (PCP4) was measured using quantitative real-time PCR (qPCR) or Western blot. Osteogenic differentiation was monitored according to the protein levels of RUNX family transcription factor 2 (RUNX2), osteopontin (OPN) and osteocalcin (OCN) by Western blot analysis, and the activity of alkaline phosphatase (ALP). Angiogenesis was evaluated by tube formation assay. The targeting relationship between miR-942-5p and circ_0019693 or PCP4 was identified using pull-down, dual-luciferase reporter, and RNA immunoprecipitation assays. Circ_0019693 was downregulated in serum samples and bone tissues from OP patients relative to normal subjects. Circ_0019693 expression was enhanced in the stages of BMSC osteogenic differentiation. Circ_0019693 overexpression enhanced the activity of ALP and the expression of RUNX2, OPN and OCN, and its overexpression also promoted angiogenesis. However, circ_0019693 knockdown played the opposite effects. MiR-942-5p was ensured to be a target of circ_0019693, and miR-942-5p enrichment reversed the effects of circ_0019693. In addition, PCP4 was a target of miR-942-5p, and miR-942-5p inhibitor-promoted BMSC osteogenic differentiation and angiogenesis were partly repressed by PCP4 knockdown. In conclusion, circ_0019693 promotes BMSC osteogenic differentiation osteogenesis-coupled angiogenesis via regulating miR-942-5p-targeted PCP4, thus blocking the development of OP.
Collapse
Affiliation(s)
| | | | - Zhenye Guo
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan City, Shanxi Province, China
| | - Huan Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan City, Shanxi Province, China
| | - Mingming Kang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan City, Shanxi Province, China
| | - Zhi Lv
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan City, Shanxi Province, China
| |
Collapse
|
40
|
Power RN, Cavanagh BL, Dixon JE, Curtin CM, O’Brien FJ. Development of a Gene-Activated Scaffold Incorporating Multifunctional Cell-Penetrating Peptides for pSDF-1α Delivery for Enhanced Angiogenesis in Tissue Engineering Applications. Int J Mol Sci 2022; 23:1460. [PMID: 35163379 PMCID: PMC8835777 DOI: 10.3390/ijms23031460] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/18/2022] Open
Abstract
Non-viral gene delivery has become a popular approach in tissue engineering, as it permits the transient delivery of a therapeutic gene, in order to stimulate tissue repair. However, the efficacy of non-viral delivery vectors remains an issue. Our lab has created gene-activated scaffolds by incorporating various non-viral delivery vectors, including the glycosaminoglycan-binding enhanced transduction (GET) peptide into collagen-based scaffolds with proven osteogenic potential. A modification to the GET peptide (FLR) by substitution of arginine residues with histidine (FLH) has been designed to enhance plasmid DNA (pDNA) delivery. In this study, we complexed pDNA with combinations of FLR and FLH peptides, termed GET* nanoparticles. We sought to enhance our gene-activated scaffold platform by incorporating GET* nanoparticles into collagen-nanohydroxyapatite scaffolds with proven osteogenic capacity. GET* N/P 8 was shown to be the most effective formulation for delivery to MSCs in 2D. Furthermore, GET* N/P 8 nanoparticles incorporated into collagen-nanohydroxyapatite (coll-nHA) scaffolds at a 1:1 ratio of collagen:nanohydroxyapatite was shown to be the optimal gene-activated scaffold. pDNA encoding stromal-derived factor 1α (pSDF-1α), an angiogenic chemokine which plays a role in BMP mediated differentiation of MSCs, was then delivered to MSCs using our optimised gene-activated scaffold platform, with the aim of significantly increasing angiogenesis as an important precursor to bone repair. The GET* N/P 8 coll-nHA scaffolds successfully delivered pSDF-1α to MSCs, resulting in a significant, sustained increase in SDF-1α protein production and an enhanced angiogenic effect, a key precursor in the early stages of bone repair.
