1
|
Heo S, Noh M, Kim Y, Park S. Stem Cell-Laden Engineered Patch: Advances and Applications in Tissue Regeneration. ACS APPLIED BIO MATERIALS 2025; 8:62-87. [PMID: 39701826 DOI: 10.1021/acsabm.4c01427] [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] [Indexed: 12/21/2024]
Abstract
Stem cell-based therapies are emerging as significant approaches in tissue engineering and regenerative medicine, applicable to both fundamental scientific research and clinical practice. Despite remarkable results in clinical studies, challenges such as poor standardization of graft tissues, limited sources, and reduced functionality have hindered the effectiveness of these therapies. In this review, we summarize the engineering approaches involved in fabricating stem cell assisted patches and the substantial strategies for designing stem cell-laden engineered patches (SCP) to complement the existing stem cell-based therapies. We then outline the potential applications of SCP in advancing tissue regeneration and regenerative medicine. By combining living stem cells with engineered patches, SCP can enhance the functions of both components, particularly for tissue engineering applications. Finally, we addressed current challenges, such as ethical considerations, high costs, and regulatory hurdles and proposed future research directions to overcome these barriers.
Collapse
Affiliation(s)
- Seyeong Heo
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Minhyeok Noh
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Yeonseo Kim
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Sunho Park
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
2
|
Mohanto N, Mondal H, Park YJ, Jee JP. Therapeutic delivery of oxygen using artificial oxygen carriers demonstrates the possibility of treating a wide range of diseases. J Nanobiotechnology 2025; 23:25. [PMID: 39827150 PMCID: PMC11742488 DOI: 10.1186/s12951-024-03060-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Artificial oxygen carriers have emerged as potential substitutes for red blood cells in situations of major blood loss, including accidents, surgical procedures, trauma, childbirth, stomach ulcers, hemorrhagic shock, and blood vessel ruptures which can lead to sudden reduction in blood volume. The therapeutic delivery of oxygen utilizing artificial oxygen carriers as red blood cell substitutes presents a promising avenue for treating a spectrum of disease models. Apart from that, the recent advancement of artificial oxygen carriers intended to supplant conventional blood transfusions draws significant attention due to the exigencies of warfare and the ongoing challenges posed by the COVID-19 pandemic. However, there is a pressing need to formulate stable, non-toxic, and immunologically inert oxygen carriers. Even though numerous challenges are encountered in the development of artificial oxygen carriers, their applicability extends to various medical treatments, encompassing elective and cardiovascular surgeries, hemorrhagic shock, decompression illness, acute stroke, myocardial infarction, sickle cell crisis, and proficient addressing conditions such as cerebral hypoxia. Therefore, this paper provides an overview of therapeutic oxygen delivery using assorted types of artificial oxygen carriers, including hemoglobin-based, perfluorocarbon-based, stem cell-derived, and oxygen micro/nanobubbles, in the treatment of diverse disease models. Additionally, it discusses the potential side effects and limitations associated with these interventions, while incorporating completed and ongoing research and recent clinical developments. Finally, the prospective solutions and general demands of the perfect artificial oxygen carriers were anticipated to be a reference for subsequent research endeavors.
Collapse
Affiliation(s)
- Nijaya Mohanto
- College of Pharmacy, Chosun University, 309 Pilmun-Daero, Dong-Gu, Gwangju, 61452, Republic of Korea
| | - Himangsu Mondal
- College of Pharmacy, Chosun University, 309 Pilmun-Daero, Dong-Gu, Gwangju, 61452, Republic of Korea
| | - Young-Joon Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi, Republic of Korea
| | - Jun-Pil Jee
- College of Pharmacy, Chosun University, 309 Pilmun-Daero, Dong-Gu, Gwangju, 61452, Republic of Korea.
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju, Republic of Korea.
| |
Collapse
|
3
|
Yang Y, Wang W, Zeng Q, Wang N, Li W, Chen B, Guan Q, Li C, Li W. Fabricating oxygen self-supplying 3D printed bioactive hydrogel scaffold for augmented vascularized bone regeneration. Bioact Mater 2024; 40:227-243. [PMID: 38973993 PMCID: PMC11226730 DOI: 10.1016/j.bioactmat.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Limited cells and factors, inadequate mechanical properties, and necrosis of defects center have hindered the wide clinical application of bone-tissue engineering scaffolds. Herein, we construct a self-oxygenated 3D printed bioactive hydrogel scaffold by integrating oxygen-generating nanoparticles and hybrid double network hydrogel structure. The hydrogel scaffold possesses the characteristics of extracellular matrix; Meanwhile, the fabricated hybrid double network structure by polyacrylamide and CaCl2-crosslinked sodium carboxymethylcellulose endows the hydrogel favorable compressive strength and 3D printability. Furthermore, the O2 generated by CaO2 nanoparticles encapsulated in ZIF-8 releases steadily and sustainably because of the well-developed microporous structure of ZIF-8, which can significantly promote cell viability and proliferation in vitro, as well as angiogenesis and osteogenic differentiation with the assistance of Zn2+. More significantly, the synergy of O2 and 3D printed pore structure can prevent necrosis of defects center and facilitate cell infiltration by providing cells the nutrients and space they need, which can further induce vascular network ingrowth and accelerate bone regeneration in all areas of the defect in vivo. Overall, this work provides a new avenue for preparing cell/factor-free bone-tissue engineered scaffolds that possess great potential for tissue regeneration and clinical alternative.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Wanmeng Wang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Qianrui Zeng
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Ning Wang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Wenbo Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Bo Chen
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Qingxin Guan
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Changyi Li
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Wei Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
4
|
Zhao J, Sarkar N, Ren Y, Pathak AP, Grayson WL. Engineering next-generation oxygen-generating scaffolds to enhance bone regeneration. Trends Biotechnol 2024:S0167-7799(24)00250-6. [PMID: 39343620 DOI: 10.1016/j.tibtech.2024.09.006] [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: 03/14/2024] [Revised: 08/08/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
In bone, an adequate oxygen (O2) supply is crucial during development, homeostasis, and healing. Oxygen-generating scaffolds (OGS) have demonstrated significant potential to enhance bone regeneration. However, the complexity of O2 delivery and signaling in vivo makes it challenging to tailor the design of OGS to precisely meet this biological requirement. We review recent advances in OGS and analyze persisting engineering and translational hurdles. We also discuss the potential of computational and machine learning (ML) models to facilitate the integration of novel imaging data with biological readouts and advanced biomanufacturing technologies. By elucidating how to tackle current challenges using cutting-edge technologies, we provide insights for transitioning from traditional to next-generation OGS to improve bone regeneration in patients.
