151
|
Kapinova A, Mazurakova A, Halasova E, Dankova Z, Büsselberg D, Costigliola V, Golubnitschaja O, Kubatka P. Underexplored reciprocity between genome-wide methylation status and long non-coding RNA expression reflected in breast cancer research: potential impacts for the disease management in the framework of 3P medicine. EPMA J 2023; 14:249-273. [PMID: 37275549 PMCID: PMC10236066 DOI: 10.1007/s13167-023-00323-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023]
Abstract
Breast cancer (BC) is the most common female malignancy reaching a pandemic scale worldwide. A comprehensive interplay between genetic alterations and shifted epigenetic regions synergistically leads to disease development and progression into metastatic BC. DNA and histones methylations, as the most studied epigenetic modifications, represent frequent and early events in the process of carcinogenesis. To this end, long non-coding RNAs (lncRNAs) are recognized as potent epigenetic modulators in pathomechanisms of BC by contributing to the regulation of DNA, RNA, and histones' methylation. In turn, the methylation status of DNA, RNA, and histones can affect the level of lncRNAs expression demonstrating the reciprocity of mechanisms involved. Furthermore, lncRNAs might undergo methylation in response to actual medical conditions such as tumor development and treated malignancies. The reciprocity between genome-wide methylation status and long non-coding RNA expression levels in BC remains largely unexplored. Since the bio/medical research in the area is, per evidence, strongly fragmented, the relevance of this reciprocity for BC development and progression has not yet been systematically analyzed. Contextually, the article aims at:consolidating the accumulated knowledge on both-the genome-wide methylation status and corresponding lncRNA expression patterns in BC andhighlighting the potential benefits of this consolidated multi-professional approach for advanced BC management. Based on a big data analysis and machine learning for individualized data interpretation, the proposed approach demonstrates a great potential to promote predictive diagnostics and targeted prevention in the cost-effective primary healthcare (sub-optimal health conditions and protection against the health-to-disease transition) as well as advanced treatment algorithms tailored to the individualized patient profiles in secondary BC care (effective protection against metastatic disease). Clinically relevant examples are provided, including mitochondrial health control and epigenetic regulatory mechanisms involved.
Collapse
Affiliation(s)
- Andrea Kapinova
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Alena Mazurakova
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Erika Halasova
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Zuzana Dankova
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | | | - Olga Golubnitschaja
- Predictive, Preventive, and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
152
|
Moura SR, Fernandes MJ, Santos SG, Almeida MI. Circular RNAs: Promising Targets in Osteoporosis. Curr Osteoporos Rep 2023; 21:289-302. [PMID: 37119447 PMCID: PMC10169890 DOI: 10.1007/s11914-023-00786-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE OF REVIEW Circular RNAs (circRNAs) are RNA transcripts derived from fragments of pre-messenger RNAs through a back-splicing process. An advantage that rises from their circular covalently closed conformation is their high stability, when compared with their linear counterparts. The current review focuses on the emerging roles of circRNAs in osteoporosis, including in osteogenic differentiation and osteoclastogenesis. Their potential as osteoporosis biomarkers will also be discussed. RECENT FINDINGS Although firstly described as non-coding, some of these single-stranded RNAs were recently reported to possess protein-coding capacity. On the other hand, the circRNAs exhibit cell and tissue-specific patterns at the transcriptome level in eukaryotes and are regulated throughout the development or disease progression. Even though thousands of these circular transcripts are listed and annotated, only a limited number of studies describe their biological role in bone processes. Recent evidence indicates inhibitory activator roles in both osteoblasts and osteoclasts differentiation and function. Latest screenings in the blood, plasma, or serum of osteoporosis patients support the potential for circRNA signature to be used as biomarkers in osteoporosis, but further validation is required. While intense research into circRNAs has been detailing their biological roles, there remains a need for standardization and further research to fulfil the future potential of this emerging and highly promising class of regulatory molecules.
Collapse
Affiliation(s)
- Sara Reis Moura
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria João Fernandes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Maria Inês Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
153
|
Zhang J, Tang S, Ding N, Ma P, Zhang Z. Surface-modified Ti 3C 2 MXene nanosheets for mesenchymal stem cell osteogenic differentiation via photothermal conversion. NANOSCALE ADVANCES 2023; 5:2921-2932. [PMID: 37260501 PMCID: PMC10228341 DOI: 10.1039/d3na00187c] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/21/2023] [Indexed: 06/02/2023]
Abstract
In the field of bone tissue engineering, the practical application of growth factors is limited by various factors such as systemic toxicity, instability, and the potential to induce inflammation. To circumvent these limitations, the use of physical signals, such as thermal stimulation, to regulate stem cells has been proposed as a promising alternative. The present study aims to investigate the potential of the two-dimensional nanomaterial Ti3C2 MXene, which exhibits unique photothermal properties, to induce osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via photothermal conversion. Surface modification of Ti3C2 MXene nanosheets with PVP (Ti3C2-PVP) was employed to enhance their colloidal stability in physiological solutions. Characterization and cellular experiments showed that Ti3C2-PVP nanosheets have favorable photothermal properties and biocompatibility. Our study demonstrated that the induction of photothermal stimulation by co-culturing Ti3C2-PVP nanosheets with BMSCs and subsequent irradiation with 808 nm NIR significantly promoted cell proliferation, adhesion and osteogenic differentiation of BMSCs. In conclusion, the results of this study suggest that Ti3C2-PVP is a promising material for bone tissue engineering applications as it can modulate the cellular functions of BMSCs through photothermal conversion.
Collapse
Affiliation(s)
- Jiebing Zhang
- School of Stomatology, Capital Medical University Beijing PR China
| | - Shuang Tang
- School of Stomatology, Capital Medical University Beijing PR China
| | - Ning Ding
- School of Stomatology, Capital Medical University Beijing PR China
| | - Ping Ma
- School of Stomatology, Capital Medical University Beijing PR China
| | - Zutai Zhang
- School of Stomatology, Capital Medical University Beijing PR China
| |
Collapse
|
154
|
Ganesh SS, Anushikaa R, Swetha Victoria VS, Lavanya K, Shanmugavadivu A, Selvamurugan N. Recent Advancements in Electrospun Chitin and Chitosan Nanofibers for Bone Tissue Engineering Applications. J Funct Biomater 2023; 14:jfb14050288. [PMID: 37233398 DOI: 10.3390/jfb14050288] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023] Open
Abstract
Treatment of large segmental bone loss caused by fractures, osteomyelitis, and non-union results in expenses of around USD 300,000 per case. Moreover, the worst-case scenario results in amputation in 10% to 14.5% of cases. Biomaterials, cells, and regulatory elements are employed in bone tissue engineering (BTE) to create biosynthetic bone grafts with effective functionalization that can aid in the restoration of such fractured bones, preventing amputation and alleviating expenses. Chitin (CT) and chitosan (CS) are two of the most prevalent natural biopolymers utilized in the fields of biomaterials and BTE. To offer the structural and biochemical cues for augmenting bone formation, CT and CS can be employed alone or in combination with other biomaterials in the form of nanofibers (NFs). When compared with several fabrication methods available to produce scaffolds, electrospinning is regarded as superior since it enables the development of nanostructured scaffolds utilizing biopolymers. Electrospun nanofibers (ENFs) offer unique characteristics, including morphological resemblance to the extracellular matrix, high surface-area-to-volume ratio, permeability, porosity, and stability. This review elaborates on the recent strategies employed utilizing CT and CS ENFs and their biocomposites in BTE. We also summarize their implementation in supporting and delivering an osteogenic response to treat critical bone defects and their perspectives on rejuvenation. The CT- and CS-based ENF composite biomaterials show promise as potential constructions for bone tissue creation.
Collapse
Affiliation(s)
- S Shree Ganesh
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Ramprasad Anushikaa
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Venkadesan Sri Swetha Victoria
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Krishnaraj Lavanya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| |
Collapse
|
155
|
Pathania AS. Crosstalk between Noncoding RNAs and the Epigenetics Machinery in Pediatric Tumors and Their Microenvironment. Cancers (Basel) 2023; 15:2833. [PMID: 37345170 DOI: 10.3390/cancers15102833] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
According to the World Health Organization, every year, an estimated 400,000+ new cancer cases affect children under the age of 20 worldwide. Unlike adult cancers, pediatric cancers develop very early in life due to alterations in signaling pathways that regulate embryonic development, and environmental factors do not contribute much to cancer development. The highly organized complex microenvironment controlled by synchronized gene expression patterns plays an essential role in the embryonic stages of development. Dysregulated development can lead to tumor initiation and growth. The low mutational burden in pediatric tumors suggests the predominant role of epigenetic changes in driving the cancer phenotype. However, one more upstream layer of regulation driven by ncRNAs regulates gene expression and signaling pathways involved in the development. Deregulation of ncRNAs can alter the epigenetic machinery of a cell, affecting the transcription and translation profiles of gene regulatory networks required for cellular proliferation and differentiation during embryonic development. Therefore, it is essential to understand the role of ncRNAs in pediatric tumor development to accelerate translational research to discover new treatments for childhood cancers. This review focuses on the role of ncRNA in regulating the epigenetics of pediatric tumors and their tumor microenvironment, the impact of their deregulation on driving pediatric tumor progress, and their potential as effective therapeutic targets.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
156
|
Zhang Y, Jiang S, Xu D, Li Z, Guo J, Li Z, Cheng G. Application of Nanocellulose-Based Aerogels in Bone Tissue Engineering: Current Trends and Outlooks. Polymers (Basel) 2023; 15:polym15102323. [PMID: 37242898 DOI: 10.3390/polym15102323] [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: 03/08/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The complex or compromised bone defects caused by osteomyelitis, malignant tumors, metastatic tumors, skeletal abnormalities, and systemic diseases are difficult to be self-repaired, leading to a non-union fracture. With the increasing demands of bone transplantation, more and more attention has been paid to artificial bone substitutes. As biopolymer-based aerogel materials, nanocellulose aerogels have been widely utilized in bone tissue engineering. More importantly, nanocellulose aerogels not only mimic the structure of the extracellular matrix but could also deliver drugs and bioactive molecules to promote tissue healing and growth. Here, we reviewed the most recent literature about nanocellulose-based aerogels, summarized the preparation, modification, composite fabrication, and applications of nanocellulose-based aerogels in bone tissue engineering, as well as giving special focus to the current limitations and future opportunities of nanocellulose aerogels for bone tissue engineering.
Collapse
Affiliation(s)
- Yaoguang Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Shengjun Jiang
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430079, China
| | - Dongdong Xu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325015, China
| | - Zubing Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jie Guo
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Zhi Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Gu Cheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
157
|
Kim MK, Paek K, Woo SM, Kim JA. Bone-on-a-Chip: Biomimetic Models Based on Microfluidic Technologies for Biomedical Applications. ACS Biomater Sci Eng 2023. [PMID: 37183366 DOI: 10.1021/acsbiomaterials.3c00066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
With the increasing importance of preclinical evaluation of newly developed drugs or treatments, in vitro organ or disease models are necessary. Although various organ-specific on-chip (organ-on-a-chip, or OOC) systems have been developed as emerging in vitro models, bone-on-a-chip (BOC) systems that recapitulate the bone microenvironment have been less developed or reviewed compared with other OOCs. The bone is one of the most dynamic organs and undergoes continuous remodeling throughout its lifetime. The aging population is growing worldwide, and healthcare costs are rising rapidly. Since in vitro BOC models that recapitulate native bone niches and pathological features can be important for studying the underlying mechanism of orthopedic diseases and predicting drug responses in preclinical trials instead of in animals, the development of biomimetic BOCs with high efficiency and fidelity will be accelerated further. Here, we review recently engineered BOCs developed using various microfluidic technologies and investigate their use to model the bone microenvironment. We have also explored various biomimetic strategies based on biological, geometrical, and biomechanical cues for biomedical applications of BOCs. Finally, we addressed the limitations and challenging issues of current BOCs that should be overcome to obtain more acceptable BOCs in the biomedical and pharmaceutical industries.
