1
|
Casarrubios L, Cicuéndez M, Polo-Montalvo A, Feito MJ, Martínez-Del-Pozo Á, Arcos D, Duarte IF, Portolés MT. Metabolomic characterization of MC3T3-E1pre-osteoblast differentiation induced by ipriflavone-loaded mesoporous nanospheres. BIOMATERIALS ADVANCES 2025; 166:214085. [PMID: 39490191 DOI: 10.1016/j.bioadv.2024.214085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
This study reports on the metabolic changes accompanying the differentiation of MC3T3-E1 osteoprogenitor cells induced by mesoporous bioactive glass nanospheres (nMBG) loaded with ipriflavone (nMBG-IP). Ipriflavone (IP) is a synthetic isoflavone known for inhibiting bone resorption, maintaining bone density, and preventing osteoporosis. Delivering IP intracellularly is a promising strategy to modulate bone remodeling at significantly lower doses compared to free drug administration. Our results demonstrate that nMBG are efficiently internalized by pre-osteoblasts and, when loaded with IP, induce their differentiation. This differentiation process is accompanied by pronounced metabolic alterations, as monitored by NMR analysis of medium supernatants and cell extracts (exo- and endo-metabolomics, respectively). The main effects include an early-stage intensification of glycolysis and changes in several metabolic pathways, such as nucleobase metabolism, osmoregulatory and antioxidant pathways, and lipid metabolism. Notably, the metabolic impacts of nMBG-IP and free IP were very similar, whereas nMBG alone induced only mild changes in the intracellular metabolic profile without affecting the cells' consumption/secretion patterns or lipid composition. This finding indicates that the observed effects are primarily related to IP-induced differentiation and that nMBG nanospheres serve as convenient carriers with both efficient internalization and minimal metabolic impact. Furthermore, the observed link between pre-osteoblast differentiation and metabolism underscores the potential of utilizing metabolites and metabolic reprogramming as strategies to modulate the osteogenic process, for instance, in the context of osteoporosis and other bone diseases.
Collapse
Affiliation(s)
- Laura Casarrubios
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Mónica Cicuéndez
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Alberto Polo-Montalvo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - María José Feito
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Álvaro Martínez-Del-Pozo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Daniel Arcos
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, Madrid 28040, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, Madrid 28040, Spain
| | - Iola F Duarte
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - María Teresa Portolés
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, Madrid 28040, Spain.
| |
Collapse
|
2
|
Fan Y, Sun J, Fan W, Zhong X, Yin Z, Su B, Yao J, Hong X, Zhai J, Wang Z, Chen H, Guo F, Wen X, Ning C, Chen L, Yu P. Three-Dimensional Semiconductor Network as Regulators of Energy Metabolism Drives Angiogenesis in Bone Regeneration. ACS NANO 2024; 18:32602-32616. [PMID: 39530623 DOI: 10.1021/acsnano.4c09971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Insufficient vascularization is a primary cause of bone implantation failure. The management of energy metabolism is crucial for the achievement of vascularized osseointegration. In light of the bone semiconductor property and the electric property of semiconductor heterojunctions, a three-dimensional semiconductor heterojunction network (3D-NTBH) implant has been devised with the objective of regulating cellular energy metabolism, thereby driving angiogenesis for bone regeneration. The three-dimensional heterojunction interfaces facilitate electron transfer and establish internal electric fields at the nanoscale interfaces. The 3D-NTBH was found to noticeably accelerate glycolysis in endothelial cells, thereby rapidly providing energy to support cellular metabolic activities and ultimately driving angiogenesis within the bone tissue. Molecular dynamic simulations have demonstrated that the 3D-NTBH facilitates the exposure of fibronectin's Arg-Gly-Asp peptide binding site, thereby regulating the glycolysis of endothelial cells. Further evidence suggests that 3D-NTBH promotes early vascular network reconstruction and bone regeneration in vivo. The findings of this research offer a promising research perspective for the design of vascularizing implants.