Collapse
Affiliation(s)
- Jingtong Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Naboneeta Sarkar
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Yunke Ren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Bal Z, Takakura N. Hydrogel Use in Osteonecrosis of the Femoral Head. Gels 2024; 10:544. [PMID: 39195073 DOI: 10.3390/gels10080544] [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: 06/10/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/29/2024] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a vascular disease of unknown etiology and can be categorized mainly into two types: non-traumatic and traumatic ONFH. Thus, understanding osteogenic-angiogenic coupling is of prime importance in finding a solution for the treatment of ONFH. Hydrogels are biomaterials that are similar to the extracellular matrix (ECM). As they are able to mimic real tissue, they meet one of the most important rules in tissue engineering. In ONFH studies, hydrogels have recently become popular because of their ability to retain water and their adjustable properties, injectability, and mimicry of natural ECM. Because bone regeneration and graft materials are very broad areas of research and ONFH is a complex situation including bone and vascular systems, and there is no settled treatment strategy for ONFH worldwide, in this review paper, we followed a top-down approach by reviewing (1) bone and bone grafting, (2) hydrogels, (3) vascular systems, and (4) ONFH and hydrogel use in ONFH with studies in the literature which show promising results in limited clinical studies. The aim of this review paper is to provide the reader with general information on every aspect of ONFH and to focus on the hydrogel used in ONFH.
Collapse
Affiliation(s)
- Zeynep Bal
- Laboratory of Signal Transduction, WPI Immunology Frontier Research Center (WPI-IFReC), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
- Department of Signal Transduction, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
| | - Nobuyuki Takakura
- Laboratory of Signal Transduction, WPI Immunology Frontier Research Center (WPI-IFReC), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
- Department of Signal Transduction, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
6
|
Ghaffari-Bohlouli P, Jafari H, Okoro OV, Alimoradi H, Nie L, Jiang G, Kakkar A, Shavandi A. Gas Therapy: Generating, Delivery, and Biomedical Applications. SMALL METHODS 2024; 8:e2301349. [PMID: 38193272 DOI: 10.1002/smtd.202301349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Oxygen (O2), nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and hydrogen (H2) with direct effects, and carbon dioxide (CO2) with complementary effects on the condition of various diseases are known as therapeutic gases. The targeted delivery and in situ generation of these therapeutic gases with controllable release at the site of disease has attracted attention to avoid the risk of gas poisoning and improve their performance in treating various diseases such as cancer therapy, cardiovascular therapy, bone tissue engineering, and wound healing. Stimuli-responsive gas-generating sources and delivery systems based on biomaterials that enable on-demand and controllable release are promising approaches for precise gas therapy. This work highlights current advances in the design and development of new approaches and systems to generate and deliver therapeutic gases at the site of disease with on-demand release behavior. The performance of the delivered gases in various biomedical applications is then discussed.
Collapse
Affiliation(s)
- Pejman Ghaffari-Bohlouli
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Hafez Jafari
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Oseweuba Valentine Okoro
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Houman Alimoradi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Lei Nie
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| |
Collapse
|
7
|
Zhu Y, Wang X, Liu R. Bioinformatics proved the existence of potential hub genes activating autophagy to participate in cartilage degeneration in osteonecrosis of the femoral head. J Mol Histol 2024; 55:539-554. [PMID: 38758521 DOI: 10.1007/s10735-024-10200-w] [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: 12/30/2023] [Accepted: 05/01/2024] [Indexed: 05/18/2024]
Abstract
The obvious degeneration of articular cartilage occurs in the late stage of osteonecrosis of the femoral head (ONFH), which aggravates the condition of ONFH. This study aimed to demonstrate aberrant activation of autophagy processes in ONFH chondrocytes through bioinformatics and to predict and identify relevant hub genes and pathways. Differentially expressed genes (DEGs) were identified using R software in the GSE74089 dataset from the GEO database. DEGs were crossed with the Human Autophagy Database (HADb) autophagy genes to screen out autophagy-related differential genes (AT-DEGs). GSEA, GSVA, GO, and KEGG pathway enrichment analyses of AT-DEGs were performed. The STRING database was used to analyze the protein-protein interaction (PPI) of the AT-DEGs network, and the MCODE and CytoHubba plugin in the Cytoscape software was used to analyze the key gene cluster module and screen the hub genes. The PPI network of hub genes was constructed using the GeneMANIA database, and functional enrichment and gene connectivity categories were analyzed. The expression levels of hub genes of related genes in the ONFH patients were verified in the dataset GSE123568, and the protein expression was verified by immunohistochemistry in tissues. The analysis of DEGs revealed abnormal autophagy in ONFH cartilage. AT-DEGs in ONFH have special enrichment in macroautophagy, autophagosome membrane, and phosphatidylinositol-3-phosphate binding. In the GSE123568 dataset, it was also found that ATG2B, ATG4B, and UVRAG were all significantly upregulated in ONFH patients. By immunohistochemistry, it was verified that ATG2B, ATG4B, and UVRAG were significantly overexpressed. These three genes regulate the occurrence and extension of autophagosomes through the PI3KC3C pathway. Finally, we determined that chondrocytes in ONFH undergo positive regulation of autophagy through the corresponding pathways involved in three genes: ATG2B, ATG4B, and UVRAG.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710004, China
| | - Xianxuan Wang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
8
|
Li X, Li L, Wang D, Zhang J, Yi K, Su Y, Luo J, Deng X, Deng F. Fabrication of polymeric microspheres for biomedical applications. MATERIALS HORIZONS 2024; 11:2820-2855. [PMID: 38567423 DOI: 10.1039/d3mh01641b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Polymeric microspheres (PMs) have attracted great attention in the field of biomedicine in the last several decades due to their small particle size, special functionalities shown on the surface and high surface-to-volume ratio. However, how to fabricate PMs which can meet the clinical needs and transform laboratory achievements to industrial scale-up still remains a challenge. Therefore, advanced fabrication technologies are pursued. In this review, we summarize the technologies used to fabricate PMs, including emulsion-based methods, microfluidics, spray drying, coacervation, supercritical fluid and superhydrophobic surface-mediated method and their advantages and disadvantages. We also review the different structures, properties and functions of the PMs and their applications in the fields of drug delivery, cell encapsulation and expansion, scaffolds in tissue engineering, transcatheter arterial embolization and artificial cells. Moreover, we discuss existing challenges and future perspectives for advancing fabrication technologies and biomedical applications of PMs.
Collapse
Affiliation(s)
- Xuebing Li
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China.
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Luohuizi Li
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China.
| | - Dehui Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China.
| | - Jun Zhang
- Shandong Pharmaceutical Glass Co. Ltd, Zibo, 256100, P. R. China
| | - Kangfeng Yi
- Shandong Pharmaceutical Glass Co. Ltd, Zibo, 256100, P. R. China
| | - Yucai Su
- Shandong Pharmaceutical Glass Co. Ltd, Zibo, 256100, P. R. China
| | - Jing Luo
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China.
| | - Xu Deng
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China.