Collapse
Affiliation(s)
- Min Kyeong Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Kyurim Paek
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Program in Biomicro System Technology, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Mi Woo
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Republic of Korea
| |
Collapse
|
158
|
Jeyaraman M, Sami A, Nallakumarasamy A, Jeyaraman N, Jain VK. Hyperbaric Oxygen Therapy in Orthopaedics: An Adjunct Therapy with an Emerging Role. Indian J Orthop 2023; 57:748-761. [PMID: 37128570 PMCID: PMC10147865 DOI: 10.1007/s43465-023-00837-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Introduction Hyperbaric oxygen therapy (HBOT) has emerged as an adjunct treatment modality in various orthopedic and rheumatological conditions. Undersea and Hyperbaric Medical Society (UHMS) defined the minimum number of HBOT cycles, dose, and frequency for various diseases. UHMS laid the 14 absolute indications for HBOT. This article deals with the mechanism of actions of HBOT and evidence of various musculoskeletal disorders where HBOT was utilized to accelerate the healing process of the diseases. Materials and methods The review literature search was conducted by using PubMed, SCOPUS, and other database of medical journals for identifying, reviewing, and evaluating the published clinical trial data, research study, and review articles for the use of HBOT in musculoskeletal disorders. Results Various clinical researchers documented cellular and biochemical advantages of HBOT which possess allodynic effects, anti-inflammatory, and prooxygenatory effects in patients with musculoskeletal conditions. Studies on the usage of HBOT in avascular necrosis and wound healing provide a platform for exploring the plausible uses of HBOT in other musculoskeletal conditions. Literature evidence states the complications associated with HBOT therapy. Conclusion The existing HBOT protocols have to be optimized for various musculoskeletal disorders. Large scale blinded RCTs have to be performed for demonstrating the level of evidence in the usage of HBOT in various musculoskeletal clinical scenarios.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600056 India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
| | - Abdus Sami
- Department of Orthopaedics, Atal Bihari Vajpayee Institute of Medical Sciences, Dr Ram Manohar Lohia Hospital, New Delhi, 110001 India
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| | - Naveen Jeyaraman
- Department of Orthopaedics, Rathimed Specialty Hospital, Chennai, Tamil Nadu 600040 India
| | - Vijay Kumar Jain
- Department of Orthopaedics, Atal Bihari Vajpayee Institute of Medical Sciences, Dr Ram Manohar Lohia Hospital, New Delhi, 110001 India
| |
Collapse
|
159
|
Hao Z, Ren L, Zhang Z, Yang Z, Wu S, Liu G, Cheng B, Wu J, Xia J. A multifunctional neuromodulation platform utilizing Schwann cell-derived exosomes orchestrates bone microenvironment via immunomodulation, angiogenesis and osteogenesis. Bioact Mater 2023; 23:206-222. [DOI: 10.1016/j.bioactmat.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/15/2022] Open
|
160
|
Valizadeh N, Salehi R, Aghazadeh M, Alipour M, Sadeghzadeh H, Mahkam M. Enhanced osteogenic differentiation and mineralization of human dental pulp stem cells using Prunus amygdalus amara (bitter almond) incorporated nanofibrous scaffold. J Mech Behav Biomed Mater 2023; 142:105790. [PMID: 37104899 DOI: 10.1016/j.jmbbm.2023.105790] [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: 02/03/2023] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 04/29/2023]
Abstract
Polyphenol extracts derived from plants are expected to have enhanced osteoblast proliferation and differentiation ability, which has gained much attention in tissue engineering applications. Herein, for the first time, we investigate the effects of Prunus amygdalus amara (bitter almond) (BA) extract loaded on poly (ε-caprolactone) (PCL)/gelatin (Gt) nanofibrous scaffolds on the osteoblast differentiation of human dental pulp stem cells (DPSCs). In this regard, BA (0, 5, 10, and 15% wt)-loaded PCL/Gt nanofibrous scaffolds were prepared by electrospinning with fiber diameters in the range of around 237-276 nm. Morphology, composition, porosity, hydrophilicity, and mechanical properties of the scaffolds were examined by FESEM, ATR-FTIR spectroscopy, BET, contact angle, and tensile tests, respectively. It was found that the addition of BA improved the tensile strength (up to 6.1 times), Young's modulus (up to 3 times), and strain at break (up to 3.2 times) compared to the neat PCL/Gt nanofibers. Evaluations of cell attachment, spreading, and proliferation were done by FESEM observation and MTT assay. Cytocompatibility studies support the biocompatible nature of BA loaded PCL/Gt scaffolds and free BA by demonstrating cell viability of more than 100% in all groups. The results of alkaline phosphatase activity and Alizarin Red assay revealed that osteogenic activity levels of BA loaded PCL/Gt scaffolds and free BA were significantly increased compared to the control group (p < 0.05, p < 0.01, p < 0.001). QRT-PCR results demonstrated that BA loaded PCL/Gt scaffolds and free BA led to a significant increase in osteoblast differentiation of DPSCs through the upregulation of osteogenic related genes compared to the control group (p < 0.05). Based on results, incorporation of BA extract in PCL/Gt scaffolds exhibited synergistic effects on the adhesion, proliferation, and osteogenesis differentiation of hDPSCs and was therefore assumed to be a favorable scaffold for bone tissue engineering applications.
Collapse
Affiliation(s)
- Nasrin Valizadeh
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Marziyeh Aghazadeh
- Stem Cell Research Center and Department of Oral Medicine, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Sadeghzadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Mahkam
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran.
| |
Collapse
|
161
|
Hayashi K, Yanagisawa T, Kishida R, Tsuchiya A, Ishikawa K. Gear-shaped carbonate apatite granules with a hexagonal macropore for rapid bone regeneration. Comput Struct Biotechnol J 2023; 21:2514-2523. [PMID: 37077175 PMCID: PMC10106487 DOI: 10.1016/j.csbj.2023.03.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Synthetic bone grafts are in high demand owing to increased age-related bone disorders in the global aging population. Here, we report fabrication of gear-shaped granules (G-GRNs) for rapid bone healing. G-GRNs possessed six protrusions and a hexagonal macropore in the granular center. These were composed of carbonate apatite, i.e., bone mineral, microspheres with ∼1-μm micropores in the spaces between the microspheres. G-GRNs formed new bone and blood vessels (both on the granular surface and within the macropores) 4 weeks after implantation in the rabbit femur defects. The formed bone structure was similar to that of cancellous bone. The bone percentage in the defect recovered to that in a normal rabbit femur at week-4 post-implantation, and the bone percentage remained constant for the following 8 weeks. Throughout the entire period, the bone percentage in the G-GRN-implanted group was ∼10% higher than that of the group implanted with conventional carbonate apatite granules. Furthermore, a portion of the G-GRNs resorbed at week-4, and resorption continued for the following 8 weeks. Thus, G-GRNs are involved in bone remodeling and are gradually replaced with new bone while maintaining a suitable bone level. These findings provide a basis for the design and fabrication of synthetic bone grafts for achieving rapid bone regeneration.
Collapse
|
162
|
Koushik TM, Miller CM, Antunes E. Bone Tissue Engineering Scaffolds: Function of Multi-Material Hierarchically Structured Scaffolds. Adv Healthc Mater 2023; 12:e2202766. [PMID: 36512599 PMCID: PMC11468595 DOI: 10.1002/adhm.202202766] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/29/2022] [Indexed: 12/15/2022]
Abstract
Bone tissue engineering (BTE) is a topic of interest for the last decade, and advances in materials, processing techniques, and the understanding of bone healing pathways have opened new avenues of research. The dual responsibility of BTE scaffolds in providing load-bearing capability and interaction with the local extracellular matrix to promote bone healing is a challenge in synthetic scaffolds. This article describes the usage and processing of multi-materials and hierarchical structures to mimic the structure of natural bone tissues to function as bioactive and load-bearing synthetic scaffolds. The first part of this literature review describes the physiology of bone healing responses and the interactions at different stages of bone repair. The following section reviews the available literature on biomaterials used for BTE scaffolds followed by some multi-material approaches. The next section discusses the impact of the scaffold's structural features on bone healing and the necessity of a hierarchical distribution in the scaffold structure. Finally, the last section of this review highlights the emerging trends in BTE scaffold developments that can inspire new tissue engineering strategies and truly develop the next generation of synthetic scaffolds.
Collapse
Affiliation(s)
- Tejas M. Koushik
- College of Science and EngineeringJames Cook UniversityTownsvilleQueensland4811Australia
| | - Catherine M. Miller
- College of Medicine and DentistryJames Cook UniversitySmithfieldQueensland4878Australia
| | - Elsa Antunes
- College of Science and EngineeringJames Cook UniversityTownsvilleQueensland4811Australia
| |
Collapse
|
163
|
Erogul O, Gobeka HH, Kasikci M, Erogul LE, Balci A. Impacts of protective face masks on ocular surface symptoms among healthcare professionals during the COVID-19 pandemic. Ir J Med Sci 2023; 192:521-526. [PMID: 35697968 PMCID: PMC9191881 DOI: 10.1007/s11845-022-03059-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/07/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND To assess the impacts of prolonged protective face masks (PFM) wear on ocular surface symptoms among healthcare professionals (HCPs), and how these symptoms affected PFM wear. METHODS Thirty-question survey forms were distributed via social media platform to 396 HCPs (110 doctors, 164 nurses, and 122 health technicians) between September 8 and 30, 2021. Participants who could not be reached via social media were given a face-to-face questionnaire. Aside from sociodemographic data, the questionnaire inquired about PFM wear, PFM types, ocular surface symptoms, and how PFM wear has changed during the COVID-19 pandemic. RESULTS A total of 74.5% of HCPs reported wearing PFMs, mostly surgical ones (76.8%), for half a day at work but not at home, with redness (29.3%) being the most frequently encountered ocular surface symptom, followed by burning (15.7%), pain (14.1%), tingling (10.9%), and rash (6.6%). The presence of associated restrictions in conjunction with PFM-related ocular symptoms was more likely in dry and hot environments. There was no significant relationship between PFM type, PFM-wearing duration, and HCPs' daily activities (p > 0.05). Despite the lack of a significant relationship between PFM types and ocular surface symptoms (p > 0.05), there was a significant relationship between PFM-wearing duration and ocular pain (p < 0.05). CONCLUSIONS PFM-related ocular surface symptoms can be alleviated by properly wearing PFMs, reducing wear time, and using long-acting topical lubricants. This could improve PFM wear compliance, prevent disease transmission, and ultimately help with COVID-19 protection.
Collapse
Affiliation(s)
- Ozgur Erogul
- Department of Ophthalmology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Hamidu Hamisi Gobeka
- Department of Ophthalmology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Murat Kasikci
- Department of Ophthalmology, Muğla Sıtkı Koçman University Education and Research Hospital, Muğla, Turkey
| | - Leyla Eryigit Erogul
- Department of Ophthalmology, Afyonkarahisar Parkhayat Hospital, Afyonkarahisar, Turkey
| | - Aydin Balci
- Department of Chest Diseases, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| |
Collapse
|
164
|
Silva-Barroso AS, Cabral CSD, Ferreira P, Moreira AF, Correia IJ. Lignin-enriched tricalcium phosphate/sodium alginate 3D scaffolds for application in bone tissue regeneration. Int J Biol Macromol 2023; 239:124258. [PMID: 37003376 DOI: 10.1016/j.ijbiomac.2023.124258] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The bone is a connective, vascularized, and mineralized tissue that confers protection to organs, and participates in the support and locomotion of the human body, maintenance of homeostasis, as well as in hematopoiesis. However, throughout the lifetime, bone defects may arise due to traumas (mechanical fractures), diseases, and/or aging, which when too extensive compromise the ability of the bone to self-regenerate. To surpass such clinical situation, different therapeutic approaches have been pursued. Rapid prototyping techniques using composite materials (consisting of ceramics and polymers) have been used to produce customized 3D structures with osteoinductive and osteoconductive properties. In order to reinforce the mechanical and osteogenic properties of these 3D structures, herein, a new 3D scaffold was produced through the layer-by-layer deposition of a tricalcium phosphate (TCP), sodium alginate (SA), and lignin (LG) mixture using the Fab@Home 3D-Plotter. Three different TCP/LG/SA formulations, LG/SA ratio 1:3, 1:2, or 1:1, were produced and subsequently evaluated to determine their suitability for bone regeneration. The physicochemical assays demonstrated that the LG inclusion improved the mechanical resistance of the scaffolds, particularly in the 1:2 ratio, since a 15 % increase in the mechanical strength was observed. Moreover, all TCP/LG/SA formulations showed an enhanced wettability and maintained their capacity to promote the osteoblasts' adhesion and proliferation as well as their bioactivity (formation of hydroxyapatite crystals). Such results support the LG inclusion and application in the development of 3D scaffolds aimed for bone regeneration.