Collapse
Affiliation(s)
- Youzhun Fan
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Jiwei Sun
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Wenjie Fan
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xianwei Zhong
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab of Green Chemical Product Technology, South China University of Technology, Guangzhou 510641, China
| | - Zhaoyi Yin
- Faculty of Materials Science and Engineering, Kunming University of Science and Technology, Kunming 650093, China
| | - Bin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Yao
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xinyu Hong
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Jinxia Zhai
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Zhengao Wang
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Haoyan Chen
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Fengyuan Guo
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiufang Wen
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab of Green Chemical Product Technology, South China University of Technology, Guangzhou 510641, China
| | - Chengyun Ning
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| | - Lili Chen
- Department of Stomatology, School of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Peng Yu
- School of Materials Science and Engineering, Guangdong Engineering Technology Research Center of Metallic Materials Surface Functionalization, National Engineering Research Center for Tissue Restoration and Reconstruction, Medical Devices Research and Testing Center, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
3
|
Hong X, Tian G, Dai B, Zhou X, Gao Y, Zhu L, Liu H, Zhu Q, Zhang L, Zhu Y, Ren D, Guo C, Nan J, Liu X, Wang J, Ren T. Copper-loaded Milk-Protein Derived Microgel Preserves Cardiac Metabolic Homeostasis After Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401527. [PMID: 39007192 PMCID: PMC11425262 DOI: 10.1002/advs.202401527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/11/2024] [Indexed: 07/16/2024]
Abstract
Myocardial Infarction (MI) is a leading cause of death worldwide. Metabolic modulation is a promising therapeutic approach to prevent adverse remodeling after MI. However, whether material-derived cues can treat MI through metabolic regulation is mainly unexplored. Herein, a Cu2+ loaded casein microgel (CuCMG) aiming to rescue the pathological intramyocardial metabolism for MI amelioration is developed. Cu2+ is an important ion factor involved in metabolic pathways, and intracardiac copper drain is observed after MI. It is thus speculated that intramyocardial supplementation of Cu2+ can rescue myocardial metabolism. Casein, a milk-derived protein, is screened out as Cu2+ carrier through molecular-docking based on Cu2+ loading capacity and accessibility. CuCMGs notably attenuate MI-induced cardiac dysfunction and maladaptive remodeling, accompanied by increased angiogenesis. The results from unbiased transcriptome profiling and oxidative phosphorylation analyses support the hypothesis that CuCMG prominently rescued the metabolic homeostasis of myocardium after MI. These findings enhance the understanding of the design and application of metabolic-modulating biomaterials for ischemic cardiomyopathy therapy.
Collapse
Affiliation(s)
- Xiaoqian Hong
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Geer Tian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Binyao Dai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xuhao Zhou
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Ying Gao
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Lianlian Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Haoran Liu
- School of Engineering, Westlake University, Hangzhou, 310023, China
| | - Qinchao Zhu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Liwen Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yang Zhu
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Daxi Ren
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, 310023, China
| | - Jinliang Nan
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Tanchen Ren
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
4
|
Yu L, Bennett CJ, Lin CH, Yan S, Yang J. Scaffold design considerations for peripheral nerve regeneration. J Neural Eng 2024; 21:041001. [PMID: 38996412 DOI: 10.1088/1741-2552/ad628d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/12/2024] [Indexed: 07/14/2024]
Abstract
Peripheral nerve injury (PNI) represents a serious clinical and public health problem due to its high incurrence and poor spontaneous recovery. Compared to autograft, which is still the best current practice for long-gap peripheral nerve defects in clinics, the use of polymer-based biodegradable nerve guidance conduits (NGCs) has been gaining momentum as an alternative to guide the repair of severe PNI without the need of secondary surgery and donor nerve tissue. However, simple hollow cylindrical tubes can barely outperform autograft in terms of the regenerative efficiency especially in critical sized PNI. With the rapid development of tissue engineering technology and materials science, various functionalized NGCs have emerged to enhance nerve regeneration over the past decades. From the aspect of scaffold design considerations, with a specific focus on biodegradable polymers, this review aims to summarize the recent advances in NGCs by addressing the onerous demands of biomaterial selections, structural designs, and manufacturing techniques that contributes to the biocompatibility, degradation rate, mechanical properties, drug encapsulation and release efficiency, immunomodulation, angiogenesis, and the overall nerve regeneration potential of NGCs. In addition, several commercially available NGCs along with their regulation pathways and clinical applications are compared and discussed. Lastly, we discuss the current challenges and future directions attempting to provide inspiration for the future design of ideal NGCs that can completely cure long-gap peripheral nerve defects.