- Shenzhen Institute for Advanced Study, University of Electronic Science and Technology of China, Shenzhen, 518110, P. R. China
| | - Fei Deng
- Department of Nephrology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
- Department of Nephrology, Sichuan Provincial People's Hospital Jinniu Hospital, Chengdu Jinniu District People's Hospital, Chengdu 610054, P. R. China.
| |
Collapse
|
9
|
Xie Z, Luo H, Wang T, Wang L, Zhang J, Dong W, Liu G, Li F, Kang Q, Zhu X, Zhang F, Peng W. METTL3 inhibits BMSC apoptosis and facilitates osteonecrosis repair via an m6A-IGF2BP2-dependent mechanism. Heliyon 2024; 10:e30195. [PMID: 38784565 PMCID: PMC11112270 DOI: 10.1016/j.heliyon.2024.e30195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Hypoxia-induced apoptosis of bone marrow mesenchymal stem cells (BMSCs) limits the efficacy of their transplantation for steroid-induced osteonecrosis of the femoral head (SONFH). As apoptosis and RNA methylation are closely related, exploring the role and mechanism of RNA methylation in hypoxic apoptosis of BMSCs is expected to identify new targets for transplantation of BMSCs for SONFH and enhance transplantation efficacy. We performed methylated RNA immunoprecipitation sequencing (MeRIP-seq) combined with RNA-seq on a hypoxia-induced apoptosis BMSC model and found that the RNA methyltransferase-like 3 (METTL3) is involved in hypoxia-induced BMSC apoptosis. The expression of METTL3 was downregulated in BMSCs after hypoxia and in BMSCs implanted in osteonecrosis areas. Knockdown of METLL3 under normoxic conditions promoted apoptosis of BMSCs. In contrast, overexpression of METTL3 promoted the survival of BMSCs under hypoxic conditions, and overexpression of METTL3 promoted the survival of BMSCs in the osteonecrosis area and the repair of the osteonecrosis area. Regarding the mechanism, the m6A levels of the mRNAs of anti-apoptotic genes Bcl-2, Mcl-1, and BIRC5 were significantly increased upon the overexpression of METTL3 under hypoxic conditions, which promoted the binding of Bcl-2, Mcl-1, and BIRC5 mRNAs to IGF2BP2, enhanced the mRNA stability, and increased the protein expression of the three anti-apoptotic genes. In conclusion, overexpression of METTL3 promoted m6A modification of mRNAs of Bcl-2, Mcl-1, and BIRC5, promoted the binding of IGF2BP2 to the above-mentioned mRNAs, enhanced mRNA stability, inhibited hypoxia-induced BMSC apoptosis, and promoted repair of SONFH, thereby providing novel targets for transplantation of BMSCs for SONFH.
Collapse
Affiliation(s)
- Zhihong Xie
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Hong Luo
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Tao Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Lei Wang
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Jian Zhang
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Wentao Dong
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Gang Liu
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Fanchao Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Qinglin Kang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xuesong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Fei Zhang
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Wuxun Peng
- Department of Emergency Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| |
Collapse
|
10
|
Norberg AE, Bakirci E, Lim KS, Dalton PD, Woodfield TBF, Lindberg GCJ. Bioassembly of hemoglobin-loaded photopolymerizable spheroids alleviates hypoxia-induced cell death. Biofabrication 2024; 16:025026. [PMID: 38373325 DOI: 10.1088/1758-5090/ad2a7d] [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: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 02/21/2024]
Abstract
The delivery of oxygen within tissue engineered constructs is essential for cell survivability; however, achieving this within larger biofabricated constructs poses a significant challenge. Efforts to overcome this limitation often involve the delivery of synthetic oxygen generating compounds. The application of some of these compounds is problematic for the biofabrication of living tissues due to inherent issues such as cytotoxicity, hyperoxia and limited structural stability due to oxygen inhibition of radical-based crosslinking processes. This study aims to develop an oxygen delivering system relying on natural-derived components which are cytocompatible, allow for photopolymerization and advanced biofabrication processes, and improve cell survivability under hypoxia (1% O2). We explore the binding of human hemoglobin (Hb) as a natural oxygen deposit within photopolymerizable allylated gelatin (GelAGE) hydrogels through the spontaneous complex formation of Hb with negatively charged biomolecules (heparin, hyaluronic acid, and bovine serum albumin). We systematically study the effect of biomolecule inclusion on cytotoxicity, hydrogel network properties, Hb incorporation efficiency, oxygen carrying capacity, cell viability, and compatibility with 3D-bioassembly processes within melt electrowritten (MEW) scaffolds. All biomolecules were successfully incorporated within GelAGE hydrogels, displaying controllable mechanical properties and cytocompatibility. Results demonstrated efficient and tailorable Hb incorporation within GelAGE-Heparin hydrogels. The developed system was compatible with microfluidics and photopolymerization processes, allowing for the production of GelAGE-Heparin-Hb spheres. Hb-loaded spheres were assembled into MEW polycaprolactone scaffolds, significantly increasing the local oxygen levels. Ultimately, cells within Hb-loaded constructs demonstrated good cell survivability under hypoxia. Taken together, we successfully developed a hydrogel system that retains Hb as a natural oxygen deposit post-photopolymerization, protecting Hb from free-radical oxidation while remaining compatible with biofabrication of large constructs. The developed GelAGE-Heparin-Hb system allows for physoxic oxygen delivery and thus possesses a vast potential for use across broad tissue engineering and biofabrication strategies to help eliminate cell death due to hypoxia.
Collapse
Affiliation(s)
- Axel E Norberg
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Khoon S Lim
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Paul D Dalton
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Tim B F Woodfield
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Gabriella C J Lindberg
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| |
Collapse
|
11
|
Gao Y, You Y, Zhang P, Yu Y, Xu Z, Wei H, Liu Z, Yu R, Jin G, Wang H, Zhang S, Li Y, Li W. Cortistatin prevents glucocorticoid-associated osteonecrosis of the femoral head via the GHSR1a/Akt pathway. Commun Biol 2024; 7:132. [PMID: 38278996 PMCID: PMC10817896 DOI: 10.1038/s42003-024-05795-5] [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: 01/15/2023] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
Long-term use of glucocorticoids (GCs) is known to be a predominant cause of osteonecrosis of the femoral head (ONFH). Moreover, GCs can mediate apoptosis of various cell types by exaggerating oxidative stress. We have previously found that Cortistatin (CST) antagonizes oxidative stress and improves cell apoptosis in several conditions. In this study, we detected that the CST expression levels were diminished in patients with ONFH compared with femoral neck fracture (FNF). In addition, a GC-induced rat ONFH model was established, which impaired bone quality in the femoral head. Then, administration of CST attenuated these ONFH phenotypes. Furthermore, osteoblast and endothelial cells were cultured and stimulated with dexamethasone (Dex) in the presence or absence of recombinant CST. As a result, Dex induced impaired anabolic metabolism of osteoblasts and suppressed tube formation in endothelial cells, while additional treatment with CST reversed this damage to the cells. Moreover, blocking GHSR1a, a well-accepted receptor of CST, or blocking the AKT signaling pathway largely abolished the protective function of CST in Dex-induced disorder of the cells. Taken together, we indicate that CST has the capability to prevent GC-induced apoptosis and metabolic disorder of osteoblasts in the pathogenesis of ONFH via the GHSR1a/AKT signaling pathway.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Yunhao You
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pengfei Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Yu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaoning Xu
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Wei
- Department of Rehabilitation, Qilu Hospital of Shandong University, Jinan, China
| | - Zhicheng Liu
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Ruixuan Yu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gaoxin Jin
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Hao Wang
- Department of Trauma Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Shuai Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China.
| | - Yuhua Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China.