Collapse
Affiliation(s)
- A S Silva-Barroso
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Cátia S D Cabral
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Paula Ferreira
- CIEPQPF-Departamento de Engenharia Química, Universidade de Coimbra, Rua Silvio Lima, 3030-790 Coimbra, Portugal; Instituto Superior de Engenharia de Coimbra, Instituto Politécnico de Coimbra, Rua Pedro Nunes, 3030-199 Coimbra, Portugal
| | - André F Moreira
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CPIRN-UDI/IPG-Center of Potential and Innovation in Natural Resources, Research Unit for Inland Development, Instituto Politécnico da Guarda, Avenida Dr. Francisco de Sá Carneiro, 6300-559 Guarda, Portugal
| | - Ilídio J Correia
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
165
|
Gulati K, Ding C, Guo T, Guo H, Yu H, Liu Y. Craniofacial therapy: advanced local therapies from nano-engineered titanium implants to treat craniofacial conditions. Int J Oral Sci 2023; 15:15. [PMID: 36977679 PMCID: PMC10050545 DOI: 10.1038/s41368-023-00220-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023] Open
Abstract
Nano-engineering-based tissue regeneration and local therapeutic delivery strategies show significant potential to reduce the health and economic burden associated with craniofacial defects, including traumas and tumours. Critical to the success of such nano-engineered non-resorbable craniofacial implants include load-bearing functioning and survival in complex local trauma conditions. Further, race to invade between multiple cells and pathogens is an important criterion that dictates the fate of the implant. In this pioneering review, we compare the therapeutic efficacy of nano-engineered titanium-based craniofacial implants towards maximised local therapy addressing bone formation/resorption, soft-tissue integration, bacterial infection and cancers/tumours. We present the various strategies to engineer titanium-based craniofacial implants in the macro-, micro- and nano-scales, using topographical, chemical, electrochemical, biological and therapeutic modifications. A particular focus is electrochemically anodised titanium implants with controlled nanotopographies that enable tailored and enhanced bioactivity and local therapeutic release. Next, we review the clinical translation challenges associated with such implants. This review will inform the readers of the latest developments and challenges related to therapeutic nano-engineered craniofacial implants.
Collapse
Affiliation(s)
- Karan Gulati
- The University of Queensland, School of Dentistry, Herston, QLD, Australia
| | - Chengye Ding
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Tianqi Guo
- The University of Queensland, School of Dentistry, Herston, QLD, Australia
| | - Houzuo Guo
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
- Department of Oral Implantology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Huajie Yu
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China.
- Fourth Clinical Division, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China.
| |
Collapse
|
166
|
Luo Y, Liu Y, Wang B, Tu X. CHIR99021-Treated Osteocytes with Wnt Activation in 3D-Printed Module Form an Osteogenic Microenvironment for Enhanced Osteogenesis and Vasculogenesis. Int J Mol Sci 2023; 24:ijms24066008. [PMID: 36983081 PMCID: PMC10052982 DOI: 10.3390/ijms24066008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Finding a bone implant that has high bioactivity that can safely drive stem cell differentiation and simulate a real in vivo microenvironment is a challenge for bone tissue engineering. Osteocytes significantly regulate bone cell fate, and Wnt-activated osteocytes can reversely regulate bone formation by regulating bone anabolism, which may improve the biological activity of bone implants. To achieve a safe application, we used the Wnt agonist CHIR99021 (C91) to treat MLO-Y4 for 24 h, in a co-culture with ST2 for 3 days after withdrawal. We found that the expression of Runx2 and Osx increased, promoted osteogenic differentiation, and inhibited adipogenic differentiation in the ST2 cells, and these effects were eliminated by the triptonide. Therefore, we hypothesized that C91-treated osteocytes form an osteogenic microenvironment (COOME). Subsequently, we constructed a bio-instructive 3D printing system to verify the function of COOME in 3D modules that mimic the in vivo environment. Within PCI3D, COOME increased the survival and proliferation rates to as high as 92% after 7 days and promoted ST2 cell differentiation and mineralization. Simultaneously, we found that the COOME-conditioned medium also had the same effects. Therefore, COOME promotes ST2 cell osteogenic differentiation both directly and indirectly. It also promotes HUVEC migration and tube formation, which can be explained by the high expression of Vegf. Altogether, these results indicate that COOME, combined with our independently developed 3D printing system, can overcome the poor cell survival and bioactivity of orthopedic implants and provide a new method for clinical bone defect repair.
Collapse
Affiliation(s)
- Yisheng Luo
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yangxi Liu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Bo Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
167
|
Müller-Heupt LK, Wiesmann-Imilowski N, Schröder S, Groß J, Ziskoven PC, Bani P, Kämmerer PW, Schiegnitz E, Eckelt A, Eckelt J, Ritz U, Opatz T, Al-Nawas B, Synatschke CV, Deschner J. Oxygen-Releasing Hyaluronic Acid-Based Dispersion with Controlled Oxygen Delivery for Enhanced Periodontal Tissue Engineering. Int J Mol Sci 2023; 24:ijms24065936. [PMID: 36983008 PMCID: PMC10059003 DOI: 10.3390/ijms24065936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Periodontitis is a chronic biofilm-associated inflammatory disease of the tooth-supporting tissues that causes tooth loss. It is strongly associated with anaerobic bacterial colonization and represents a substantial global health burden. Due to a local hypoxic environment, tissue regeneration is impaired. Oxygen therapy has shown promising results as a potential treatment of periodontitis, but so far, local oxygen delivery remains a key technical challenge. An oxygen (O2)-releasing hyaluronic acid (HA)-based dispersion with a controlled oxygen delivery was developed. Cell viability of primary human fibroblasts, osteoblasts, and HUVECs was demonstrated, and biocompatibility was tested using a chorioallantoic membrane assay (CAM assay). Suppression of anaerobic growth of Porphyromonas gingivalis was shown using the broth microdilution assay. In vitro assays showed that the O2-releasing HA was not cytotoxic towards human primary fibroblasts, osteoblasts, and HUVECs. In vivo, angiogenesis was enhanced in a CAM assay, although not to a statistically significant degree. Growth of P. gingivalis was inhibited by CaO2 concentrations higher than 256 mg/L. Taken together, the results of this study demonstrate the biocompatibility and selective antimicrobial activity against P. gingivalis for the developed O2-releasing HA-based dispersion and the potential of O2-releasing biomaterials for periodontal tissue regeneration.
Collapse
Affiliation(s)
- Lena Katharina Müller-Heupt
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Nadine Wiesmann-Imilowski
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeck Str. 1, 55131 Mainz, Germany
| | - Sofia Schröder
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Jonathan Groß
- Department of Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Pablo Cores Ziskoven
- Department of Periodontology and Operative Dentistry, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Philipp Bani
- Department of Periodontology and Operative Dentistry, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Peer Wolfgang Kämmerer
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Eik Schiegnitz
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | - Anja Eckelt
- WEE-Solve GmbH, Auf der Burg 6, 55130 Mainz, Germany
| | - John Eckelt
- WEE-Solve GmbH, Auf der Burg 6, 55130 Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Till Opatz
- Department of Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| | | | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany
| |
Collapse
|
168
|
Mabroum H, El Baza H, Ben Youcef H, Oudadesse H, Noukrati H, Barroug A. Design of Antibacterial Apatitic Composite Cement Loaded with Ciprofloxacin: Investigations on the Physicochemical Properties, Release Kinetics, and Antibacterial Activity. Int J Pharm 2023; 637:122861. [PMID: 36948475 DOI: 10.1016/j.ijpharm.2023.122861] [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: 01/12/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
This work aims to develop an injectable and antibacterial composite cement for bone substitution and prevention/treatment of bone infections. This cement is composed of calcium phosphate, calcium carbonate, bioactive glass, sodium alginate, and ciprofloxacin. The effect of ciprofloxacin on the microstructure, chemical composition, setting properties, cohesion, injectability, and compressive strength was investigated. The in vitro drug release kinetics and the antibacterial activity of ciprofloxacin-loaded composites against staphylococcus aureus and Escherichia coli pathogens were investigated. XRD and FTIR analysis demonstrated that the formulated cements are composed of a nanocrystalline carbonated apatite analogous to the mineral part of the bone. The evaluation of the composite cement's properties revealed that the incorporation of 3 and 9 wt% of ciprofloxacin affects the microstructural and physicochemical properties of the cement, resulting in a prolonged setting time, and a slight decrease in injectability and compressive strength. The in vitro drug release study revealed sustained release profiles over 18 days. The amounts of ciprofloxacin released per day (0.2 -15.2 mg/L) depend on the cement composition and the amount of ciprofloxacin incorporated. The antibacterial activity of ciprofloxacin-loaded cement composites attested to their effectiveness to inhibit the growth of Staphylococcus aureus and Escherichia coli.
Collapse
Affiliation(s)
- Hanaa Mabroum
- Cadi Ayyad University, Faculty of Sciences Semlalia, 2390, 40000, Marrakech, Morocco; Institute of Biological Sciences, ISSB, Faculty of medical sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Hamza El Baza
- Institute of Biological Sciences, ISSB, Faculty of medical sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Hicham Ben Youcef
- High Throughput Multidisciplinary Research Laboratory (HTMR), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | | | - Hassan Noukrati
- Institute of Biological Sciences, ISSB, Faculty of medical sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Allal Barroug
- Cadi Ayyad University, Faculty of Sciences Semlalia, 2390, 40000, Marrakech, Morocco; Institute of Biological Sciences, ISSB, Faculty of medical sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| |
Collapse
|
169
|
Deng H, Wang J, An R. Hyaluronic acid-based hydrogels: As an exosome delivery system in bone regeneration. Front Pharmacol 2023; 14:1131001. [PMID: 37007032 PMCID: PMC10063825 DOI: 10.3389/fphar.2023.1131001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Exosomes are extracellular vesicles (EVs) containing various ingredients such as DNA, RNA, lipids and proteins, which play a significant role in intercellular communication. Numerous studies have demonstrated the important role of exosomes in bone regeneration through promoting the expression of osteogenic-related genes and proteins in mesenchymal stem cells. However, the low targeting ability and short circulating half-life of exosomes limited their clinical application. In order to solve those problems, different delivery systems and biological scaffolds have been developed. Hydrogel is a kind of absorbable biological scaffold composed of three-dimensional hydrophilic polymers. It not only has excellent biocompatibility and superior mechanical strength but can also provide a suitable nutrient environment for the growth of the endogenous cells. Thus, the combination between exosomes and hydrogels can improve the stability and maintain the biological activity of exosomes while achieving the sustained release of exosomes in the bone defect sites. As an important component of the extracellular matrix (ECM), hyaluronic acid (HA) plays a critical role in various physiological and pathological processes such as cell differentiation, proliferation, migration, inflammation, angiogenesis, tissue regeneration, wound healing and cancer. In recent years, hyaluronic acid-based hydrogels have been used as an exosome delivery system for bone regeneration and have displayed positive effects. This review mainly summarized the potential mechanism of HA and exosomes in promoting bone regeneration and the application prospects and challenges of hyaluronic acid-based hydrogels as exosome delivery devices in bone regeneration.
Collapse
Affiliation(s)
| | | | - Ran An
- *Correspondence: Jiecong Wang, ; Ran An,
| |
Collapse
|
170
|
Recent progressions in biomedical and pharmaceutical applications of chitosan nanoparticles: A comprehensive review. Int J Biol Macromol 2023; 231:123354. [PMID: 36681228 DOI: 10.1016/j.ijbiomac.2023.123354] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
Nowadays, the most common approaches in the prognosis, diagnosis, and treatment of diseases are along with undeniable limitations. Thus, the ever-increasing need for using biocompatible natural materials and novel practical modalities is required. Applying biomaterials, such as chitosan nanoparticles (CS NPs: FDA-approved long-chain polymer of N-acetyl-glucosamine and D-glucosamine for some pharmaceutical applications), can serve as an appropriate alternative to overcome these limitations. Recently, the biomedical applications of CS NPs have extensively been investigated. These NPs and their derivatives can not only prepare through different physical and chemical approaches but also modify with various molecules and bioactive materials. The potential properties of CS NPs, such as biocompatibility, biodegradability, serum stability, solubility, non-immunogenicity, anti-inflammatory properties, appropriate pharmacokinetics and pharmacodynamics, and so forth, have made them excellent candidates for biomedical applications. Therefore, CS NPs have efficiently applied for various biomedical applications, like regenerative medicine and tissue engineering, biosensors for the detection of microorganisms, and drug delivery systems (DDS) for the suppression of diseases. These NPs possess a high level of biosafety. In summary, CS NPs have the potential ability for biomedical and clinical applications, and it would be remarkably beneficial to develop new generations of CS-based material for the future of medicine.