Collapse
Affiliation(s)
- Le Yu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Carly Jane Bennett
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Chung-Hsun Lin
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Jian Yang
- Biomedical Engineering Program, Westlake University, Hangzhou, Zhejiang 310030, People's Republic of China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang 310030, People's Republic of China
| |
Collapse
|
5
|
Srivastava GK, Martinez-Rodriguez S, Md Fadilah NI, Looi Qi Hao D, Markey G, Shukla P, Fauzi MB, Panetsos F. Progress in Wound-Healing Products Based on Natural Compounds, Stem Cells, and MicroRNA-Based Biopolymers in the European, USA, and Asian Markets: Opportunities, Barriers, and Regulatory Issues. Polymers (Basel) 2024; 16:1280. [PMID: 38732749 PMCID: PMC11085499 DOI: 10.3390/polym16091280] [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: 02/09/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024] Open
Abstract
Wounds are breaks in the continuity of the skin and underlying tissues, resulting from external causes such as cuts, blows, impacts, or surgical interventions. Countless individuals suffer minor to severe injuries, with unfortunate cases even leading to death. In today's scenario, several commercial products are available to facilitate the healing process of wounds, although chronic wounds still present more challenges than acute wounds. Nevertheless, the huge demand for wound-care products within the healthcare sector has given rise to a rapidly growing market, fostering continuous research and development endeavors for innovative wound-healing solutions. Today, there are many commercially available products including those based on natural biopolymers, stem cells, and microRNAs that promote healing from wounds. This article explores the recent breakthroughs in wound-healing products that harness the potential of natural biopolymers, stem cells, and microRNAs. A comprehensive exploration is undertaken, covering not only commercially available products but also those still in the research phase. Additionally, we provide a thorough examination of the opportunities, obstacles, and regulatory considerations influencing the potential commercialization of wound-healing products across the diverse markets of Europe, America, and Asia.
Collapse
Affiliation(s)
- Girish K. Srivastava
- Departamento de Cirugía, Oftalmología, Otorrinolaringología y Fisioterapia, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain;
- Instituto Universitario de Oftalmobiología Aplicada, Facultad de Medicina, Universidad de Valladolid, 47011 Valladolid, Spain;
| | - Sofia Martinez-Rodriguez
- Instituto Universitario de Oftalmobiología Aplicada, Facultad de Medicina, Universidad de Valladolid, 47011 Valladolid, Spain;
| | - Nur Izzah Md Fadilah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.I.M.F.); (D.L.Q.H.); (M.B.F.)
| | - Daniel Looi Qi Hao
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.I.M.F.); (D.L.Q.H.); (M.B.F.)
- My Cytohealth Sdn. Bhd., Kuala Lumpur 56000, Malaysia
| | - Gavin Markey
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Londonderry BT47 6SB, UK; (G.M.); (P.S.)
| | - Priyank Shukla
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Londonderry BT47 6SB, UK; (G.M.); (P.S.)
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.I.M.F.); (D.L.Q.H.); (M.B.F.)