Collapse
Affiliation(s)
- Rachael N. Power
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), D02 YN77 Dublin, Ireland; (R.N.P.); (C.M.C.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI, D02 YN77 Dublin, Ireland
| | | | - James E. Dixon
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Caroline M. Curtin
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), D02 YN77 Dublin, Ireland; (R.N.P.); (C.M.C.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), D02 YN77 Dublin, Ireland; (R.N.P.); (C.M.C.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI, D02 YN77 Dublin, Ireland
| |
Collapse
|
41
|
Extracellular Vesicles in Musculoskeletal Regeneration: Modulating the Therapy of the Future. Cells 2021; 11:cells11010043. [PMID: 35011605 PMCID: PMC8750529 DOI: 10.3390/cells11010043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
Tissue regeneration is a hot topic in health sciences, particularly because effective therapies promoting the healing of several cell types are lacking, specifically those of the musculoskeletal system. Mesenchymal Stem/Stromal Cells (MSCs) have been identified as crucial players in bone homeostasis, and are considered a promising therapy for diseases such as osteoarthritis (OA) and Rheumatoid Arthritis (RA). However, some known drawbacks limit their use, particularly ethical issues and immunological rejections. Thus, MSCs byproducts, namely Extracellular Vesicles (EVs), are emerging as potential solutions to overcome some of the issues of the original cells. EVs can be modulated by either cellular preconditioning or vesicle engineering, and thus represent a plastic tool to be implemented in regenerative medicine. Further, the use of biomaterials is important to improve EV delivery and indirectly to modulate their content and secretion. This review aims to connect the dots among MSCs, EVs, and biomaterials, in the context of musculoskeletal diseases.
Collapse
|
42
|
Krstić J, Mojsilović S, Mojsilović SS, Santibanez JF. Regulation of the mesenchymal stem cell fate by interleukin-17: Implications in osteogenic differentiation. World J Stem Cells 2021; 13:1696-1713. [PMID: 34909118 PMCID: PMC8641017 DOI: 10.4252/wjsc.v13.i11.1696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/14/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023] Open
Abstract
Bone regeneration is a tightly regulated process that ensures proper repair and functionality after injury. The delicate balance between bone formation and resorption is governed by cytokines and signaling molecules released during the inflammatory response. Interleukin (IL)-17A, produced in the early phase of inflammation, influences the fate of osteoprogenitors. Due to their inherent capacity to differentiate into osteoblasts, mesenchymal stem/stromal cells (MSCs) contribute to bone healing and regeneration. This review presents an overview of IL-17A signaling and the leading cellular and molecular mechanisms by which it regulates the osteogenic differentiation of MSCs. The main findings demonstrating IL-17A’s influence on osteoblastogenesis are described. To this end, divergent information exists about the capacity of IL-17A to regulate MSCs’ osteogenic fate, depending on the tissue context and target cell type, along with contradictory findings in the same cell types. Therefore, we summarize the data showing both the pro-osteogenic and anti-osteogenic roles of IL-17, which may help in the understanding of IL-17A function in bone repair and regeneration.