Collapse
|
171
|
Zhang X, Zhao Q, Zhou N, Liu Y, Qin K, Buhl EM, Wang X, Hildebrand F, Balmayor ER, Greven J. Osteoblast derived extracellular vesicles induced by dexamethasone: A novel biomimetic tool for enhancing osteogenesis in vitro. Front Bioeng Biotechnol 2023; 11:1160703. [PMID: 37020508 PMCID: PMC10069331 DOI: 10.3389/fbioe.2023.1160703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Extracellular vesicles (EVs) are newly appreciated communicators involved in intercellular crosstalk, and have emerged as a promising biomimetic tool for bone tissue regeneration, overcoming many of the limitations associated with cell-based therapies. However, the significance of osteoblast-derived extracellular vesicles on osteogenesis has not been fully established. In this present study, we aim to investigate the therapeutic potential of extracellular vesicles secreted from consecutive 14 days of dexamethasone-stimulated osteoblasts (OB-EVDex) to act as a biomimetic tool for regulating osteogenesis, and to elucidate the underlying mechanisms. OB-EVdex treated groups are compared to the clinically used osteo-inductor of BMP-2 as control. Our findings revealed that OB-EVDex have a typical bilayer membrane nanostructure of, with an average diameter of 178 ± 21 nm, and that fluorescently labeled OB-EVDex were engulfed by osteoblasts in a time-dependent manner. The proliferation, attachment, and viability capacities of OB-EVDex-treated osteoblasts were significantly improved when compared to untreated cells, with the highest proliferative rate observed in the OB-EVDex + BMP-2 group. Notably, combinations of OB-EVDex and BMP-2 markedly promoted osteogenic differentiation by positively upregulating osteogenesis-related gene expression levels of RUNX2, BGLAP, SPP1, SPARC, Col 1A1, and ALPL relative to BMP-2 or OB-EVDex treatment alone. Mineralization assays also showed greater pro-osteogenic potency after combined applications of OB-EVDex and BMP-2, as evidenced by a notable increase in mineralized nodules (calcium deposition) revealed by Alkaline Phosphatase (ALP), Alizarin Red Alizarin Red staining (ARS), and von Kossa staining. Therefore, our findings shed light on the potential of OB-EVDex as a new therapeutic option for enhancing osteogenesis.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
- *Correspondence: Xing Zhang, ; Qun Zhao, ; Xinhong Wang,
| | - Qun Zhao
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
- *Correspondence: Xing Zhang, ; Qun Zhao, ; Xinhong Wang,
| | - Nan Zhou
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Yu Liu
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Kang Qin
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology and Medical Clinic II, University Hospital RWTH Aachen, Aachen, Germany
| | - Xinhong Wang
- Department of Orthopedics, The Affliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huai’an, Jiangsu, China
- *Correspondence: Xing Zhang, ; Qun Zhao, ; Xinhong Wang,
| | - Frank Hildebrand
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Elizabeth R. Balmayor
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Johannes Greven
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
172
|
Ye J, Liu N, Li Z, Liu L, Zheng M, Wen X, Wang N, Xu Y, Sun B, Zhou Q. Injectable, Hierarchically Degraded Bioactive Scaffold for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:11458-11473. [PMID: 36827205 DOI: 10.1021/acsami.2c18824] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Bioactive materials play vital roles in the repair of critical bone defects. However, bone tissue engineering and regenerative medicine are still challenged by the need to repair bone defects evenly and completely. In this study, we functionally simulated the natural creeping substitution process of autologous bone repair by constructing an injectable, hierarchically degradable bioactive scaffold with a composite hydrogel, decalcified bone matrix (DBM) particles, and bone morphogenetic protein 2. This composite scaffold exhibited superior mechanical properties. The scaffold promoted cell proliferation and osteogenic differentiation through multiple signaling pathways. The hierarchical degradation rates of the crosslinked hydrogel and DBM particles accelerated tissue ingrowth and bone formation with a naturally woven bone-like structure in vivo. In the rat calvarial critical defect repair model, the composite scaffold provided even and complete repair of the entire defect area while also integrating the new and host bone effectively. Our results indicate that this injectable, hierarchically degradable bioactive scaffold promotes bone regeneration and provides a promising strategy for evenly and completely repairing the bone defects.
Collapse
Affiliation(s)
- Jixing Ye
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ningyuan Liu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Zongxin Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Liehua Liu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ming Zheng
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xueping Wen
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Nan Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yanqin Xu
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Biemin Sun
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Qiang Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| |
Collapse
|
173
|
Heng BC, Bai Y, Li X, Meng Y, Lu Y, Zhang X, Deng X. The bioelectrical properties of bone tissue. Animal Model Exp Med 2023; 6:120-130. [PMID: 36856186 PMCID: PMC10158952 DOI: 10.1002/ame2.12300] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/18/2022] [Indexed: 03/02/2023] Open
Abstract
Understanding the bioelectrical properties of bone tissue is key to developing new treatment strategies for bone diseases and injuries, as well as improving the design and fabrication of scaffold implants for bone tissue engineering. The bioelectrical properties of bone tissue can be attributed to the interaction of its various cell lineages (osteocyte, osteoblast and osteoclast) with the surrounding extracellular matrix, in the presence of various biomechanical stimuli arising from routine physical activities; and is best described as a combination and overlap of dielectric, piezoelectric, pyroelectric and ferroelectric properties, together with streaming potential and electro-osmosis. There is close interdependence and interaction of the various electroactive and electrosensitive components of bone tissue, including cell membrane potential, voltage-gated ion channels, intracellular signaling pathways, and cell surface receptors, together with various matrix components such as collagen, hydroxyapatite, proteoglycans and glycosaminoglycans. It is the remarkably complex web of interactive cross-talk between the organic and non-organic components of bone that define its electrophysiological properties, which in turn exerts a profound influence on its metabolism, homeostasis and regeneration in health and disease. This has spurred increasing interest in application of electroactive scaffolds in bone tissue engineering, to recapitulate the natural electrophysiological microenvironment of healthy bone tissue to facilitate bone defect repair.
Collapse
Affiliation(s)
- Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, PR China.,School of Medical and Life Sciences, Sunway University, Subang Jaya, Malaysia
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yanze Meng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China.,National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China.,National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| |
Collapse
|
174
|
Yu W, Wang HL, Zhang J, Yin C. The effects of epigenetic modifications on bone remodeling in age-related osteoporosis. Connect Tissue Res 2023; 64:105-116. [PMID: 36271658 DOI: 10.1080/03008207.2022.2120392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE As the population ages, there is an increased risk of fracture and morbidity diseases associated with aging, such as age-related osteoporosis and other bone diseases linked to aging skeletons. RESULTS Several bone-related cells, including multipotent bone mesenchymal stem cells, osteoblasts that form bone tissue, and osteoclasts that break it down, are in symbiotic relationships throughout life. Growing evidence indicates that epigenetic modifications of cells caused by aging contribute to compromised bone remodeling and lead to osteoporosis. A number of epigenetic mechanisms are at play, including DNA/RNA modifications, histone modifications, microRNAs (miRNAs), and long noncoding RNAs (lncRNAs), as well as chromatin remodeling. CONCLUSION In this review, we summarized the epigenetic modifications of different bone-related cells during the development and progression of osteoporosis associated with aging. Additionally, we described a compensatory recovery mechanism under epigenetic regulation that may lead to new strategies for regulating bone remodeling in age-related osteoporosis.
Collapse
Affiliation(s)
- Wenyue Yu
- School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - He-Ling Wang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Jianying Zhang
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Chengcheng Yin
- School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| |
Collapse
|
175
|
Xie S, Choudhari S, Wu CL, Abramson K, Corcoran D, Gregory SG, Thimmapuram J, Guilak F, Little D. Aging and obesity prime the methylome and transcriptome of adipose stem cells for disease and dysfunction. FASEB J 2023; 37:e22785. [PMID: 36794668 PMCID: PMC10561192 DOI: 10.1096/fj.202201413r] [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: 08/31/2022] [Revised: 12/20/2022] [Accepted: 01/09/2023] [Indexed: 02/17/2023]
Abstract
The epigenome of stem cells occupies a critical interface between genes and environment, serving to regulate expression through modification by intrinsic and extrinsic factors. We hypothesized that aging and obesity, which represent major risk factors for a variety of diseases, synergistically modify the epigenome of adult adipose stem cells (ASCs). Using integrated RNA- and targeted bisulfite-sequencing in murine ASCs from lean and obese mice at 5- and 12-months of age, we identified global DNA hypomethylation with either aging or obesity, and a synergistic effect of aging combined with obesity. The transcriptome of ASCs in lean mice was relatively stable to the effects of age, but this was not true in obese mice. Functional pathway analyses identified a subset of genes with critical roles in progenitors and in diseases of obesity and aging. Specifically, Mapt, Nr3c2, App, and Ctnnb1 emerged as potential hypomethylated upstream regulators in both aging and obesity (AL vs. YL and AO vs. YO), and App, Ctnnb1, Hipk2, Id2, and Tp53 exhibited additional effects of aging in obese animals. Furthermore, Foxo3 and Ccnd1 were potential hypermethylated upstream regulators of healthy aging (AL vs. YL), and of the effects of obesity in young animals (YO vs. YL), suggesting that these factors could play a role in accelerated aging with obesity. Finally, we identified candidate driver genes that appeared recurrently in all analyses and comparisons undertaken. Further mechanistic studies are needed to validate the roles of these genes capable of priming ASCs for dysfunction in aging- and obesity-associated pathologies.
Collapse
Affiliation(s)
- Shaojun Xie
- Bioinformatics Core, Purdue University, 1022 Young Hall, 155 S. Grant Street, West Lafayette, IN 47907
| | - Sulbha Choudhari
- Bioinformatics Core, Purdue University, 1022 Young Hall, 155 S. Grant Street, West Lafayette, IN 47907
- Advanced Biomedical Computational Science, Bioinformatics and Computational Science, Frederick National Laboratory for Cancer Research, 8560 Progress Drive, Frederick, MD 2170
| | - Chia-Lung Wu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, 14611
| | - Karen Abramson
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701
| | - David Corcoran
- Genomic Analysis and Bioinformatics Shared Resource, Duke Center for Genomic and Computational Biology, 101 Science Drive, Duke University Medical Center Box 3382, Durham, NC 27708
- Lineberger Bioinformatics Core, 5200 Marsico Hall, University of North Carolina-Chapel Hill, Chapel Hill, NC 27516
| | - Simon G. Gregory
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701
- Department of Neurology, Duke University School of Medicine, 311 Research Drive, Durham, NC 27710
| | - Jyothi Thimmapuram
- Bioinformatics Core, Purdue University, 1022 Young Hall, 155 S. Grant Street, West Lafayette, IN 47907
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, 4515 McKinley Ave., St. Louis, MO 63110
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis Missouri 63110
| | - Dianne Little
- Departments of Basic Medical Sciences and Biomedical Engineering, Purdue University, 2186 Lynn Hall, 625 Harrison St, West Lafayette, IN 47907-2026
| |
Collapse
|
176
|
Miao Y, Chen Y, Luo J, Liu X, Yang Q, Shi X, Wang Y. Black phosphorus nanosheets-enabled DNA hydrogel integrating 3D-printed scaffold for promoting vascularized bone regeneration. Bioact Mater 2023; 21:97-109. [PMID: 36093326 PMCID: PMC9417961 DOI: 10.1016/j.bioactmat.2022.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/10/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
The classical 3D-printed scaffolds have attracted enormous interests in bone regeneration due to the customized structural and mechanical adaptability to bone defects. However, the pristine scaffolds still suffer from the absence of dynamic and bioactive microenvironment that is analogous to natural extracellular matrix (ECM) to regulate cell behaviour and promote tissue regeneration. To address this challenge, we develop a black phosphorus nanosheets-enabled dynamic DNA hydrogel to integrate with 3D-printed scaffold to build a bioactive gel-scaffold construct to achieve enhanced angiogenesis and bone regeneration. The black phosphorus nanosheets reinforce the mechanical strength of dynamic self-healable hydrogel and endow the gel-scaffold construct with preserved protein binding to achieve sustainable delivery of growth factor. We further explore the effects of this activated construct on both human umbilical vein endothelial cells (HUVECs) and mesenchymal stem cells (MSCs) as well as in a critical-sized rat cranial defect model. The results confirm that the gel-scaffold construct is able to promote the growth of mature blood vessels as well as induce osteogenesis to promote new bone formation, indicating that the strategy of nano-enabled dynamic hydrogel integrated with 3D-printed scaffold holds great promise for bone tissue engineering. Therapeutic VEGF-engineered black phosphorus nanosheets are incorporated into DNA hydrogels. Nano-enabled DNA hydrogel integrating with 3D-printed scaffold builds gel-scaffold construct. Gel-scaffold construct upregulates the expression of genes and proteins related to angiogenesis and osteogenesis. Gel-scaffold construct accelerates the formation of early vascular network and new bone tissue.