| | - Weiwei Li
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
12
|
Zhao J, Zhou C, Xiao Y, Zhang K, Zhang Q, Xia L, Jiang B, Jiang C, Ming W, Zhang H, Long H, Liang W. Oxygen generating biomaterials at the forefront of regenerative medicine: advances in bone regeneration. Front Bioeng Biotechnol 2024; 12:1292171. [PMID: 38282892 PMCID: PMC10811251 DOI: 10.3389/fbioe.2024.1292171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Globally, an annual count of more than two million bone transplants is conducted, with conventional treatments, including metallic implants and bone grafts, exhibiting certain limitations. In recent years, there have been significant advancements in the field of bone regeneration. Oxygen tension regulates cellular behavior, which in turn affects tissue regeneration through metabolic programming. Biomaterials with oxygen release capabilities enhance therapeutic effectiveness and reduce tissue damage from hypoxia. However, precise control over oxygen release is a significant technical challenge, despite its potential to support cellular viability and differentiation. The matrices often used to repair large-size bone defects do not supply enough oxygen to the stem cells being used in the regeneration process. Hypoxia-induced necrosis primarily occurs in the central regions of large matrices due to inadequate provision of oxygen and nutrients by the surrounding vasculature of the host tissues. Oxygen generating biomaterials (OGBs) are becoming increasingly significant in enhancing our capacity to facilitate the bone regeneration, thereby addressing the challenges posed by hypoxia or inadequate vascularization. Herein, we discussed the key role of oxygen in bone regeneration, various oxygen source materials and their mechanism of oxygen release, the fabrication techniques employed for oxygen-releasing matrices, and novel emerging approaches for oxygen delivery that hold promise for their potential application in the field of bone regeneration.
Collapse
Affiliation(s)
- Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, China
| | - Yang Xiao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Kunyan Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Qiang Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bo Jiang
- Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chanyi Jiang
- Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenyi Ming
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengjian Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
13
|
Tsubosaka M, Maruyama M, Lui E, Kushioka J, Toya M, Gao Q, Shen H, Li X, Chow SKH, Zhang N, Yang YP, Goodman SB. Preclinical models for studying corticosteroid-induced osteonecrosis of the femoral head. J Biomed Mater Res B Appl Biomater 2024; 112:e35360. [PMID: 38247252 DOI: 10.1002/jbm.b.35360] [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: 02/23/2023] [Revised: 11/02/2023] [Accepted: 11/27/2023] [Indexed: 01/23/2024]
Abstract
Nontraumatic osteonecrosis of the femoral head (ONFH) is a refractory condition that commonly results in femoral head collapse and degenerative arthritis of the hip. In the early stages, surgical procedures for hip preservation, including core decompression (CD), have been developed to prevent progressive collapse of the femoral head. Optimization of bone regeneration and biological augmentation may further enhance the therapeutic efficacy of CD for ONFH. Thus, combining CD with cell-based therapy has recently been proposed. In fact, patients treated with cell-based therapy using autologous bone marrow concentrate demonstrate improved survivorship of the femoral head, compared with conventional CD alone. Preclinical research studies to investigate adjunctive therapies for CD often utilize the rabbit model of corticosteroid-induced ONFH. Mesenchymal stem cells (MSCs) are known to promote osteogenesis and angiogenesis, and decrease inflammation in bone. Local drug delivery systems have the potential to achieve targeted therapeutic effects by precisely controlling the drug release rate. Scaffolds can provide an osteoconductive structural framework to facilitate the repair of osteonecrotic bone tissue. We focused on the combination of both cell-based and scaffold-based therapies for bone tissue regeneration in ONFH. We hypothesized that combining CD and osteoconductive scaffolds would provide mechanical strength and structural cell guidance; and that combining CD and genetically modified (GM) MSCs to express relevant cytokines, chemokines, and growth factors would promote bone tissue repair. We developed GM MSCs that overexpress the anti-inflammatory, pro-reconstructive cytokines platelet-derived growth factor-BB to provide MSCs with additional benefits and investigated the efficacy of combinations of these GM MSCs and scaffolds for treatment of ONFH in skeletally mature male New Zealand white rabbits. In the future, the long-term safety, efficacy, durability, and cost-effectiveness of these and other biological and mechanical treatments must be demonstrated for the patients affected by ONFH.
Collapse
Affiliation(s)
- Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
14
|
Pan Q, Su W, Yao Y. Progress in microsphere-based scaffolds in bone/cartilage tissue engineering. Biomed Mater 2023; 18:062004. [PMID: 37751762 DOI: 10.1088/1748-605x/acfd78] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Bone/cartilage repair and regeneration have been popular and difficult issues in medical research. Tissue engineering is rapidly evolving to provide new solutions to this problem, and the key point is to design the appropriate scaffold biomaterial. In recent years, microsphere-based scaffolds have been considered suitable scaffold materials for bone/cartilage injury repair because microporous structures can form more internal space for better cell proliferation and other cellular activities, and these composite scaffolds can provide physical/chemical signals for neotissue formation with higher efficiency. This paper reviews the research progress of microsphere-based scaffolds in bone/chondral tissue engineering, briefly introduces types of microspheres made from polymer, inorganic and composite materials, discusses the preparation methods of microspheres and the exploration of suitable microsphere pore size in bone and cartilage tissue engineering, and finally details the application of microsphere-based scaffolds in biomimetic scaffolds, cell proliferation and drug delivery systems.
Collapse
Affiliation(s)
- Qian Pan
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Weixian Su
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Yongchang Yao
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| |
Collapse
|
15
|
Balaji S, Short WD, Padon BW, Belgodere JA, Jimenez SE, Deoli NT, Guidry AC, Green JC, Prajapati TJ, Farouk F, Kaul A, Son D, Jung OS, Astete CE, Kim M, Jung JP. Injectable Antioxidant and Oxygen-Releasing Lignin Composites to Promote Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:18639-18652. [PMID: 37022100 PMCID: PMC10119855 DOI: 10.1021/acsami.2c22982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/27/2023] [Indexed: 06/19/2023]
Abstract
The application of engineered biomaterials for wound healing has been pursued since the beginning of tissue engineering. Here, we attempt to apply functionalized lignin to confer antioxidation to the extracellular microenvironments of wounds and to deliver oxygen from the dissociation of calcium peroxide for enhanced vascularization and healing responses without eliciting inflammatory responses. Elemental analysis showed 17 times higher quantity of calcium in the oxygen-releasing nanoparticles. Lignin composites including the oxygen-generating nanoparticles released around 700 ppm oxygen per day at least for 7 days. By modulating the concentration of the methacrylated gelatin, we were able to maintain the injectability of lignin composite precursors and the stiffness of lignin composites suitable for wound healing after photo-cross-linking. In situ formation of lignin composites with the oxygen-releasing nanoparticles enhanced the rate of tissue granulation, the formation of blood vessels, and the infiltration of α-smooth muscle actin+ fibroblasts into the wounds over 7 days. At 28 days after surgery, the lignin composite with oxygen-generating nanoparticles remodeled the collagen architecture, resembling the basket-weave pattern of unwounded collagen with minimal scar formation. Thus, our study shows the potential of functionalized lignin for wound-healing applications requiring balanced antioxidation and controlled release of oxygen for enhanced tissue granulation, vascularization, and maturation of collagen.