Collapse
Affiliation(s)
- Jelena Krstić
- Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria
| | - Slavko Mojsilović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11129, Serbia
| | - Sonja S Mojsilović
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, Belgrade 11129, Serbia
| | - Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O’Higgins, Chile 8370993, Chile
| |
Collapse
|
43
|
Valdés-Fernández J, López-Martínez T, Ripalda-Cemboráin P, Calvo IA, Sáez B, Romero-Torrecilla JA, Aldazabal J, Muiños-López E, Montiel V, Orbe J, Rodríguez JA, Páramo JA, Prósper F, Granero-Moltó F. Molecular and Cellular Mechanisms of Delayed Fracture Healing in Mmp10 (Stromelysin 2) Knockout Mice. J Bone Miner Res 2021; 36:2203-2213. [PMID: 34173256 DOI: 10.1002/jbmr.4403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 11/08/2022]
Abstract
The remodeling of the extracellular matrix is a central function in endochondral ossification and bone homeostasis. During secondary fracture healing, vascular invasion and bone growth requires the removal of the cartilage intermediate and the coordinate action of the collagenase matrix metalloproteinase (MMP)-13, produced by hypertrophic chondrocytes, and the gelatinase MMP-9, produced by cells of hematopoietic lineage. Interfering with these MMP activities results in impaired fracture healing characterized by cartilage accumulation and delayed vascularization. MMP-10, Stromelysin 2, a matrix metalloproteinase with high homology to MMP-3 (Stromelysin 1), presents a wide range of putative substrates identified in vitro, but its targets and functions in vivo and especially during fracture healing and bone homeostasis are not well defined. Here, we investigated the role of MMP-10 through bone regeneration in C57BL/6 mice. During secondary fracture healing, MMP-10 is expressed by hematopoietic cells and its maximum expression peak is associated with cartilage resorption at 14 days post fracture (dpf). In accordance with this expression pattern, when Mmp10 is globally silenced, we observed an impaired fracture-healing phenotype at 14 dpf, characterized by delayed cartilage resorption and TRAP-positive cell accumulation. This phenotype can be rescued by a non-competitive transplant of wild-type bone marrow, indicating that MMP-10 functions are required only in cells of hematopoietic linage. In addition, we found that this phenotype is a consequence of reduced gelatinase activity and the lack of proMMP-9 processing in macrophages. Our data provide evidence of the in vivo function of MMP-10 during endochondral ossification and defines the macrophages as the lead cell population in cartilage removal and vascular invasion. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | | | - Purificación Ripalda-Cemboráin
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Isabel A Calvo
- Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Borja Sáez
- Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | - Javier Aldazabal
- Tissue Engineering Group, TECNUN-Universidad de Navarra, San Sebastián, Spain
| | | | - Verónica Montiel
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Josune Orbe
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain
| | - José Antonio Rodríguez
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain
| | - José Antonio Páramo
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prósper
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Froilán Granero-Moltó
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
44
|
Park KR, Park JE, Kim B, Kwon IK, Hong JT, Yun HM. Calycosin-7-O-β-Glucoside Isolated from Astragalus membranaceus Promotes Osteogenesis and Mineralization in Human Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms222111362. [PMID: 34768792 PMCID: PMC8583672 DOI: 10.3390/ijms222111362] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/21/2022] Open
Abstract
Stem cells have received attention in various diseases, such as inflammatory, cancer, and bone diseases. Mesenchymal stem cells (MSCs) are multipotent stem cells that are critical for forming and repairing bone tissues. Herein, we isolated calycosin-7-O-β-glucoside (Caly) from the roots of Astragalus membranaceus, which is one of the most famous medicinal herbs, and investigated the osteogenic activities of Caly in MSCs. Caly did not affect cytotoxicity against MSCs, whereas Caly enhanced cell migration during the osteogenesis of MSCs. Caly increased the expression and enzymatic activities of ALP and the formation of mineralized nodules during the osteogenesis of MSCs. The osteogenesis and bone-forming activities of Caly are mediated by bone morphogenetic protein 2 (BMP2), phospho-Smad1/5/8, Wnt3a, phospho-GSK3β, and phospho-AKT, inducing the expression of runt-related transcription factor 2 (RUNX2). In addition, Caly-mediated osteogenesis and RUNX2 expression were attenuated by noggin and wortmannin. Moreover, the effects were validated in pre-osteoblasts committed to the osteoblast lineages from MSCs. Overall, our results provide novel evidence that Caly stimulates osteoblast lineage commitment of MSCs by triggering RUNX2 expression, suggesting Caly as a potential anabolic drug to prevent bone diseases.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea;
- Medical Device Research Center, Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Korea;
| | - Ji Eun Park
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea; (J.E.P.); (B.K.)
| | - Bomi Kim
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea; (J.E.P.); (B.K.)
| | - Il Keun Kwon
- Medical Device Research Center, Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Korea;
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si 28160, Korea
- Correspondence: (J.T.H.); (H.-M.Y.); Tel.: +82-02-961-0691 (H.-M.Y.); Fax: +82-02-960-1457 (H.-M.Y.)