Collapse
Affiliation(s)
- Yali Miao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China.
| | - Jinshui Luo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Xiao Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Qian Yang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Xuetao Shi
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China.
| | - Yingjun Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China.
| |
Collapse
|
177
|
Okutan AE, Gürün E, Surucu S, Kehribar L, Mahiroğulları M. Morphological Changes in the Tibial Tunnel After ACL Reconstruction With the Outside-In Technique and Adjustable Suspensory Fixation. Orthop J Sports Med 2023; 11:23259671231155153. [PMID: 36875338 PMCID: PMC9983096 DOI: 10.1177/23259671231155153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/08/2023] [Indexed: 03/05/2023] Open
Abstract
Background Anterior cruciate ligament reconstruction (ACLR) using the complete tibial tunnel technique and adjustable-loop cortical suspensory fixation is known to leave a "dead space" that holds the loop device in the tibial tunnel. The consequence of the dead space and its effect on graft healing are still uncertain. Purpose To investigate morphological changes in the tibial tunnel and their effect on graft healing, and to identify factors affecting bone healing in the tibial loop tunnel after ACLR with a quadrupled semitendinosus tendon autograft using adjustable suspensory fixation. Study Design Case series; Level of evidence, 4. Methods Included were 48 patients (34 male, 14 female; mean age, 25.2 ± 5.6 years) who underwent ACLR with a quadrupled semitendinosus tendon autograft using adjustable suspensory fixation. To evaluate tibial tunnel morphology, computed tomography was performed at 1 day and 6 months postoperatively. At 1 year postoperatively, graft healing was assessed on magnetic resonance imaging using the graft signal-to-noise quotient (SNQ). Multivariate regression and correlation analyses were performed to determine any associations between volumetric changes in bone healing and operative variables. Results At 6 months after ACLR, a mean of 63.2% of the tibial loop tunnel was filled by bone. Multivariate regression analysis showed that remnant preservation was significantly associated with the loop tunnel filling rate (P < .001). At 1 year after ACLR, the tibial loop tunnel was almost completely closed (98.5%). There were no correlations between loop tunnel volume and graft integration or graft SNQ. A significant but weak correlation was found between graft tunnel volume and intratunnel graft SNQ (P = .10) as well as integration grade in the tibial tunnel (P = .30). Conclusion Excellent bone filling in the tibial loop tunnel was seen at 1 year after ACLR. Remnant preservation was significantly associated with the loop tunnel filling rate. A weak correlation was found between graft tunnel volume and intratunnel graft SNQ as well as integration grade in the tibial tunnel.
Collapse
Affiliation(s)
- Ahmet Emin Okutan
- Department of Orthopedics and Traumatology, Faculty of Medicine, Samsun University, Samsun, Turkey
| | - Enes Gürün
- Department of Radiology, Faculty of Medicine, Samsun University, Samsun, Turkey
| | - Serkan Surucu
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Lokman Kehribar
- Department of Orthopedics and Traumatology, Faculty of Medicine, Samsun University, Samsun, Turkey
| | - Mahir Mahiroğulları
- Department of Orthopedics and Traumatology, Memorial Sisli Hospital, Istanbul, Turkey
| |
Collapse
|
178
|
Hang R, Wang Z, Wang H, Zhang Y, Zhao Y, Bai L, Yao X. Matrix stiffness-induced platelet activation determines immunomodulation of macrophages. BIOMATERIALS ADVANCES 2023; 148:213356. [PMID: 36848742 DOI: 10.1016/j.bioadv.2023.213356] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/11/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023]
Abstract
Although various bone defect repair materials have been used clinically, the influence of the material properties on bone repair and regeneration as well as the underlying mechanisms are not fully understood. We hypothesize that the material stiffness affects initial platelet activation during hemostasis phase, which in turn mediates subsequent osteoimmunomodulation of macrophages, finally determining clinical outcomes. To verify the hypothesis, the present work used polyacrylamide hydrogels with different stiffness (10, 70, and 260 kPa) as model materials to investigate matrix stiffness induced platelet activation behavior and its mediation on osteoimmunomodulation of macrophages. The results showed that the matrix stiffness was positively related with activation degree of platelets. However, the extracts of platelets incubated on middle-stiff matrix polarized macrophages to pro-healing M2 phenotype when compared with that on soft and stiff matrixes. ELISA results showed when compared with that on soft and stiff matrixes, the platelets incubated on middle-stiff matrix released more TGF-β and PGE2, both of which could polarize macrophages to M2 phenotype. The M2 macrophages could promote angiogenesis of endothelial cells and osteogenesis of bone marrow mesenchymal stem cells, two important and coupled processes involved in bone repair and regeneration. These findings suggest bone repair materials with 70 kPa stiffness can mediate proper platelet activation, which can polarize macrophages to pro-healing M2 phenotype, potentially contributing to bone repair and regeneration.
Collapse
Affiliation(s)
- Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Zhenlong Wang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Hui Wang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi Xinhua Chemical Defense Equipment Research Institute Co., Ltd, Taiyuan 030008, China
| | - Yi Zhang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yuyu Zhao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
179
|
Liu J, Yang L, Liu K, Gao F. Hydrogel scaffolds in bone regeneration: Their promising roles in angiogenesis. Front Pharmacol 2023; 14:1050954. [PMID: 36860296 PMCID: PMC9968752 DOI: 10.3389/fphar.2023.1050954] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Bone tissue engineering (BTE) has become a hopeful potential treatment strategy for large bone defects, including bone tumors, trauma, and extensive fractures, where the self-healing property of bone cannot repair the defect. Bone tissue engineering is composed of three main elements: progenitor/stem cells, scaffold, and growth factors/biochemical cues. Among the various biomaterial scaffolds, hydrogels are broadly used in bone tissue engineering owing to their biocompatibility, controllable mechanical characteristics, osteoconductive, and osteoinductive properties. During bone tissue engineering, angiogenesis plays a central role in the failure or success of bone reconstruction via discarding wastes and providing oxygen, minerals, nutrients, and growth factors to the injured microenvironment. This review presents an overview of bone tissue engineering and its requirements, hydrogel structure and characterization, the applications of hydrogels in bone regeneration, and the promising roles of hydrogels in bone angiogenesis during bone tissue engineering.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Lili Yang
- Department of Spinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kexin Liu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Gao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China,*Correspondence: Feng Gao,
| |
Collapse
|
180
|
Clancy J, Hoffmann CS, Pickett BE. Transcriptomics secondary analysis of severe human infection with SARS-CoV-2 identifies gene expression changes and predicts three transcriptional biomarkers in leukocytes. Comput Struct Biotechnol J 2023; 21:1403-1413. [PMID: 36785619 PMCID: PMC9908618 DOI: 10.1016/j.csbj.2023.02.003] [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: 08/18/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
SARS-CoV-2 is the causative agent of COVID-19, which has greatly affected human health since it first emerged. Defining the human factors and biomarkers that differentiate severe SARS-CoV-2 infection from mild infection has become of increasing interest to clinicians. To help address this need, we retrieved 269 public RNA-seq human transcriptome samples from GEO that had qualitative disease severity metadata. We then subjected these samples to a robust RNA-seq data processing workflow to calculate gene expression in PBMCs, whole blood, and leukocytes, as well as to predict transcriptional biomarkers in PBMCs and leukocytes. This process involved using Salmon for read mapping, edgeR to calculate significant differential expression levels, and gene ontology enrichment using Camera. We then performed a random forest machine learning analysis on the read counts data to identify genes that best classified samples based on the COVID-19 severity phenotype. This approach produced a ranked list of leukocyte genes based on their Gini values that includes TGFBI, TTYH2, and CD4, which are associated with both the immune response and inflammation. Our results show that these three genes can potentially classify samples with severe COVID-19 with accuracy of ∼88% and an area under the receiver operating characteristic curve of 92.6--indicating acceptable specificity and sensitivity. We expect that our findings can help contribute to the development of improved diagnostics that may aid in identifying severe COVID-19 cases, guide clinical treatment, and improve mortality rates.
Collapse
|
181
|
Prospective use of amniotic mesenchymal stem cell metabolite products for tissue regeneration. J Biol Eng 2023; 17:11. [PMID: 36759827 PMCID: PMC9912508 DOI: 10.1186/s13036-023-00331-1] [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: 09/09/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Chronic disease can cause tissue and organ damage constituting the largest obstacle to therapy which, in turn, reduces patients' quality-adjusted life-year. Degenerative diseases such as osteoporosis, Alzheimer's disease, Parkinson's disease, and infectious conditions such as hepatitis, cause physical injury to organs. Moreover, damage resulting from chronic conditions such as diabetes can also culminate in the loss of organ function. In these cases, organ transplantation constitutes the therapy of choice, despite the associated problems of immunological rejection, potential disease transmission, and high morbidity rates. Tissue regeneration has the potential to heal or replace tissues and organs damaged by age, disease, or trauma, as well as to treat disabilities. Stem cell use represents an unprecedented strategy for these therapies. However, product availability and mass production remain challenges. A novel therapeutic alternative involving amniotic mesenchymal stem cell metabolite products (AMSC-MP) has been developed using metabolites from stem cells which contain cytokines and growth factors. Its potential role in regenerative therapy has recently been explored, enabling broad pharmacological applications including various gastrointestinal, lung, bladder and renal conditions, as well as the treatment of bone wounds, regeneration and skin aging due to its low immunogenicity and anti-inflammatory effects. The various kinds of growth factors present in AMSC-MP, namely bFGF, VEGF, TGF-β, EGF and KGF, have their respective functions and activities. Each growth factor is formed by different proteins resulting in molecules with various physicochemical properties and levels of stability. This knowledge will assist in the manufacture and application of AMSC-MP as a therapeutic agent.
Collapse
|
182
|
Li Z, Xue L, Wang P, Ren X, Zhang Y, Wang C, Sun J. Biological Scaffolds Assembled with Magnetic Nanoparticles for Bone Tissue Engineering: A Review. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1429. [PMID: 36837058 PMCID: PMC9961196 DOI: 10.3390/ma16041429] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are widely used in bone tissue engineering because of their unique physical and chemical properties and their excellent biocompatibility. Under the action of a magnetic field, SPIONs loaded in a biological scaffold can effectively promote osteoblast proliferation, differentiation, angiogenesis, and so on. SPIONs have very broad application prospects in bone repair, bone reconstruction, bone regeneration, and other fields. In this paper, several methods for forming biological scaffolds via the biological assembly of SPIONs are reviewed, and the specific applications of these biological scaffolds in bone tissue engineering are discussed.
Collapse
Affiliation(s)
- Zheng Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Bioscience and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Le Xue
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Bioscience and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Peng Wang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Bioscience and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Xueqian Ren
- Clinical Medical Engineering Department, The Affiliated Zhongda Hospital of Southeast University Medical School, Nanjing 210009, China
| | - Yunyang Zhang
- Center of Modern Analysis, Nanjing University, Nanjing 210000, China
| | - Chuan Wang
- Naval Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Jianfei Sun
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Bioscience and Medical Engineering, Southeast University, Nanjing 210009, China
| |
Collapse
|
183
|
Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells 2023; 12:cells12040521. [PMID: 36831188 PMCID: PMC9954309 DOI: 10.3390/cells12040521] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.