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group, Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Institute for Health Research San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain
- Silk Biomed SL, 28260 Madrid, Spain
- Bioactive Surfaces SL, 28260 Madrid, Spain
- Omnia Mater SL, 28009 Madrid, Spain
| |
Collapse
|
6
|
Liu X, Wan X, Sui B, Hu Q, Liu Z, Ding T, Zhao J, Chen Y, Wang ZL, Li L. Piezoelectric hydrogel for treatment of periodontitis through bioenergetic activation. Bioact Mater 2024; 35:346-361. [PMID: 38379699 PMCID: PMC10876489 DOI: 10.1016/j.bioactmat.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/26/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
The impaired differentiation ability of resident cells and disordered immune microenvironment in periodontitis pose a huge challenge for bone regeneration. Herein, we construct a piezoelectric hydrogel to rescue the impaired osteogenic capability and rebuild the regenerative immune microenvironment through bioenergetic activation. Under local mechanical stress, the piezoelectric hydrogel generated piezopotential that initiates osteogenic differentiation of inflammatory periodontal ligament stem cells (PDLSCs) via modulating energy metabolism and promoting adenosine triphosphate (ATP) synthesis. Moreover, it also reshapes an anti-inflammatory and pro-regenerative niche through switching M1 macrophages to the M2 phenotype. The synergy of tilapia gelatin and piezoelectric stimulation enhances in situ regeneration in periodontal inflammatory defects of rats. These findings pave a new pathway for treating periodontitis and other immune-related bone defects through piezoelectric stimulation-enabled energy metabolism modulation and immunomodulation.
Collapse
Affiliation(s)
- Xin Liu
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, PR China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, PR China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Baiyan Sui
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, PR China
| | - Quanhong Hu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, PR China
| | - Zhirong Liu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, PR China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Tingting Ding
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, PR China
| | - Jiao Zhao
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, PR China
| | - Yuxiao Chen
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, PR China
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, PR China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, PR China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
7
|
Jiao X, Wu F, Yue X, Yang J, Zhang Y, Qiu J, Ke X, Sun X, Zhao L, Xu C, Li Y, Yang X, Yang G, Gou Z, Zhang L. New insight into biodegradable macropore filler on tuning mechanical properties and bone tissue ingrowth in sparingly dissolvable bioceramic scaffolds. Mater Today Bio 2024; 24:100936. [PMID: 38234459 PMCID: PMC10792586 DOI: 10.1016/j.mtbio.2023.100936] [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: 10/22/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024] Open
Abstract
Structural parameters of the implants such as shape, size, and porosity of the pores have been extensively investigated to promote bone tissue repair, however, it is unknown how the pore interconnectivity affects the bone growth behaviors in the scaffolds. Herein we systematically evaluated the effect of biodegradable bioceramics as a secondary phase filler in the macroporous networks on the mechanical and osteogenic behaviors in sparingly dissolvable bioceramic scaffolds. The pure hardystonite (HT) scaffolds with ∼550 & 800 μm in pore sizes were prepared by digital light processing, and then the Sr-doped calcium silicate (SrCSi) bioceramic slurry without and with 30 % organic porogens were intruded into the HT scaffolds with 800 μm pore size and sintered at 1150 °C. It indicated that the organic porogens could endow spherical micropores in the SrCSi filler, and the invasion of the SrCSi component could not only significantly enhance the compressive strength and modulus of the HT-based scaffolds, but also induce osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). The pure HT scaffolds showed extremely slow bio-dissolution in Tris buffer after immersion for 8 weeks (∼1 % mass decay); in contrast, the SrCSi filler would readily dissolve into the aqueous medium and produced a steady mass decay (>6 % mass loss). In vivo experiments in rabbit femoral bone defect models showed that the pure HT scaffolds showed bone tissue ingrowth but the bone growth was impeded in the SrCSi-intruded scaffolds within 4 weeks; however, the group with higher porosity of SrCSi filler showed appreciable osteogenesis after 8 weeks of implantation and the whole scaffold was uniformly covered by new bone tissues after 16 weeks. These findings provide some new insights that the pore interconnectivity is not inevitable to impede bone ingrowth with the prolongation of implantation time, and such a highly biodegradable and bioactive filler intrusion strategy may be beneficial for optimizing the performances of scaffolds in bone regenerative medicine applications.