Collapse
Affiliation(s)
- Swathi Balaji
- Division
of Pediatric Surgery, Department of Surgery, Texas Children’s
Hospital and Baylor College of Medicine, Feigin Center at Texas Children’s Hospital, 1102 Bates Ave, C.450.05, Houston, Texas 77030, United States of America
| | - Walker D. Short
- Division
of Pediatric Surgery, Department of Surgery, Texas Children’s
Hospital and Baylor College of Medicine, Feigin Center at Texas Children’s Hospital, 1102 Bates Ave, C.450.05, Houston, Texas 77030, United States of America
| | - Benjamin W. Padon
- Division
of Pediatric Surgery, Department of Surgery, Texas Children’s
Hospital and Baylor College of Medicine, Feigin Center at Texas Children’s Hospital, 1102 Bates Ave, C.450.05, Houston, Texas 77030, United States of America
| | - Jorge A. Belgodere
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| | - Sarah E. Jimenez
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| | - Naresh T. Deoli
- Louisiana
Accelerator Center, University of Louisiana
at Lafayette, 20 Cajundome Boulevard, Lafayette, Louisiana 70506, United States of America
| | - Anna C. Guidry
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| | - Justin C. Green
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| | - Tanuj J. Prajapati
- Division
of Pediatric Surgery, Department of Surgery, Texas Children’s
Hospital and Baylor College of Medicine, Feigin Center at Texas Children’s Hospital, 1102 Bates Ave, C.450.05, Houston, Texas 77030, United States of America
| | - Fayiz Farouk
- Division
of Pediatric Surgery, Department of Surgery, Texas Children’s
Hospital and Baylor College of Medicine, Feigin Center at Texas Children’s Hospital, 1102 Bates Ave, C.450.05, Houston, Texas 77030, United States of America
| | - Aditya Kaul
- Division
of Pediatric Surgery, Department of Surgery, Texas Children’s
Hospital and Baylor College of Medicine, Feigin Center at Texas Children’s Hospital, 1102 Bates Ave, C.450.05, Houston, Texas 77030, United States of America
| | - Dongwan Son
- Department
of Chemistry and Chemical Engineering, Inha
University, Incheon 22212, Republic of Korea
| | - Olivia S. Jung
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| | - Carlos E. Astete
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| | - Myungwoong Kim
- Department
of Chemistry and Chemical Engineering, Inha
University, Incheon 22212, Republic of Korea
| | - Jangwook P. Jung
- Department
of Biological Engineering, Louisiana State
University, 149 E.B. Doran Hall, Baton Rouge, Louisiana 70803, United States of America
| |
Collapse
|
16
|
Wang T, Xie ZH, Wang L, Luo H, Zhang J, Dong WT, Zheng XH, Ye C, Tian XB, Liu G, Zhu XS, Li YL, Kang QL, Zhang F, Peng WX. LncAABR07053481 inhibits bone marrow mesenchymal stem cell apoptosis and promotes repair following steroid-induced avascular necrosis. Commun Biol 2023; 6:365. [PMID: 37012358 PMCID: PMC10070412 DOI: 10.1038/s42003-023-04661-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 03/03/2023] [Indexed: 04/05/2023] Open
Abstract
The osteonecrotic area of steroid-induced avascular necrosis of the femoral head (SANFH) is a hypoxic microenvironment that leads to apoptosis of transplanted bone marrow mesenchymal stem cells (BMSCs). However, the underlying mechanism remains unclear. Here, we explore the mechanism of hypoxic-induced apoptosis of BMSCs, and use the mechanism to improve the transplantation efficacy of BMSCs. Our results show that the long non-coding RNA AABR07053481 (LncAABR07053481) is downregulated in BMSCs and closely related to the degree of hypoxia. Overexpression of LncAABR07053481 could increase the survival rate of BMSCs. Further exploration of the downstream target gene indicates that LncAABR07053481 acts as a molecular "sponge" of miR-664-2-5p to relieve the silencing effect of miR-664-2-5p on the target gene Notch1. Importantly, the survival rate of BMSCs overexpressing LncAABR07053481 is significantly improved after transplantation, and the repair effect of BMSCs in the osteonecrotic area is also improved. This study reveal the mechanism by which LncAABR07053481 inhibits hypoxia-induced apoptosis of BMSCs by regulating the miR-664-2-5p/Notch1 pathway and its therapeutic effect on SANFH.
Collapse
Affiliation(s)
- Tao Wang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Zhi-Hong Xie
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Lei Wang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Hong Luo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Jian Zhang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Wen-Tao Dong
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Xiao-Han Zheng
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Chuan Ye
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Xiao-Bin Tian
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Gang Liu
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Xue-Song Zhu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, P.R. China
| | - Yan-Lin Li
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, P.R. China
| | - Qing-Lin Kang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, P.R. China
| | - Fei Zhang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
| | - Wu-Xun Peng
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
| |
Collapse
|
17
|
Nikolopoulos VK, Augustine R, Camci-Unal G. Harnessing the potential of oxygen-generating materials and their utilization in organ-specific delivery of oxygen. Biomater Sci 2023; 11:1567-1588. [PMID: 36688522 PMCID: PMC10015602 DOI: 10.1039/d2bm01329k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The limited availability of transplantable organs hinders the success of patient treatment through organ transplantation. In addition, there are challenges with immune rejection and the risk of disease transmission when receiving organs from other individuals. Tissue engineering aims to overcome these challenges by generating functional three-dimensional (3D) tissue constructs. When developing tissues or organs of a particular shape, structure, and size as determined by the specific needs of the therapeutic intervention, a tissue specific oxygen supply to all parts of the tissue construct is an utmost requirement. Moreover, the lack of a functional vasculature in engineered tissues decreases cell survival upon implantation in the body. Oxygen-generating materials can alleviate this challenge in engineered tissue constructs by providing oxygen in a sustained and controlled manner. Oxygen-generating materials can be incorporated into 3D scaffolds allowing the cells to receive and utilize oxygen efficiently. In this review, we present an overview of the use of oxygen-generating materials in various tissue engineering applications in an organ specific manner as well as their potential use in the clinic.