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea;
- Correspondence: (J.T.H.); (H.-M.Y.); Tel.: +82-02-961-0691 (H.-M.Y.); Fax: +82-02-960-1457 (H.-M.Y.)
| |
Collapse
|
45
|
Ding Q, Liu H, Liu L, Ma C, Qin H, Wei Y, Ren Y. Deletion of p16 accelerates fracture healing in geriatric mice. Am J Transl Res 2021; 13:11107-11125. [PMID: 34786046 PMCID: PMC8581914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/05/2021] [Indexed: 06/13/2023]
Abstract
The biomarker p16 plays a role in aging and is upregulated in aged organs and cells, including bone marrow mesenchymal stem cells (BM-MSCs), which play a leading role in fracture healing. Several studies have reported delayed fracture healing in geriatric mice. However, the relationship between p16 expression and fracture healing in geriatric mice remains poorly understood. In this study, we found that fracture healing was accelerated in p16 deletion (p16-/-) mice, and the number of migrated BM-MSCs from p16-/- mice increased. The expressions of SDF-1 and CXCR4 were also upregulated in p16-/- mice. Increased cell percentage at S phase in cell cycle, enhanced expressions of CDK4/6, pRB, and E2F1, decreased expression of RB, and elevated expressions of SOX9, PCNA, and COL2A1 were detected in p16-/- mice. The expressions of COL10A1, MMP13, OSTERIX, and COL1A1 were also high in p16-/- mice. Moreover, the expressions of p-AKT, p-mTOR, HIF-1α, and VEGF-A in BM-MSCs and expression of VEGF-A in callus were upregulated in p16-/- mice. The expression of VEGF in the serum of p16-/- mice was also higher than that of wild type mice. Thus, deletion of p16 enhances migration, division, and differentiation of BM-MSCs, promotes proliferation and maturation of chondrocytes, activates osteoblastogenesis, and facilitates vascularization to accelerate fracture healing, providing a novel strategy to treat fracture in the elderly.
Collapse
Affiliation(s)
- Qirui Ding
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| | - Huan Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| | - Lijia Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| | - Cheng Ma
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| | - Haonan Qin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| | - Yifan Wei
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| | - Yongxin Ren
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing 210029, Jiangsu Province, P. R. China
| |
Collapse
|
46
|
Kirankumar S, Gurusamy N, Rajasingh S, Sigamani V, Vasanthan J, Perales SG, Rajasingh J. Modern approaches on stem cells and scaffolding technology for osteogenic differentiation and regeneration. Exp Biol Med (Maywood) 2021; 247:433-445. [PMID: 34648374 DOI: 10.1177/15353702211052927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The process of bone repair has always been a natural mystery. Although bones do repair themselves, supplemental treatment is required for the initiation of the self-regeneration process. Predominantly, surgical procedures are employed for bone regeneration. Recently, cell-based therapy for bone regeneration has proven to be more effective than traditional methods, as it eliminates the immune risk and painful surgeries. In clinical trials, various stem cells, especially mesenchymal stem cells, have shown to be more efficient for the treatment of several bone-related diseases, such as non-union fracture, osteogenesis imperfecta, osteosarcoma, and osteoporosis. Furthermore, the stem cells grown in a suitable three-dimensional scaffold support were found to be more efficient for osteogenesis. It has been shown that the three-dimensional bioscaffolds support and simulate an in vivo environment, which helps in differentiation of stem cells into bone cells. Bone regeneration in patients with bone disorders can be improved through modification of stem cells with several osteogenic factors or using stem cells as carriers for osteogenic factors. In this review, we focused on the various types of stem cells and scaffolds that are being used for bone regeneration. In addition, the molecular mechanisms of various transcription factors, signaling pathways that support bone regeneration and the senescence of the stem cells, which limits bone regeneration, have been discussed.