Collapse
|
184
|
Hao D, Liu R, Fernandez TG, Pivetti C, Jackson JE, Kulubya ES, Jiang HJ, Ju HY, Liu WL, Panitch A, Lam KS, Leach JK, Farmer DL, Wang A. A bioactive material with dual integrin-targeting ligands regulates specific endogenous cell adhesion and promotes vascularized bone regeneration in adult and fetal bone defects. Bioact Mater 2023; 20:179-193. [PMID: 35663336 PMCID: PMC9160290 DOI: 10.1016/j.bioactmat.2022.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 12/18/2022] Open
Abstract
Significant progress has been made in designing bone materials capable of directing endogenous cells to promote vascularized bone regeneration. However, current strategies lack regulation of the specific endogenous cell populations for vascularized bone regeneration, thus leading to adverse tissue formation and decreased regenerative efficiency. Here, we engineered a biomaterial to regulate endogenous cell adhesion and promote vascularized bone regeneration. The biomaterial works by presenting two synthetic ligands, LLP2A and LXW7, explicitly targeting integrins α4β1 and αvβ3, respectively, expressed on the surfaces of the cells related to bone formation and vascularization, such as mesenchymal stem cells (MSCs), osteoblasts, endothelial progenitor cells (EPCs), and endothelial cells (ECs). In vitro, the LLP2A/LXW7 modified biomaterial improved the adhesion of MSCs, osteoblasts, EPCs, and ECs via integrin α4β1 and αvβ3, respectively. In an adult rat calvarial bone defect model, the LLP2A/LXW7 modified biomaterial enhanced bone formation and vascularization by synergistically regulating endogenous cells with osteogenic and angiogenic potentials, such as DLX5+ cells, osteocalcin+ cells, CD34+/CD45- cells and CD31+ cells. In a fetal sheep spinal bone defect model, the LLP2A/LXW7 modified biomaterial augmented bone formation and vascularization without any adverse effects. This innovative biomaterial offers an off-the-shelf, easy-to-use, and biologically safe product suitable for vascularized bone regeneration in both fetal and adult disease environments. Two integrin-binding ligands for constructing vascularized bone biomaterial. Extracellular matrix (ECM)-mimicking collagen-based biomaterial with specific integrin binding sites for cell adhesion. Biomaterial regulates adhesion of endogenous stem cells with osteogenic and angiogenic potentials. Biomaterial promotes vascularized bone formation in adult and fetal bone defects without safety issues. An easy-to-make and off-the-shelf biomaterial for treatment of clinical bone diseases.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Tomas Gonzalez Fernandez
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Christopher Pivetti
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Jordan Elizabeth Jackson
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Edwin Samuel Kulubya
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Hong-Jiang Jiang
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Hai-Yang Ju
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Wen-Liang Liu
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Alyssa Panitch
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
- Corresponding author. Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA, 95817, USA.
| |
Collapse
|
185
|
Wang X, Zheng W, Bai Z, Huang S, Jiang K, Liu H, Liu L. Mimicking bone matrix through coaxial electrospinning of core-shell nanofibrous scaffold for improving neurogenesis bone regeneration. BIOMATERIALS ADVANCES 2023; 145:213246. [PMID: 36549151 DOI: 10.1016/j.bioadv.2022.213246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/22/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
There is a significant clinical demand for bone repair materials with high efficacy. This study was designed to fabricate nanofibrous scaffolds to promote bone defect regeneration using magnesium doped mesoporous bioactive glass (MBG), a fusion protein Osteocalcin-Osteopontin-Biglycan (OOB), silk fibroin (SF) and nerve growth factor (NGF) for facilitating accelerated bone formation. We found that MBG adsorbed with OOB (OOB@MBG) as core, and SF adsorbed with NGF (SF@NGF) as shell to fabricate the nanofibrous scaffolds (OOB@MBG/NGF@SF) through coaxial electrospinning. OOB@MBG/NGF@SF scaffolds could effectively mimic the component and structure of bone matrix. Interestingly, we observed that OOB@MBG/NGF@SF scaffolds could substantially promote bone mesenchymal stem cells (BMSCs) osteogenesis through stimulating Erk1/2 activated Runx2 and mTOR pathway, and it could also activate the expression level of various osteogenic marker genes. Intriguingly, OOB@MBG/NGF@SF scaffolds could also enhance BMSCs induced neural differentiation cells differentiated into neuron, and activate the expression of the different neuron specific marker genes. Moreover, it was found that OOB@MBG/NGF@SF scaffolds accelerated bone regeneration with neurogenesis, and new neurons were formed in Haversian canal in vivo. Consistent with these observations, we found that Erk1/2 and mTOR signaling pathways also regulated osteogenesis with the neurogenesis process from RNA sequencing result. Overall, our findings provided novel evidence suggesting that OOB@MBG/NGF@SF scaffolds could function as a potential biomaterial in accelerating bone defect regeneration with neurogenesis, as well as in recovering the motor ability and improving the quality of life of patients.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China.
| | - Weijia Zheng
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China
| | - Zhenzu Bai
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China
| | - Shan Huang
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China
| | - Kai Jiang
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China
| | - Haoming Liu
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China
| | - Long Liu
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, PR China
| |
Collapse
|
186
|
Poly(lactic acid) and Nanocrystalline Cellulose Methacrylated Particles for Preparation of Cryogelated and 3D-Printed Scaffolds for Tissue Engineering. Polymers (Basel) 2023; 15:polym15030651. [PMID: 36771954 PMCID: PMC9920993 DOI: 10.3390/polym15030651] [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: 12/09/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023] Open
Abstract
Different parts of bones possess different properties, such as the capacity for remodeling cell content, porosity, and protein composition. For various traumatic or surgical tissue defects, the application of tissue-engineered constructs seems to be a promising strategy. Despite significant research efforts, such constructs are still rarely available in the clinic. One of the reasons is the lack of resorbable materials, whose properties can be adjusted according to the intended tissue or tissue contacts. Here, we present our first results on the development of a toolbox, by which the scaffolds with easily tunable mechanical and biological properties could be prepared. Biodegradable poly(lactic acid) and nanocrystalline cellulose methacrylated particles were obtained, characterized, and used for preparation of three-dimensional scaffolds via cryogelation and 3D printing approaches. The composition of particles-based ink for 3D printing was optimized in order to allow formation of stable materials. Both the modified-particle cytotoxicity and the matrix-supported cell adhesion were evaluated and visualized in order to confirm the perspectives of materials application.
Collapse
|
187
|
Bie H, Chen H, Shan L, Tan CY, Al-Furjan MSH, Ramesh S, Gong Y, Liu YF, Zhou RG, Yang W, Wang H. 3D Printing and Performance Study of Porous Artificial Bone Based on HA-ZrO 2-PVA Composites. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1107. [PMID: 36770115 PMCID: PMC9919799 DOI: 10.3390/ma16031107] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
An ideal artificial bone implant should have similar mechanical properties and biocompatibility to natural bone, as well as an internal structure that facilitates stomatal penetration. In this work, 3D printing was used to fabricate and investigate artificial bone composites based on HA-ZrO2-PVA. The composites were proportionally configured using zirconia (ZrO2), hydroxyapatite (HA) and polyvinyl alcohol (PVA), where the ZrO2 played a toughening role and PVA solution served as a binder. In order to obtain the optimal 3D printing process parameters for the composites, a theoretical model of the extrusion process of the composites was first established, followed by the optimization of various parameters including the spray head internal diameter, extrusion pressure, extrusion speed, and extrusion line width. The results showed that, at the optimum parameters of a spray head diameter of 0.2 mm, extrusion pressure values ranging from 1-3 bar, a line spacing of 0.8-1.5 mm, and a spray head displacement range of 8-10 mm/s, a better structure of biological bone scaffolds could be obtained. The mechanical tests performed on the scaffolds showed that the elastic modulus of the artificial bone scaffolds reached about 174 MPa, which fulfilled the biomechanical requirements of human bone. According to scanning electron microscope observation of the scaffold sample, the porosity of the scaffold sample was close to 65%, which can well promote the growth of chondrocytes and angiogenesis. In addition, c5.18 chondrocytes were used to verify the biocompatibility of the composite materials, and the cell proliferation was increased by 100% when compared with that of the control group. The results showed that the composite has good biocompatibility.
Collapse
Affiliation(s)
- Hongling Bie
- Artificial Intelligence Applications College, Shanghai Urban Construction Vocational College, Shanghai 201415, China
| | - Honghao Chen
- School of Mechanical Engineering, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Lijun Shan
- Center of Advanced Manufacturing and Material Processing, Department of Mechanical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - C. Y. Tan
- Center of Advanced Manufacturing and Material Processing, Department of Mechanical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - M. S. H. Al-Furjan
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
- Collaborative Innovation Center of High-End Laser Manufacturing Equipment (National “2011 Plan”), Zhejiang University of Technology, Hangzhou 310023, China
| | - S. Ramesh
- Center of Advanced Manufacturing and Material Processing, Department of Mechanical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Youping Gong
- School of Mechanical Engineering, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Y. F. Liu
- Collaborative Innovation Center of High-End Laser Manufacturing Equipment (National “2011 Plan”), Zhejiang University of Technology, Hangzhou 310023, China
- Key Laboratory of E&M, Zhejiang University of Technology, Ministry of Education & Zhejiang Province, Hangzhou 310023, China
| | - R. G. Zhou
- School of Mechanical Engineering, Hangzhou Dianzi University, Hangzhou 310018, China
- Wenzhou Institute of Hangzhou Dianzi University, 3-4/F, Building B, Zhejiang Yungu, Nanyang Avenue, Yaoxi Street, Hangzhou 325038, China
| | - Weibo Yang
- Zhejiang Guanlin Machinery Limited Company, Huzhou 313300, China
| | - Honghua Wang
- Zhejiang Guanlin Machinery Limited Company, Huzhou 313300, China
| |
Collapse
|
188
|
Xiao X, Liu Z, Shu R, Wang J, Zhu X, Bai D, Lin H. Periodontal bone regeneration with a degradable thermoplastic HA/PLCL bone graft. J Mater Chem B 2023; 11:772-786. [PMID: 36444735 DOI: 10.1039/d2tb02123d] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Strategic bone grafts are required to regenerate periodontal bone defects owing to limited self-healing. Current bioceramic particle or deproteinized bovine bone (DBB) products are not able to ideally meet clinical requirements, such as insufficient operability and slow degradation rates. Herein, a strong-interacted bone graft was designed and synthesized by modifying hydroxyapatite (HA) with a lactide-caprolactone copolymer (PLCL) to improve component homogeneity and mechanical properties. The physical-chemical analysis indicated that HA particles were homogenously distributed in HA/PLCL bone grafts, possessed outstanding thermoplasticity, and facilitated clinic operability and initial mechanical support. The in vitro study suggested that HA/PLCL bone graft degraded in a spatiotemporal model. Micropores were formed on the non-porous surface at the beginning, and interconnected porous structures were gradually generated. Furthermore, HA/PLCL bone grafts exhibited excellent biocompatibility and osteogenic ability as revealed in vitro cell culture and in vivo animal experiments. When applied to rat periodontal bone defects, the HA/PLCL bone graft showed a non-inferior bone regeneration compared to the commercial DBB. This study proposes a potential bone graft for periodontal bone repair with thermoplastic, spatiotemporal degraded, and osteogenic characteristics.
Collapse
Affiliation(s)
- Xueling Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Zhanhong Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610064, China
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jiangyue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610064, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Hai Lin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610064, China
| |
Collapse
|
189
|
The Current Status, Prospects, and Challenges of Shape Memory Polymers Application in Bone Tissue Engineering. Polymers (Basel) 2023; 15:polym15030556. [PMID: 36771857 PMCID: PMC9920657 DOI: 10.3390/polym15030556] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/28/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Bone defects can occur after severe trauma, infection, or bone tumor resection surgery, which requires grafting to repair the defect when it reaches a critical size, as the bone's self-healing ability is insufficient to complete the bone repair. Natural bone grafts or artificial bone grafts, such as bioceramics, are currently used in bone tissue engineering, but the low availability of bone and high cost limit these treatments. Therefore, shape memory polymers (SMPs), which combine biocompatibility, biodegradability, mechanical properties, shape tunability, ease of access, and minimally invasive implantation, have received attention in bone tissue engineering in recent years. Here, we reviewed the various excellent properties of SMPs and their contribution to bone formation in experiments at the cellular and animal levels, respectively, especially for the repair of defects in craniomaxillofacial (CMF) and limb bones, to provide new ideas for the application of these new SMPs in bone tissue engineering.