Collapse
Affiliation(s)
- Xiaoyi Jiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Fanghui Wu
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Xusong Yue
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Jun Yang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Yan Zhang
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jiandi Qiu
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Xiurong Ke
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Xiaoliang Sun
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Liben Zhao
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Chuchu Xu
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yifan Li
- Department of Orthopaedics, The First Affiliated Hospital, School of Medicine of Zhejiang University, Hangzhou, 310003, China
| | - Xianyan Yang
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou, 310058, China
| | - Guojing Yang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Zhongru Gou
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou, 310058, China
| | - Lei Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| |
Collapse
|
8
|
Wang H, Huddleston S, Yang J, Ameer GA. Enabling Proregenerative Medical Devices via Citrate-Based Biomaterials: Transitioning from Inert to Regenerative Biomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306326. [PMID: 38043945 DOI: 10.1002/adma.202306326] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/03/2023] [Indexed: 12/05/2023]
Abstract
Regenerative medicine aims to restore tissue and organ function without the use of prosthetics and permanent implants. However, achieving this goal has been elusive, and the field remains mostly an academic discipline with few products widely used in clinical practice. From a materials science perspective, barriers include the lack of proregenerative biomaterials, a complex regulatory process to demonstrate safety and efficacy, and user adoption challenges. Although biomaterials, particularly biodegradable polymers, can play a major role in regenerative medicine, their suboptimal mechanical and degradation properties often limit their use, and they do not support inherent biological processes that facilitate tissue regeneration. As of 2020, nine synthetic biodegradable polymers used in medical devices are cleared or approved for use in the United States of America. Despite the limitations in the design, production, and marketing of these devices, this small number of biodegradable polymers has dominated the resorbable medical device market for the past 50 years. This perspective will review the history and applications of biodegradable polymers used in medical devices, highlight the need and requirements for regenerative biomaterials, and discuss the path behind the recent successful introduction of citrate-based biomaterials for manufacturing innovative medical products aimed at improving the outcome of musculoskeletal surgeries.
Collapse
Affiliation(s)
- Huifeng Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Samantha Huddleston
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Jian Yang
- Biomedical Engineering Program, School of Engineering, Westlake University, Hangzhou, Zhejiang, 310030, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, 60611, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
9
|
Kapnick SM, Martin CA, Jewell CM. Engineering metabolism to modulate immunity. Adv Drug Deliv Rev 2024; 204:115122. [PMID: 37935318 PMCID: PMC10843796 DOI: 10.1016/j.addr.2023.115122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Metabolic programming and reprogramming have emerged as pivotal mechanisms for altering immune cell function. Thus, immunometabolism has become an attractive target area for treatment of immune-mediated disorders. Nonetheless, many hurdles to delivering metabolic cues persist. In this review, we consider how biomaterials are poised to transform manipulation of immune cell metabolism through integrated control of metabolic configurations to affect outcomes in autoimmunity, regeneration, transplant, and cancer. We emphasize the features of nanoparticles and other biomaterials that permit delivery of metabolic cues to the intracellular compartment of immune cells, or strategies for altering signals in the extracellular space. We then provide perspectives on the potential for reciprocal regulation of immunometabolism by the physical properties of materials themselves. Lastly, opportunities for clinical translation are highlighted. This discussion contributes to our understanding of immunometabolism, biomaterials-based strategies for altering metabolic configurations in immune cells, and emerging concepts in this evolving field.
Collapse
Affiliation(s)
- Senta M Kapnick
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA
| | - Corinne A Martin
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10 N Green Street, Baltimore, MD, USA; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S Greene Street, Suite N9E17, Baltimore, MD, USA.