Collapse
Affiliation(s)
- Vasilios K Nikolopoulos
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | - Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
18
|
Augustine R, Gezek M, Seray Bostanci N, Nguyen A, Camci-Unal G. Oxygen-Generating Scaffolds: One Step Closer to the Clinical Translation of Tissue Engineered Products. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023; 455:140783. [PMID: 36644784 PMCID: PMC9835968 DOI: 10.1016/j.cej.2022.140783] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The lack of oxygen supply in engineered constructs has been an ongoing challenge for tissue engineering and regenerative medicine. Upon implantation of an engineered tissue, spontaneous blood vessel formation does not happen rapidly, therefore, there is typically a limited availability of oxygen in engineered biomaterials. Providing oxygen in large tissue-engineered constructs is a major challenge that hinders the development of clinically relevant engineered tissues. Similarly, maintaining adequate oxygen levels in cell-laden tissue engineered products during transportation and storage is another hurdle. There is an unmet demand for functional scaffolds that could actively produce and deliver oxygen, attainable by incorporating oxygen-generating materials. Recent approaches include encapsulation of oxygen-generating agents such as solid peroxides, liquid peroxides, and fluorinated substances in the scaffolds. Recent approaches to mitigate the adverse effects, as well as achieving a sustained and controlled release of oxygen, are discussed. Importance of oxygen-generating materials in various tissue engineering approaches such as ex vivo tissue engineering, in situ tissue engineering, and bioprinting are highlighted in detail. In addition, the existing challenges, possible solutions, and future strategies that aim to design clinically relevant multifunctional oxygen-generating biomaterials are provided in this review paper.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Angelina Nguyen
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
19
|
Liu S, Yang H, Zhang L, Bianco A, Ma B, Ge S. Multifunctional barrier membranes promote bone regeneration by scavenging H2O2, generating O2, eliminating inflammation, and regulating immune response. Colloids Surf B Biointerfaces 2023. [DOI: 10.1016/j.colsurfb.2023.113147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
20
|
Lnc Tmem235 promotes repair of early steroid-induced osteonecrosis of the femoral head by inhibiting hypoxia-induced apoptosis of BMSCs. Exp Mol Med 2022; 54:1991-2006. [PMID: 36380019 PMCID: PMC9723185 DOI: 10.1038/s12276-022-00875-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/24/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) have been used in the treatment of early steroid-induced osteonecrosis of the femoral head (SONFH). However, the hypoxic microenvironment in the osteonecrotic area leads to hypoxia-induced apoptosis of transplanted BMSCs, which limits their efficacy. Therefore, approaches that inhibit hypoxia-induced apoptosis of BMSCs are promising for augmenting the efficacy of BMSC transplantation. Our present study found that under hypoxia, the expression of the long noncoding RNA (Lnc) transmembrane protein 235 (Tmem235) was downregulated, the expression of Bcl-2-associated X protein was upregulated, the expression of B-cell lymphoma-2 protein was downregulated, and the apoptotic rate of BMSCs was over 70%. However, overexpression of Lnc Tmem235 reversed hypoxia-induced apoptosis of BMSCs and promoted their survival. These results demonstrated that Lnc Tmem235 effectively inhibited hypoxia-induced apoptosis of BMSCs. Mechanistically, we found that Lnc Tmem235 exhibited competitive binding to miR-34a-3p compared with BIRC5 mRNA, which is an inhibitor of apoptosis; this competitive binding relieved the silencing effect of miR-34a-3p on BIRC5 mRNA to ultimately inhibit hypoxia-induced apoptosis of BMSCs by promoting the expression of BIRC5. Furthermore, we cocultured BMSCs overexpressing Lnc Tmem235 with xenogeneic antigen-extracted cancellous bone to construct tissue-engineered bone to repair a model of early SONFH in vivo. The results showed that overexpression of Lnc Tmem235 effectively reduced apoptosis of BMSCs in the hypoxic microenvironment of osteonecrosis and improved the effect of BMSC transplantation. Taken together, our findings show that Lnc Tmem235 inhibited hypoxia-induced apoptosis of BMSCs by regulating the miR-34a-3p/BIRC5 axis, thus improving the transplantation efficacy of BMSCs for treating early SONFH.
Collapse
|
21
|
徐 鑫, 范 骁, 吴 鑫, 时 利, 王 培, 高 福, 孙 伟, 李 子. [Protective effect of Kaempferol on endothelial cell injury in glucocorticoid induced osteonecrosis of the femoral head]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:1277-1287. [PMID: 36310467 PMCID: PMC9626266 DOI: 10.7507/1002-1892.202204028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/23/2022] [Indexed: 01/24/2023]
Abstract
Objective To explore the effect of Kaempferol on bone microvascular endothelial cells (BMECs) in glucocorticoid induced osteonecrosis of the femoral head (GIONFH) in vitro. Methods BMECs were isolated from cancellous bone of femoral head or femoral neck donated voluntarily by patients with femoral neck fracture. BMECs were identified by von Willebrand factor and CD31 immunofluorescence staining and tube formation assay. The cell counting kit 8 (CCK-8) assay was used to screen the optimal concentration and the time point of dexamethasone (Dex) to inhibit the cell activity and the optimal concentration of Kaempferol to improve the inhibition of Dex. Then the BMECs were divided into 4 groups, namely, the cell group (group A), the cells treated with optimal concentration of Dex group (group B), the cells treated with optimal concentration of Dex+1 μmol/L Kaempferol group (group C), and the cells treated with optimal concentration of Dex+5 μmol/L Kaempferol group (group D). EdU assay, in vitro tube formation assay, TUNEL staining assay, Annexin Ⅴ/propidium iodide (PI) staining assay, Transwell migration assay, scratch healing assay, and Western blot assay were used to detect the effect of Kaempferol on the proliferation, tube formation, apoptosis, migration, and protein expression of BMECs treated with Dex. Results The cultured cells were identified as BMECs. CCK-8 assay showed that the optimal concentration and the time point of Dex to inhibit cell activity was 300 μmol/L for 24 hours, and the optimal concentration of Kaempferol to improve the inhibitory activity of Dex was 1 μmol/L. EdU and tube formation assays showed that the cell proliferation rate, tube length, and number of branch points were significantly lower in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). TUNEL and Annexin V/PI staining assays showed that the rates of TUNEL positive cells and apoptotic cells were significantly higher in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). Scratch healing assay and Transwell migration assay showed that the scratch healing rate and the number of migration cells were significantly lower in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). Western blot assay demonstrated that the relative expressions of Cleaved Caspase-3 and Bax proteins were significantly higher in groups B-D than in group A, and in groups B and D than in group C ( P<0.05); the relative expressions of matrix metalloproteinase 2, Cyclin D1, Cyclin E1, VEGFA, and Bcl2 proteins were significantly lower in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). Conclusion Kaempferol can alleviate the damage and dysfunction of BMECs in GIONFH.
Collapse
Affiliation(s)
- 鑫 徐
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 骁宇 范
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 鑫杰 吴
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 利军 时
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 培旭 王
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 福强 高
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 伟 孙
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
- 北京大学中日友好临床医学院骨科(北京 100029)Department of Orthopedics, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, P. R. China
| | - 子荣 李
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| |
Collapse
|
22
|
Zheng LW, Lan CN, Kong Y, Liu LH, Fan YM, Zhang CJ. Exosomal miR-150 derived from BMSCs inhibits TNF-α-mediated osteoblast apoptosis in osteonecrosis of the femoral head by GREM1/NF-κB signaling. Regen Med 2022; 17:739-753. [PMID: 35938412 DOI: 10.2217/rme-2021-0169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: The purpose of this study was to investigate the functions of exosomal miR-150 derived from bone marrow mesenchymal stem cells in osteonecrosis of the femoral head (ONFH). Materials & methods: Cell viability and apoptosis were detected using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry. Alizarin red staining was performed to detect calcium deposits. A rat model was established to assess the effects of exosomal miR-150 on ONFH in vivo. Results: Exosomes or exosomal miR-150 derived from bone marrow mesenchymal stem cells inhibited TNF-α-induced osteoblast apoptosis and promoted osteogenic differentiation and autophagy. Exosomal miR-150 suppressed apoptosis and induced autophagy in TNF-α-treated osteoblasts by regulating the GREM1/NF-κB axis. Exosomal miR-150 also improved the pathological features of ONFH in vivo. Conclusion: Exosomal miR-150 alleviates ONFH by mediating the GREM1/NF-κB axis. This study provides a potential therapeutic strategy for ONFH.