Collapse
|
190
|
He C, Lv Q, Liu Z, Long S, Li H, Xiao Y, Yang X, Liu Y, Liu C, Wang Z. Random and aligned electrostatically spun PLLA nanofibrous membranes enhance bone repair in mouse femur midshaft defects. J Biomater Appl 2023; 37:1582-1592. [PMID: 36662630 DOI: 10.1177/08853282221144220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Long-segment bone defects are a common clinical challenge and abstract biomaterials are a promising therapy. Poly-L-lactic acid (PLLA) nanofibrous membranes prepared by electrostatic spinning have a good bone repair potential. However, there are random and aligned surface morphologies of electrostatic spun PLLA nanofibrous membranes, which can affect the migration, proliferation, and differentiation ability of cells. The role of surface morphology in the repair of long bone defects in vivo is currently unknown. In this study, random and aligned electrostatically spun PLLA nanofibrous membranes were prepared, characterised, and implanted into a femur midshaft defect mouse model. The ability of electrostatically spun PLLA nanofibrous membranes to enhance bone repair was tested using X-ray photography, high-resolution micro-computed tomography (micro-CT), and pathological section specimens. The results showed that both random and aligned electrostatically spun PLLA nanofibrous membranes enhanced bone regeneration at bone defects, but the aligned ones exhibited superior results. These results provide a theoretical basis for engineering the surface morphology of bone repair materials.
Collapse
Affiliation(s)
- Chengkai He
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Basic Medical School of Kunming Medical University, Kunming, China
| | - Qiong Lv
- Outpatient Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhui Liu
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shengyu Long
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haohan Li
- The First Clinical College of Kunming Medical University, Kunming, China
| | - Ya Xiao
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Xingyu Yang
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Yuhang Liu
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Cai Liu
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Zhihua Wang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Yunnan Provincial Clinical Medical for Bone and Joint Diseases, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
191
|
Roumani S, Jeanneau C, Giraud T, Cotten A, Laucournet M, Sohier J, Pithioux M, About I. Osteogenic Potential of a Polyethylene Glycol Hydrogel Functionalized with Poly-Lysine Dendrigrafts (DGL) for Bone Regeneration. MATERIALS (BASEL, SWITZERLAND) 2023; 16:ma16020862. [PMID: 36676600 PMCID: PMC9863473 DOI: 10.3390/ma16020862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/28/2022] [Accepted: 01/12/2023] [Indexed: 05/27/2023]
Abstract
Resorbable hydrogels are widely used as scaffolds for tissue engineering. These hydrogels can be modified by grafting dendrimer-linked functionalized molecules (dendrigrafts). Our aim was to develop a tunable poly(L-lysine) dendrigrafts (DGL)/PEG-based hydrogel with an inverse porosity and to investigate its osteogenic potential. DGL/PEG hydrogels were emulsified in a surfactant-containing oil solution to form microspheres. The toxicity was evaluated on Human Vascular Endothelial Cells (HUVECs) and Bone Marrow Mesenchymal Stem Cells (hMSCs) with Live/Dead and MTT assays. The effects on HUVECs were investigated through C5 Complement expression by RT-PCR and C5a/TGF-β1 secretion by ELISA. Recruitment of hMSCs was investigated using Boyden chambers and their osteogenic differentiation was studied by measuring Alkaline Phosphatase activity (ALP) and BMP-2 secretion by ELISA. Adjusting the stirring speed during the emulsification allowed to obtain spherical microspheres with tunable diameters (10-1600 µm). The cell viability rate with the hydrogel was 95 and 100% with HUVECs and hMSCs, respectively. Incubating HUVECs with the biomaterial induced a 5-fold increase in TGF-β1 and a 3-fold increase in Complement C5a release. Furthermore, HUVEC supernatants obtained after incubation with the hydrogel induced a 2.5-fold increase in hMSC recruitment. The hydrogel induced a 3-fold increase both in hMSC ALP activity and BMP-2 secretion. Overall, the functionalized hydrogel enhanced the osteogenic potential by interacting with endothelial cells and hMSCs and represents a promising tool for bone tissue engineering.
Collapse
Affiliation(s)
- Sandra Roumani
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France
| | | | - Thomas Giraud
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France
- APHM, Hôpital Timone, Pôle Odontologie, 13005 Marseille, France
| | - Aurélie Cotten
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France
| | - Marc Laucournet
- Laboratory for Tissue Biology and Therapeutic Engineering (LBTI), UMR 5305, CNRS, Lyon University, 69367 Lyon, France
| | - Jérôme Sohier
- Laboratory for Tissue Biology and Therapeutic Engineering (LBTI), UMR 5305, CNRS, Lyon University, 69367 Lyon, France
| | - Martine Pithioux
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France
- Aix-Marseille University, APHM, CNRS, ISM, Sainte-Marguerite Hospital, Institute for Locomotion, Department of Orthopaedics and Traumatology, 13009 Marseille, France
| | - Imad About
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France
| |
Collapse
|
192
|
Wang Z, Yi X, Liu Y, Liu Q, Li Z, Yu A. Differential expression profiles and functional prediction of circRNA in mice with traumatic heterotopic ossification. Front Immunol 2023; 13:1090529. [PMID: 36713424 PMCID: PMC9878564 DOI: 10.3389/fimmu.2022.1090529] [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: 11/05/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023] Open
Abstract
Background Traumatic heterotopic ossification (HO) is an intractable sequela incited by inflammatory insult. To date, the exact molecular mechanisms of traumatic HO formation remain unclear. Recent studies have indicated that circular RNAs (circRNAs) participate in various human skeletal diseases. Although the formation of HO recapitulates many programs during bone development and remodeling, few data are available concerning whether circRNAs could participate in this pathological osteogenesis. Methods To investigate the differentially expressed circRNAs (DE-circRNAs) in HO formation, microarray assay was performed to analyze the circRNA expression profile in four pairs of mice HO tissues and normal tissues. Then, qRT-PCR was applied to verify the microarray data. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed the biological functions of the differentially expressed circRNAs target genes. Cytoscape software was used to construct the circRNA-miRNA-mRNA network for circRNAs with different expression levels as well as the target genes. Results We demonstrated that 491 circRNAs were significantly differentially expressed in mouse HO tissues by a fold-change ≥ 2 and p-value ≤ 0.05. Among them, the expressions of 168 circRNAs were increased, while 323 were decreased. The expression levels of 10 selected circRNAs were verified successfully by qRT-PCR. GO analysis exhibited that these DE-circRNAs participated in a series of cellular processes. KEGG pathway analysis revealed that multiple upregulated and downregulated pathways were closely related to the DE-circRNAs in HO mice. The circRNA-miRNA-mRNA networks demonstrated that DE-circRNAs may be involved in the pathological osteogenesis of HO through the circRNA-targeted miRNA-mRNA axis. Conclusion Our study first demonstrated the expression profiles and predicted the potential functions of DE-circRNAs in mice traumatic HO, which may shed new light on the elucidation of mechanisms as well as provide novel potential peripheral biological diagnostic markers and therapeutic targets for traumatic HO.
Collapse
Affiliation(s)
| | | | | | - Qiaoyun Liu
- *Correspondence: Qiaoyun Liu, ; Zonghuan Li, ; Aixi Yu,
| | - Zonghuan Li
- *Correspondence: Qiaoyun Liu, ; Zonghuan Li, ; Aixi Yu,
| | - Aixi Yu
- *Correspondence: Qiaoyun Liu, ; Zonghuan Li, ; Aixi Yu,
| |
Collapse
|
193
|
Nogueira AVB, Lopes MES, Marcantonio CC, Salmon CR, Mofatto LS, Deschner J, Nociti-Junior FH, Cirelli JA. Obesity Modifies the Proteomic Profile of the Periodontal Ligament. Int J Mol Sci 2023; 24:ijms24021003. [PMID: 36674516 PMCID: PMC9861657 DOI: 10.3390/ijms24021003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023] Open
Abstract
This study aimed to assess the obesity effects on the proteomic profile of the periodontal ligament of rats submitted to obesity induction by a high-fat diet. Eight Holtzman rats were divided into control (n = 3) and obese (n = 5) groups. The maxillae were histologically processed for laser capture microdissection of the periodontal ligament of the first maxillary molars. Peptide mixtures were analyzed by LC-MS/MS. A total of 1379 proteins were identified in all groups. Among them, 335 (24.30%) were exclusively detected in the obese group, while 129 (9.35%) proteins were uniquely found in the control group. Out of the 110 (7.98%) differentially abundant proteins, 10 were more abundant and 100 had decreased abundance in the obese group. A gene ontology analysis showed some proteins related to obesity in the “extracellular exosome” term among differentially identified proteins in the gene ontology cellular component terms Prelp, Sec13, and Sod2. These three proteins were upregulated in the obese group (p < 0.05), as shown by proteomic and immunohistochemistry analyses. In summary, our study presents novel evidence that the proteomic profile of the periodontal ligament is altered in experimental obesity induction, providing a list of differentially abundant proteins associated with obesity, which indicates that the periodontal ligament is responsive to obesity.
Collapse
Affiliation(s)
- Andressa V. B. Nogueira
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
- Correspondence: (A.V.B.N.); (J.A.C.); Tel.: +49-0-6131-17-7091 (A.V.B.N.); +55-16-3301-6375 (J.A.C.)
| | - Maria Eduarda S. Lopes
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
| | - Camila C. Marcantonio
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
| | - Cristiane R. Salmon
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas—UNICAMP, Piracicaba 13414-903, São Paulo, Brazil
| | - Luciana S. Mofatto
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas—UNICAMP, Campinas 13083-862, São Paulo, Brazil
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Francisco H. Nociti-Junior
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas—UNICAMP, Piracicaba 13414-903, São Paulo, Brazil
- São Leopoldo Mandic Research Center, Campinas 13045-755, São Paulo, Brazil
| | - Joni A. Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, São Paulo, Brazil
- Correspondence: (A.V.B.N.); (J.A.C.); Tel.: +49-0-6131-17-7091 (A.V.B.N.); +55-16-3301-6375 (J.A.C.)
| |
Collapse
|
194
|
Darshna, Kumar R, Srivastava P, Chandra P. Bioengineering of bone tissues using bioreactors for modulation of mechano-sensitivity in bone. Biotechnol Genet Eng Rev 2023:1-41. [PMID: 36596226 DOI: 10.1080/02648725.2022.2162249] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023]
Abstract
Since the last decade, significant developments have been made in the area of bone tissue engineering associated with the emergence of novel biomaterials as well as techniques of scaffold fabrication. Despite all these developments, the translation from research findings to clinical applications is still very limited. Manufacturing the designed tissue constructs in a scalable manner remains the most challenging aspect. This bottleneck could be overcome by using bioreactors for the manufacture of these tissue constructs. In this review, a current scenario of bone injuries/defects and the cause of the translational gap between laboratory research and clinical use has been emphasized. Furthermore, various bioreactors being used in the area of bone tissue regeneration in recent studies have been highlighted along with their advantages and limitations. A vivid literature survey on the ideal attributes of bioreactors has been accounted, viz. dynamic, versatile, automated, reproducible and commercialization aspects. Additionally, the illustration of computational approaches that should be combined with bone tissue engineering experiments using bioreactors to simulate and optimize cellular growth in bone tissue constructs has also been done extensively.