| |
Collapse
|
10
|
Wu M, Zhao Y, Tao M, Fu M, Wang Y, Liu Q, Lu Z, Guo J. Malate-Based Biodegradable Scaffolds Activate Cellular Energetic Metabolism for Accelerated Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50836-50853. [PMID: 37903387 DOI: 10.1021/acsami.3c09394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
The latest advancements in cellular bioenergetics have revealed the potential of transferring chemical energy to biological energy for therapeutic applications. Despite efforts, a three-dimensional (3D) scaffold that can induce long-term bioenergetic effects and facilitate tissue regeneration remains a big challenge. Herein, the cellular energetic metabolism promotion ability of l-malate, an important intermediate of the tricarboxylic acid (TCA) cycle, was proved, and a series of bioenergetic porous scaffolds were fabricated by synthesizing poly(diol l-malate) (PDoM) prepolymers via a facial one-pot polycondensation of l-malic acid and aliphatic diols, followed by scaffold fabrication and thermal-cross-linking. The degradation products of the developed PDoM scaffolds can regulate the metabolic microenvironment by entering mitochondria and participating in the TCA cycle to elevate intracellular adenosine triphosphate (ATP) levels, thus promoting the cellular biosynthesis, including the production of collagen type I (Col1a1), fibronectin 1 (Fn1), and actin alpha 2 (Acta2/α-Sma). The porous PDoM scaffold was demonstrated to support the growth of the cocultured mesenchymal stem cells (MSCs) and promote their secretion of bioactive molecules [such as vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1), and basic fibroblast growth factor (bFGF)], and this stem cells-laden scaffold architecture was proved to accelerate wound healing in a critical full-thickness skin defect model on rats.
Collapse
Affiliation(s)
- Min Wu
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yitao Zhao
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Meihan Tao
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Meimei Fu
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yue Wang
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Qi Liu
- Regenerative Medicine and Tissue Repair Research Center, Huangpu Institute of Materials, Guangzhou 511363, P. R. China
| | - Zhihui Lu
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Regenerative Medicine and Tissue Repair Research Center, Huangpu Institute of Materials, Guangzhou 511363, P. R. China
| | - Jinshan Guo
- Department of Histology and Embryology, GDMPA Key Laboratory of Key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
11
|
Pota G, Silvestri B, Vitiello G, Gallucci N, Di Girolamo R, Scialla S, Raucci MG, Ambrosio L, Di Napoli M, Zanfardino A, Varcamonti M, Pezzella A, Luciani G. Towards nanostructured red-ox active bio-interfaces: Bioinspired antibacterial hybrid melanin-CeO 2 nanoparticles for radical homeostasis. BIOMATERIALS ADVANCES 2023; 153:213558. [PMID: 37467646 DOI: 10.1016/j.bioadv.2023.213558] [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] [Received: 02/04/2023] [Revised: 05/29/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Redox-active nano-biointerfaces are gaining weight in the field of regenerative medicine since they can act as enzymes in regulating physiological processes and enabling cell homeostasis, as well as the defense against pathogen aggression. In particular, cerium oxide nanoparticles (CeO2 NPs) stand as intriguing enzyme-mimicking nanoplatforms, owing to the reversible Ce+3/Ce+4 surface oxidation state. Moreover, surface functionalization leads to higher catalytic activity and selectivity, as well as more tunable enzyme-mimicking performances. Conjugation with melanin is an adequate strategy to boost and enrich CeO2 NPs biological features, because of melanin redox properties accounting for intrinsic antioxidant, antimicrobial and anti-inflammatory power. Herein, hybrid Melanin/CeO2 nanostructures were designed by simply coating the metal-oxide nanoparticles with melanin chains, obtained in-situ through ligand-to-metal charge transfer mechanism, according to a bioinspired approach. Obtained hybrid nanostructures underwent detailed physico-chemical characterization. Morphological and textural features were investigated through TEM, XRD and N2 physisorption. The nature of nanoparticle-melanin interaction was analyzed through FTIR, UV-vis and EPR spectroscopy. Melanin-coated hybrid nanostructures exhibited a relevant antioxidant activity, confirmed by a powerful quenching effect for DPPH radical, reaching 81 % inhibition at 33 μg/mL. A promising anti-inflammatory efficacy of the melanin-coated hybrid nanostructures was validated through a significant inhibition of BSA denaturation after 3 h. Meanwhile, the enzyme-mimicking activity was corroborated by a prolonged peroxidase activity after 8 h at 100 μg/mL and a relevant catalase-like action, by halving the H2O2 level in 30 min at 50 μg/mL. Antimicrobial assays attested that conjugation with melanin dramatically boosted CeO2 biocide activity against both Gram (-) and Gram (+) strains. Cytocompatibility tests demonstrated that the melanin coating not only enhanced the CeO2 nanostructures biomimicry, resulting in improved cell viability for human dermal fibroblast cells (HDFs), but mostly they proved that Melanin-CeO2 NPs were able to control the oxidative stress, modulating the production of nitrite and reactive oxygen species (ROS) levels in HDFs, under physiological conditions. Such remarkable outcomes make hybrid melanin-CeO2 nanozymes, promising redox-active interfaces for regenerative medicine.