Collapse
Affiliation(s)
- Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Selene G Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
47
|
Potter ML, Smith K, Vyavahare S, Kumar S, Periyasamy-Thandavan S, Hamrick M, Isales CM, Hill WD, Fulzele S. Characterization of Differentially Expressed miRNAs by CXCL12/SDF-1 in Human Bone Marrow Stromal Cells. Biomol Concepts 2021; 12:132-143. [PMID: 34648701 DOI: 10.1515/bmc-2021-0015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 01/08/2023] Open
Abstract
Stromal cell-derived factor 1 (SDF-1) is known to influence bone marrow stromal cell (BMSC) migration, osteogenic differentiation, and fracture healing. We hypothesize that SDF-1 mediates some of its effects on BMSCs through epigenetic regulation, specifically via microRNAs (miRNAs). MiRNAs are small non-coding RNAs that target specific mRNA and prevent their translation. We performed global miRNA analysis and determined several miRNAs were differentially expressed in response to SDF-1 treatment. Gene Expression Omnibus (GEO) dataset analysis showed that these miRNAs play an important role in osteogenic differentiation and fracture healing. KEGG and GO analysis indicated that SDF-1 dependent miRNAs changes affect multiple cellular pathways, including fatty acid biosynthesis, thyroid hormone signaling, and mucin-type O-glycan biosynthesis pathways. Furthermore, bioinformatics analysis showed several miRNAs target genes related to stem cell migration and differentiation. This study's findings indicated that SDF-1 induces some of its effects on BMSCs function through miRNA regulation.
Collapse
Affiliation(s)
| | - Kathryn Smith
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA
| | - Sagar Vyavahare
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA
| | - Sandeep Kumar
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA
| | | | - Mark Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA.,Institute of Healthy Aging, Augusta University, Augusta, GA
| | - Carlos M Isales
- Institute of Healthy Aging, Augusta University, Augusta, GA.,Departments of Medicine, Augusta University, Augusta, GA
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403.,Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA.,Institute of Healthy Aging, Augusta University, Augusta, GA.,Departments of Medicine, Augusta University, Augusta, GA.,Department of Orthopedics, Augusta University, Augusta, GA
| |
Collapse
|
48
|
Bone marrow CD73 + mesenchymal stem cells display increased stemness in vitro and promote fracture healing in vivo. Bone Rep 2021; 15:101133. [PMID: 34632004 PMCID: PMC8493579 DOI: 10.1016/j.bonr.2021.101133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent and considered to be of great potential for regenerative medicine. We could show recently (Breitbach, Kimura et al. 2018) that a subpopulation of MSCs as well as sinusoidal endothelial cells (sECs) in the bone marrow (BM) of CD73-EGFP reporter mice could be labeled in vivo. We took advantage of this model to explore the plasticity and osteogenic potential of CD73-EGFP+ MSCs in vitro and their role in the regenerative response upon bone lesion in vivo. Herein we show that isolated CD73-EGFP+ MSCs displayed more pronounced stemness and stronger in vitro differentiation capacity into the osteogenic lineage compared to CD73-EGFP− MSCs. In a bone fracture model, endogenous BM-resident CD73-EGFP+ MSCs were found to migrate to the fracture site and differentiate into cartilage and bone cells. Our analysis also showed that CD73-EGFP+ sECs contributed to the neovascularization of the fracture site. In addition, grafting of CD73-EGFP+ MSCs into acute bone lesions revealed their capacity to differentiate into chondrocytes and osteocytes in vivo and their contribution to callus formation in the regeneration process of fracture healing. Thus, CD73+ MSCs display enhanced stemness and osteogenic differentiation potential in vitro and in vivo illustrating a prominent role of the CD73+ MSC subpopulation to promote fracture repair. CD73+ mesenchymal stem cells (MSCs) display pronounced proliferation potential. CD73+ MSCs show high in vitro osteogenic differentiation capacity. Endogenous bone marrow-resident CD73+ MSCs contribute to fracture healing. Grafted CD73+ MSCs promote bone formation upon fracture repair.