Collapse
Affiliation(s)
- Darshna
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Rahul Kumar
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pradeep Srivastava
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
195
|
Hu D, Cai Y, He L, Jiang H. Identification of a CircRNA-miRNA-mRNA Network and Integrated Analysis of Immune Infiltration in Oral Squamous Cell Carcinoma. J Cancer 2023; 14:250-261. [PMID: 36741263 PMCID: PMC9891867 DOI: 10.7150/jca.79967] [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: 10/17/2022] [Accepted: 12/12/2022] [Indexed: 01/04/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a highly invasive type of head and neck cancer. Circular RNA (circRNA) acts as a competing endogenous RNA (ceRNA) and involves in pathogenesis of many diseases. However, the circRNA-miRNA-mRNA network and relationship between ceRNA and immune infiltration in OSCC remain unknown. In this study, we established a ceRNA network, including 89 circRNAs, 43 miRNAs and 223 mRNAs, and found that 233 genes are mainly related to malignant signalling pathways (including "Integrin family cell surface interactions" and "Epithelial-to-mesenchymal transition" pathways) and five potential biomarkers (SLC20A1, PITX2, hsa-mir-135b, hsa-mir-377 and hsa-let-7c). Meanwhile, we established a prognostic model based on clinical risk, and revealed the relationship between immune infiltrating cells and biomarkers in OSCC. Taken together, our study is helpful to reveal the pathogenesis of oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Dehua Hu
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Yufeng Cai
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Lan He
- College of Biology, Hunan University, Changsha, Hunan 410078, China
| | - Hao Jiang
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China.,✉ Corresponding author: Hao Jiang, 172# Tongzipo Road, School of Life Sciences, Central South University, Changsha, Hunan 410011, People's Republic of China. Tel: +86-13671769729.
| |
Collapse
|
196
|
Fu M, Li J, Liu M, Yang C, Wang Q, Wang H, Chen B, Fu Q, Sun G. Sericin/Nano-Hydroxyapatite Hydrogels Based on Graphene Oxide for Effective Bone Regeneration via Immunomodulation and Osteoinduction. Int J Nanomedicine 2023; 18:1875-1895. [PMID: 37051313 PMCID: PMC10084881 DOI: 10.2147/ijn.s399487] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/05/2023] [Indexed: 04/14/2023] Open
Abstract
Background Immune responses and osteogenesis differentiation induced by implants are crucial for bone tissue regeneration. Consideration of only one of those properties is not sufficient. To investigate the synergistic actions, we designed alginate/graphene oxide/sericin/nanohydroxyapatite (Alg/GO/Ser/nHAP) nanocomposite hydrogels with both osteoimmunomodulatory and osteoinductive activities. This study aimed to explore the effect of hydrogel with osteoimmunomodulatory properties on promoting osteogenesis of bone marrow stem cells (BMSCs). Methods Alg/GO/Ser/nHAP nanocomposite hydrogel was fabricated and was characterized by SEM, FTIR, XRD, stress-strain, rheology, swelling and degradation. After the impact of sericin on M2 macrophage polarization was identified, the BMSCs viability and adhesion were evaluated by CCK8 assay, live/dead staining, cytoskeleton staining. The cell osteogenic differentiation was observed by ALP/ARS staining, immunofluorescence staining, RT-PCR, and Western blotting, respectively. Rat cranial defect model was used to assess osteoimmunomodulatory effects of scaffolds in vivo by micro‑computed tomographic, histological, and immunohistochemical analyses after 8 weeks of healing. Results In vitro experiments revealed that the hydrogel presented desirable mechanical strength, stability, porosity, and biocompatibility. Significantly, sericin and nHAP appeared to exert synergistic effects on bone regeneration. Sericin was observed to inhibit the immune response by inducing macrophage M2-type polarization to create a positive osteoimmune microenvironment, contributing to improving osseointegration at the bone-implant interface to promote osteogenesis. However, the osteogenic differentiation in rat BMSCs was further enhanced by combining nHAP and sericin in the nanocomposite hydrogel. Eventually, the hydrogel was implanted into the rat cranial defect model, assisting in the reduction of local inflammation and efficient bone regeneration. Conclusion The nanocomposite hydrogel stimulated bone formation by the synergistic effects of immunomodulation of macrophage polarization by sericin and direct osteogenic induction by nHAP, demonstrating that such a scaffold that modulates the osteoimmune microenvironment to promote osteogenesis is a promising approach for the development of bone tissue engineering implants in the future.
Collapse
Affiliation(s)
- Mei Fu
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Jun Li
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Mingchong Liu
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Chensong Yang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Qidong Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Hongrui Wang
- Department of Orthopedic Trauma, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, People’s Republic of China
| | - Bingdi Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Qingge Fu
- Department of Orthopedic Trauma, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, People’s Republic of China
| | - Guixin Sun
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Correspondence: Guixin Sun; Qingge Fu, Email ;
| |
Collapse
|
197
|
Wang YX, Peng ZL, Sun ZW, Pan YJ, Ai H, Mai ZH. MiR-20a promotes osteogenic differentiation in bone marrow-derived mesenchymal stem/stromal cells and bone repair of the maxillary sinus defect model in rabbits. Front Bioeng Biotechnol 2023; 11:1127908. [PMID: 37091341 PMCID: PMC10113429 DOI: 10.3389/fbioe.2023.1127908] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction: This study aimed to determine whether miR-20 promoted osteogenic differentiation in bone marrow-derived mesenchymal stem/stromal cells (BMSCs) and accelerated bone formation in the maxillary sinus bone defect model in rabbits. Methods: BMSCs were transfected with miR-20a or anti-miR-20a for 12 h, followed by detection of RUNX2, Sp7 mRNA, bone morphogenetic protein 2 (BMP2), and RUNX2 protein expression. Alkaline phosphatase (ALP) activity and Alizarin Red S staining were used to detect calcified nodule deposition. In the rabbit maxillary sinus bone defect model, miR-20a loaded with AAV and BMP2 protein were mixed with Bio-Oss bone powder for filling the bone defect. At 4 weeks and 8 weeks, bone density was detected by cone beam computed tomography (CBCT), and new bone, osteoblasts, and collagen type 1 were evaluated by hematoxylin and eosin (HE) staining and immunohistochemical (IHC) staining. Results: Overexpression of miR-20a enhanced the mRNA and protein levels of BMP2, RUNX2, and SP7, the activity of ALP, and the levels of matrix mineralization, whereas the levels and activity of the aforementioned factors were decreased by anti-miR-20a treatment of BMSCs. Furthermore, miR-20a significantly increased the bone density, the number of osteoblasts, and the secretion of collagen type 1 in bone defects compared with Bio-Oss bone powder in the rabbit maxillary sinus bone defect model. Conclusion: Overall, miR-20a can induce osteogenic differentiation in BMSCs and accelerate bone formation of maxillary sinus defects in rabbits.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Orthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhu-Li Peng
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhi-Wen Sun
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yan-Jun Pan
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hong Ai
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhi-Hui Mai
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Zhi-Hui Mai,
| |
Collapse
|
198
|
Sinha SP, Bajracharya M, Huang CS, Ko EWC. Does the floor of the maxillary sinus affect tooth movement for premolar extraction space closure? Clin Oral Investig 2023; 27:299-304. [PMID: 36104604 DOI: 10.1007/s00784-022-04723-3] [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: 02/14/2022] [Accepted: 09/06/2022] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The study investigated orthodontic tooth movement by comparing various horizontal and angular measures of the teeth having different sinus-root relationship (SRR) of patients undergoing first premolar extraction treatment protocols. MATERIALS AND METHODS This retrospective cross-sectional study included 45 Taiwanese adult patients, classified as the control group (n = 15) and the sinus group (n = 30) according to SRR. The control group had type I SRR (sinus floor above the level connecting the buccal and palatal root apices), whereas the sinus group included patients with type V SRR (the buccal and palatal root displayed apical protrusion into the sinus floor). Morphology was identified using pretreatment orthopantomograms. Lateral cephalometric measurements were conducted before treatment (T1) and at debonding (T2). RESULTS Post debonding, there were no statistically significant difference in the two groups. However, the incisors in the control group became marginally more upright than those of the sinus group, and the molars demonstrated tipping movement with minimal anchorage loss in the sinus group than in the control group. The total treatment time was not significantly different between the 2 groups. CONCLUSION The study revealed that the sinus-root relationship does not affect the orthodontic tooth movement. However, a more extensive 3-D study with larger sample size is strongly recommended before coming to any conclusion. CLINICAL RELEVANCE The lining of the floor of the maxillary sinus does not affect the orthodontic tooth movement.
Collapse
Affiliation(s)
- Suraj Prasad Sinha
- Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Manish Bajracharya
- Dental Department, Orthodontic Unit, National Academy of Medical Sciences, Bir Hospital, Kathmandu, Nepal
| | - Chiung-Shing Huang
- Graduate Institute of Craniofacial and Dental Science, Chang Gung University, Taoyuan, Taiwan.,Department of Craniofacial Orthodontic, Chang Gung Memorial Hospital, 6F, 199, Tung Hwa North Road, Taipei, 105, Taiwan
| | - Ellen Wen-Ching Ko
- Graduate Institute of Craniofacial and Dental Science, Chang Gung University, Taoyuan, Taiwan. .,Department of Craniofacial Orthodontic, Chang Gung Memorial Hospital, 6F, 199, Tung Hwa North Road, Taipei, 105, Taiwan. .,Department of Dentistry, Chang Gung Memorial Hospital, Linkous, Taiwan.
| |
Collapse
|
199
|
Zhang Q, Long Y, Jin L, Li C, Long J. Non-coding RNAs regulate the BMP/Smad pathway during osteogenic differentiation of stem cells. Acta Histochem 2023; 125:151998. [PMID: 36630753 DOI: 10.1016/j.acthis.2023.151998] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
MicroRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) are involved in the regulation of bone metabolism. The BMP/Smad pathway is a key signaling pathway for classical regulation of osteogenic differentiation. Non-coding RNAs (ncRNAs) and the BMP/Smad pathway both have important roles for osteogenic differentiation of stem cells, bone regeneration, and development of bone diseases. There is increasing evidence that ncRNAs interact with the BMP/Smad pathway to regulate not only osteogenic differentiation of stem cells but also progression of bone diseases, such as osteoporosis (OP), myeloma, and osteonecrosis of the femoral head (ONFH), by controlling the expression of bone disease-related genes. Therefore, ncRNAs that interact with BMP/Smad pathway molecules are potential targets for bone regeneration as well as bone disease diagnosis, prevention, and treatment. However, despite extensive studies on ncRNAs associated with the BMP/Smad pathway and osteogenic differentiation of stem cells, there is a lack of comparability. Moreover, some bone disease-associated ncRNAs with low abundance can be difficult to detect and there is a lack of mature delivery systems for their stable translocation to target sites, thus limiting their application. In this review, we summarize the research progress on interactions between ncRNAs and the BMP/Smad pathway during osteogenic differentiation of various stem cells and in the regulation of bone regeneration and bone diseases.
Collapse
Affiliation(s)
- Qiuling Zhang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Yifei Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Liangyu Jin
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Chenghao Li
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
200
|
Wang Z, Wen S, Zhong M, Yang Z, Xiong W, Zhang K, Yang S, Li H, Guo S. Epigenetics: Novel crucial approach for osteogenesis of mesenchymal stem cells. J Tissue Eng 2023; 14:20417314231175364. [PMID: 37342486 PMCID: PMC10278427 DOI: 10.1177/20417314231175364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/26/2023] [Indexed: 06/23/2023] Open
Abstract
Bone has a robust regenerative potential, but its capacity to repair critical-sized bone defects is limited. In recent years, stem cells have attracted significant interest for their potential in tissue engineering. Applying mesenchymal stem cells (MSCs) for enhancing bone regeneration is a promising therapeutic strategy. However, maintaining optimal cell efficacy or viability of MSCs is limited by several factors. Epigenetic modification can cause changes in gene expression levels without changing its sequence, mainly including nucleic acids methylation, histone modification, and non-coding RNAs. This modification is believed to be one of the determinants of MSCs fate and differentiation. Understanding the epigenetic modification of MSCs can improve the activity and function of stem cells. This review summarizes recent advances in the epigenetic mechanisms of MSCs differentiation into osteoblast lineages. We expound that epigenetic modification of MSCs can be harnessed to treat bone defects and promote bone regeneration, providing potential therapeutic targets for bone-related diseases.
Collapse
Affiliation(s)
- Zhaohua Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Si Wen
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Meiqi Zhong
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ziming Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Kuo Zhang
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning Province, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Huizheng Li
- Department of Otorhinolaryngology & Head and Neck Surgery, Dalian Friendship Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|