Collapse
Affiliation(s)
- Giulio Pota
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Brigida Silvestri
- Department of Civil, Architectural and Environmental Engineering, University of Naples Federico II, Via Claudio 21, 80125 Naples, Italy
| | - Giuseppe Vitiello
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy; CSGI, Center for Colloid and Surface Science, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Noemi Gallucci
- Department of Chemical Sciences, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Rocco Di Girolamo
- Department of Chemical Sciences, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Stefania Scialla
- Institute for Polymers Composites and Biomaterials (IPCB) CNR Via Campi Flegrei 34, I-80078 Pozzuoli, NA, Italy
| | - Maria Grazia Raucci
- Institute for Polymers Composites and Biomaterials (IPCB) CNR Via Campi Flegrei 34, I-80078 Pozzuoli, NA, Italy
| | - Luigi Ambrosio
- Institute for Polymers Composites and Biomaterials (IPCB) CNR Via Campi Flegrei 34, I-80078 Pozzuoli, NA, Italy
| | - Michela Di Napoli
- Department of Biology, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Anna Zanfardino
- Department of Biology, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Mario Varcamonti
- Department of Biology, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Alessandro Pezzella
- Institute for Polymers Composites and Biomaterials (IPCB) CNR Via Campi Flegrei 34, I-80078 Pozzuoli, NA, Italy; Department of Physics "Ettore Pancini", University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy; National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti, 9, 50121 Florence, Italy
| | - Giuseppina Luciani
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy.
| |
Collapse
|
12
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
13
|
Rosado-Galindo H, Domenech M. Substrate topographies modulate the secretory activity of human bone marrow mesenchymal stem cells. Stem Cell Res Ther 2023; 14:208. [PMID: 37605275 PMCID: PMC10441765 DOI: 10.1186/s13287-023-03450-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/11/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) secrete a diversity of factors with broad therapeutic potential, yet current culture methods limit potency outcomes. In this study, we used topographical cues on polystyrene films to investigate their impact on the secretory profile and potency of bone marrow-derived MSCs (hBM-MSCs). hBM-MSCs from four donors were cultured on topographic substrates depicting defined roughness, curvature, grooves and various levels of wettability. METHODS The topographical PS-based array was developed using razor printing, polishing and plasma treatment methods. hBM-MSCs from four donors were purchased from RoosterBio and used in co-culture with peripheral blood mononuclear cells (PBMCs) from Cell Applications Inc. in an immunopotency assay to measure immunosuppressive capacity. Cells were cultured on low serum (2%) for 24-48 h prior to analysis. Image-based analysis was used for cell quantification and morphology assessment. Metabolic activity of BM-hMSCs was measured as the mitochondrial oxygen consumption rate using an extracellular flux analyzer. Conditioned media samples of BM-hMSCs were used to quantify secreted factors, and the data were analyzed using R statistics. Enriched bioprocesses were identify using the Gene Ontology tool enrichGO from the clusterprofiler. One-way and two-way ANOVAs were carried out to identify significant changes between the conditions. Results were deemed statistically significant for combined P < 0.05 for at least three independent experiments. RESULTS Cell viability was not significantly affected in the topographical substrates, and cell elongation was enhanced at least twofold in microgrooves and surfaces with a low contact angle. Increased cell elongation correlated with a metabolic shift from oxidative phosphorylation to a glycolytic state which is indicative of a high-energy state. Differential protein expression and gene ontology analyses identified bioprocesses enriched across donors associated with immune modulation and tissue regeneration. The growth of peripheral blood mononuclear cells (PBMCs) was suppressed in hBM-MSCs co-cultures, confirming enhanced immunosuppressive potency. YAP/TAZ levels were found to be reduced on these topographies confirming a mechanosensing effect on cells and suggesting a potential role in the immunomodulatory function of hMSCs. CONCLUSIONS This work demonstrates the potential of topographical cues as a culture strategy to improve the secretory capacity and enrich for an immunomodulatory phenotype in hBM-MSCs.