Collapse
Affiliation(s)
- Li-Wen Zheng
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chun-Na Lan
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Ying Kong
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Li-Hong Liu
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yong-Mei Fan
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chang-Jie Zhang
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|
23
|
Chen P, Liu Y, Liu W, Wang Y, Liu Z, Rong M. Impact of High-Altitude Hypoxia on Bone Defect Repair: A Review of Molecular Mechanisms and Therapeutic Implications. Front Med (Lausanne) 2022; 9:842800. [PMID: 35620712 PMCID: PMC9127390 DOI: 10.3389/fmed.2022.842800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/15/2022] [Indexed: 11/23/2022] Open
Abstract
Reaching areas at altitudes over 2,500–3,000 m above sea level has become increasingly common due to commerce, military deployment, tourism, and entertainment. The high-altitude environment exerts systemic effects on humans that represent a series of compensatory reactions and affects the activity of bone cells. Cellular structures closely related to oxygen-sensing produce corresponding functional changes, resulting in decreased tissue vascularization, declined repair ability of bone defects, and longer healing time. This review focuses on the impact of high-altitude hypoxia on bone defect repair and discusses the possible mechanisms related to ion channels, reactive oxygen species production, mitochondrial function, autophagy, and epigenetics. Based on the key pathogenic mechanisms, potential therapeutic strategies have also been suggested. This review contributes novel insights into the mechanisms of abnormal bone defect repair in hypoxic environments, along with therapeutic applications. We aim to provide a foundation for future targeted, personalized, and precise bone regeneration therapies according to the adaptation of patients to high altitudes.
Collapse
Affiliation(s)
- Pei Chen
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yushan Liu
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Wenjing Liu
- Department of Prosthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yarong Wang
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Ziyi Liu
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Mingdeng Rong
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Ma J, Sun Y, Zhou H, Li X, Bai Y, Liang C, Jia X, Zhang P, Yang L. Animal Models of Femur Head Necrosis for Tissue Engineering and Biomaterials Research. Tissue Eng Part C Methods 2022; 28:214-227. [PMID: 35442092 DOI: 10.1089/ten.tec.2022.0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Femur head necrosis, also known as osteonecrosis of the femoral head (ONFH), is a widespread disabling pathology mostly affecting young and middle-aged population and one of the major causes of total hip arthroplasty in the elderly. Currently, there are limited number of different clinical or medication options for the treatment or the reversal of progressive ONFH, but their clinical outcomes are neither satisfactory nor consistent. In pursuit of more reliable therapeutic strategies for ONFH, including recently emerged tissue engineering and biomaterials approaches, in vivo animal models are extremely important for therapeutic efficacy evaluation and mechanistic exploration. Based on the better understanding of pathogenesis of ONFH, animal modeling method has evolved into three major routes, including steroid-, alcohol-, and injury/trauma-induced osteonecrosis, respectively. There is no consensus yet on a standardized ONFH animal model for tissue engineering and biomaterial research; therefore, appropriate animal modeling method should be carefully selected depending on research purposes and scientific hypotheses. In this work, mainstream types of ONFH animal model and their modeling techniques are summarized, showing both merits and demerits for each. In addition, current studies and experimental techniques of evaluating therapeutic efficacy on the treatment of ONFH using animal models are also summarized, along with discussions on future directions related to tissue engineering and biomaterial research. Impact statement Exploration of tissue engineering and biomaterial-based therapeutic strategy for the treatment of femur head necrosis is important since there are limited options available with satisfactory clinical outcomes. To promote the translation of these technologies from benchwork to bedside, animal model should be carefully selected to provide reliable results and clinical outcome prediction. Therefore, osteonecrosis of the femoral head animal modeling methods as well as associated tissue engineering and biomaterial research are overviewed and discussed in this work, as an attempt to provide guidance for model selection and optimization in tissue engineering and biomaterial translational studies.
Collapse
Affiliation(s)
- Jiali Ma
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Yuting Sun
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Huan Zhou
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China.,Center for Health Sciences and Engineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yanjie Bai
- School of Chemical Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Chunyong Liang
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China.,Changzhou Blon Minimally Invasive Medical Device Technology Co. Ltd., Jiangsu, People's Republic of China
| | - Xiaowei Jia
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Lei Yang
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China.,Center for Health Sciences and Engineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| |
Collapse
|
25
|
Wang P, Meng X, Wang R, Yang W, Yang L, Wang J, Wang DA, Fan C. Biomaterial Scaffolds Made of Chemically Cross-Linked Gelatin Microsphere Aggregates (C-GMSs) Promote Vascularized Bone Regeneration. Adv Healthc Mater 2022; 11:e2102818. [PMID: 35306762 DOI: 10.1002/adhm.202102818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/09/2022] [Indexed: 12/14/2022]
Abstract
Various scaffolding systems have been attempted to facilitate vascularization in tissue engineering by optimizing biophysical properties (e.g., vascular-like structures, porous architectures, surface topographies) or loading biochemical factors (e.g., growth factors, hormones). However, vascularization during ossification remains an unmet challenge that hampers the repair of large bone defects. In this study, reconstructing vascularized bones in situ against critical-sized bone defects is endeavored using newly developed scaffolds made of chemically cross-linked gelatin microsphere aggregates (C-GMSs). The rationale of this design lies in the creation and optimization of cell-material interfaces to enhance focal adhesion, proliferation, and function of anchorage-dependent functional cells. In vitro trials are carried out by coculturing human aortic endothelial cells (HAECs) and murine osteoblast precursor cells (MC3T3-E1) within C-GMS scaffolds, in which endothelialized bone-like constructs are yielded. Angiogenesis and osteogenesis induced by C-GMSs scaffold are further confirmed via subcutaneous-embedding trials in nude mice. In situ trials for the repair of critical-sized femoral defects are subsequently performed in rats. The acellular C-GMSs with interconnected macropores, exhibit the capability to recruit the endogenous cells (e.g., bone-forming cells, vascular forming cells, immunocytes) and then promote vascularized bone regeneration as well as integration with host bone.