Collapse
|
49
|
Venkataiah VS, Yahata Y, Kitagawa A, Inagaki M, Kakiuchi Y, Nakano M, Suzuki S, Handa K, Saito M. Clinical Applications of Cell-Scaffold Constructs for Bone Regeneration Therapy. Cells 2021; 10:2687. [PMID: 34685667 PMCID: PMC8534498 DOI: 10.3390/cells10102687] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/20/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Bone tissue engineering (BTE) is a process of combining live osteoblast progenitors with a biocompatible scaffold to produce a biological substitute that can integrate into host bone tissue and recover its function. Mesenchymal stem cells (MSCs) are the most researched post-natal stem cells because they have self-renewal properties and a multi-differentiation capacity that can give rise to various cell lineages, including osteoblasts. BTE technology utilizes a combination of MSCs and biodegradable scaffold material, which provides a suitable environment for functional bone recovery and has been developed as a therapeutic approach to bone regeneration. Although prior clinical trials of BTE approaches have shown promising results, the regeneration of large bone defects is still an unmet medical need in patients that have suffered a significant loss of bone function. In this present review, we discuss the osteogenic potential of MSCs in bone tissue engineering and propose the use of immature osteoblasts, which can differentiate into osteoblasts upon transplantation, as an alternative cell source for regeneration in large bone defects.
Collapse
Affiliation(s)
- Venkata Suresh Venkataiah
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Yoshio Yahata
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Akira Kitagawa
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
- OsteRenatos Ltd., Sendai Capital Tower 2F, 4-10-3 Central, Aoba-ku, Sendai 980-0021, Japan
| | - Masahiko Inagaki
- National Institute of Advanced Industrial Science and Technology, 2266-98 Anagahora, Nagoya 463-8560, Japan;
| | - Yusuke Kakiuchi
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Masato Nakano
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Shigeto Suzuki
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Keisuke Handa
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
- Department of Oral Science, Division of Oral Biochemistry, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Masahiro Saito
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
- OsteRenatos Ltd., Sendai Capital Tower 2F, 4-10-3 Central, Aoba-ku, Sendai 980-0021, Japan
| |
Collapse
|
50
|
Neumeister MW, Winters JN, Maduakolum E. Phalangeal and Metacarpal Fractures of the Hand: Preventing Stiffness. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2021; 9:e3871. [PMID: 34729287 PMCID: PMC8553242 DOI: 10.1097/gox.0000000000003871] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 11/26/2022]
Abstract
Fractures of the phalangeal or metacarpal bones of the hand are common. Many of these fractures are treated without surgery. However, both conservative and operative management of fractures of the hand can result in stiffness. Stiffness is the most common complication in the management of hand fractures. The key to preventing stiffness is early range of motion exercises. This article challenges many of the current treatment regimens offered to patients with the so-called unstable fractures. The evaluation of the patients' function is the primary factor that should determine the course of conservation versus operative management. X-rays do not demonstrate function and therefore act as an adjunct only to the care of the patient. The goal of treating hand fractures is to restore function. Early motion may not only improve healing but may also hasten the return to normal hand function. The tenets of how to prevent stiffness are described in this review.
Collapse
Affiliation(s)
- Michael W. Neumeister
- From the Institute for Plastic Surgery, Southern Illinois University School of Medicine, Springfield, Illinois
| | - James N. Winters
- Institute for Plastic Surgery, Southern Illinois University School of Medicine, Springfield, Illinois
| | | |
Collapse
|