Collapse
Affiliation(s)
- Peiyan Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, 266021, P. R. China
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| | - Xinyue Meng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, 266021, P. R. China
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| | - Runze Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| | - Wei Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, 266021, P. R. China
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| | - Lanting Yang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| | - Jianxun Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, P. R. China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518057, P. R. China
| | - Changjiang Fan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, 266021, P. R. China
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, Shandong, 266071, P. R. China
| |
Collapse
|
26
|
Zhang XX, Liang X, Li SR, Guo KJ, Li DF, Li TF. Bone Marrow Mesenchymal Stem Cells Overexpressing HIF-1α Prevented the Progression of Glucocorticoid-Induced Avascular Osteonecrosis of Femoral Heads in Mice. Cell Transplant 2022; 31:9636897221082687. [PMID: 35287482 PMCID: PMC8928352 DOI: 10.1177/09636897221082687] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoid (GC)-induced avascular osteonecrosis of femoral head (AOFH) is a devastating complication, and no cures are currently available for it. Previous studies have demonstrated that implantation of bone marrow mesenchymal stem cells (BMMSCs) may prevent the progression of pre-collapse AOFH. Based on previous observations, we hypothesized that GCs induce AOFH via the COX-2 (cyclooxygenase-2)-PGE-2 (prostaglandin E2)-HIF-1α (hypoxia-inducible factor-1α) axis, and that modification of BMMSCs may improve the efficacy of their implantation. BMMSCs isolated from wild-type (WT) mice were treated with dexamethasone (Dex) and the results showed that Dex repressed the expression of COX-2. Femoral head samples harvested from both WT and COX-2 knock-out (COX-2-/-) mice were subjected to micro-computed tomography and histological examinations. Compared with their WT littermates, COX-2-/- mice had larger trabecular separations, diminished microvasculature, and reduced HIF-1α expression in their femoral heads. In vitro angiogenesis assays with tube formation and fetal metatarsal sprouting demonstrated that Dex repressed angiogenesis and PGE-2 antagonized its effects. An AOFH model was successfully established in C57BL/6J mice. In vitro experiment showed that BMMSCs infected with Lentivirus encoding HIF-1α (Lenti-HIF-1α) resulted in a robust increase in the production of HIF-1α protein. Implantation of BMMSCs overexpressing HIF-1α into femoral heads of AOFH mice significantly reduced osteonecrotic areas and enhanced bone repair, thus largely preserving the structural integrity of femoral heads. Our studies provide strong rationales for early intervention with core decompression and implantation of modified BMMSCs for GC-induced AOFH, which may spare patients from expensive and difficult surgical procedures.
Collapse
Affiliation(s)
- Xin-Xin Zhang
- Department of Rheumatology, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| | - Xu Liang
- Department of Rheumatology, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| | - Sen-Rui Li
- Department of Rheumatology, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| | - Kuang-Jin Guo
- Department of Rheumatology, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| | - Dai-Feng Li
- Department of Orthopaedics, Zhengzhou University First Affiliated Hospital, Zhengzhou, China.,Department of Magnetic Resonance Imaging, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| | - Tian-Fang Li
- Department of Rheumatology, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| |
Collapse
|
27
|
林 苗, 杨 继, 闫 文, 胡 宁, 刘 子, 张 莉, 李 豫. [Research progress of tissue engineering technology in promoting revascularization of necrotic femoral bone tissue]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:1479-1485. [PMID: 34779177 PMCID: PMC8586766 DOI: 10.7507/1002-1892.202105047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/19/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To summarize the research progress of tissue engineering technology to promote bone tissue revascularization in osteonecrosis of the femoral head (ONFH). METHODS The relevant domestic and foreign literature in recent years was extensively reviewed. The mechanism of femoral head vascularization and the application progress of tissue engineering technology in the promotion of ONFH bone tissue revascularization were summarized. RESULTS Rebuilding or improving the blood supply of the femoral head is the key to the treatment of ONFH. Tissue engineering is a hot spot in current research. It mainly focuses on the three elements of seed cells, scaffold materials, and angiogenic growth factors, combined with three-dimensional printing technology and drug delivery systems to promote the revascularization of the femoral bone tissue. CONCLUSION The strategy of revascularization of the femoral head can improve the local blood supply and delay or even reverse the progression of ONFH disease.
Collapse
Affiliation(s)
- 苗远 林
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| | - 继滨 杨
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| | - 文强 闫
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| | - 宁 胡
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| | - 子铭 刘
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| | - 莉 张
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| | - 豫皖 李
- 遵义医科大学附属医院(贵州遵义 563000)Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou, 563000, P.R.China
| |
Collapse
|
28
|
He S, Qin T. [Research progress of interfacial tissue engineering in rotator cuff repair]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:1341-1351. [PMID: 34651491 DOI: 10.7507/1002-1892.202104064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective To summarize the research progress of interfacial tissue engineering in rotator cuff repair. Methods The recent literature at home and abroad concerning interfacial tissue engineering in rotator cuff repair was analysed and summarized. Results Interfacial tissue engineering is to reconstruct complex and hierarchical interfacial tissues through a variety of methods to repair or regenerate damaged joints of different tissues. Interfacial tissue engineering in rotator cuff repair mainly includes seed cells, growth factors, biomaterials, oxygen concentration, and mechanical stimulation. Conclusion The best strategy for rotator cuff healing and regeneration requires not only the use of biomaterials with gradient changes, but also the combination of seed cells, growth factors, and specific culture conditions (such as oxygen concentration and mechanical stimulation). However, the clinical transformation of the relevant treatment is still a very slow process.
Collapse
Affiliation(s)
- Shukun He
- Laboratory of Stem Cells and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Tingwu Qin
- Laboratory of Stem Cells and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| |
Collapse
|
29
|
Yang X, Wang Y, Zhou Y, Chen J, Wan Q. The Application of Polycaprolactone in Three-Dimensional Printing Scaffolds for Bone Tissue Engineering. Polymers (Basel) 2021; 13:polym13162754. [PMID: 34451293 PMCID: PMC8400029 DOI: 10.3390/polym13162754] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/25/2021] [Accepted: 08/12/2021] [Indexed: 02/05/2023] Open
Abstract
Bone tissue engineering commonly encompasses the use of three-dimensional (3D) scaffolds to provide a suitable microenvironment for the propagation of cells to regenerate damaged tissues or organs. 3D printing technology has been extensively applied to allow direct 3D scaffolds manufacturing. Polycaprolactone (PCL) has been widely used in the fabrication of 3D scaffolds in the field of bone tissue engineering due to its advantages such as good biocompatibility, slow degradation rate, the less acidic breakdown products in comparison to other polyesters, and the potential for loadbearing applications. PCL can be blended with a variety of polymers and hydrogels to improve its properties or to introduce new PCL-based composites. This paper describes the PCL used in developing state of the art of scaffolds for bone tissue engineering. In this review, we provide an overview of the 3D printing techniques for the fabrication of PCL-based composite scaffolds and recent studies on applications in different clinical situations. For instance, PCL-based composite scaffolds were used as an implant surgical guide in dental treatment. Furthermore, future trend and potential clinical translations will be discussed.
Collapse
Affiliation(s)
- Xiangjun Yang
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.Y.); (Y.W.); (Y.Z.)
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuting Wang
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.Y.); (Y.W.); (Y.Z.)
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ying Zhou
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.Y.); (Y.W.); (Y.Z.)
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junyu Chen
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.Y.); (Y.W.); (Y.Z.)
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (J.C.); (Q.W.)
| | - Qianbing Wan
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (X.Y.); (Y.W.); (Y.Z.)
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (J.C.); (Q.W.)
| |
Collapse
|