Collapse
Affiliation(s)
- Heizel Rosado-Galindo
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA
| | - Maribella Domenech
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
| |
Collapse
|
14
|
Maduka CV, Habeeb OM, Kuhnert MM, Hakun M, Goodman SB, Contag CH. Glycolytic reprogramming underlies immune cell activation by polyethylene wear particles. BIOMATERIALS ADVANCES 2023; 152:213495. [PMID: 37301057 DOI: 10.1016/j.bioadv.2023.213495] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 04/20/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Primary total joint arthroplasties (TJAs) are widely and successfully applied reconstructive procedures to treat end-stage arthritis. Nearly 50 % of TJAs are now performed in young patients, posing a new challenge: performing TJAs which last a lifetime. The urgency is justified because subsequent TJAs are costlier and fraught with higher complication rates, not to mention the toll taken on patients and their families. Polyethylene particles, generated by wear at joint articulations, drive aseptic loosening by inciting insidious inflammation associated with surrounding bone loss. Down modulating polyethylene particle-induced inflammation enhances integration of implants to bone (osseointegration), preventing loosening. A promising immunomodulation strategy could leverage immune cell metabolism, however, the role of immunometabolism in polyethylene particle-induced inflammation is unknown. Our findings reveal that immune cells exposed to sterile or contaminated polyethylene particles show fundamentally altered metabolism, resulting in glycolytic reprogramming. Inhibiting glycolysis controlled inflammation, inducing a pro-regenerative phenotype that could enhance osseointegration.
Collapse
Affiliation(s)
- Chima V Maduka
- Comparative Medicine & Integrative Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Oluwatosin M Habeeb
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Maxwell M Kuhnert
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Maxwell Hakun
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University, CA 94063, USA; Department of Bioengineering, Stanford University, CA 94305, USA
| | - Christopher H Contag
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA; Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 48864, USA.
| |
Collapse
|
15
|
Kang Z, Wu B, Zhang L, Liang X, Guo D, Yuan S, Xie D. Metabolic regulation by biomaterials in osteoblast. Front Bioeng Biotechnol 2023; 11:1184463. [PMID: 37324445 PMCID: PMC10265685 DOI: 10.3389/fbioe.2023.1184463] [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: 03/11/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
The repair of bone defects resulting from high-energy trauma, infection, or pathological fracture remains a challenge in the field of medicine. The development of biomaterials involved in the metabolic regulation provides a promising solution to this problem and has emerged as a prominent research area in regenerative engineering. While recent research on cell metabolism has advanced our knowledge of metabolic regulation in bone regeneration, the extent to which materials affect intracellular metabolic remains unclear. This review provides a detailed discussion of the mechanisms of bone regeneration, an overview of metabolic regulation in bone regeneration in osteoblasts and biomaterials involved in the metabolic regulation for bone regeneration. Furthermore, it introduces how materials, such as promoting favorable physicochemical characteristics (e.g., bioactivity, appropriate porosity, and superior mechanical properties), incorporating external stimuli (e.g., photothermal, electrical, and magnetic stimulation), and delivering metabolic regulators (e.g., metal ions, bioactive molecules like drugs and peptides, and regulatory metabolites such as alpha ketoglutarate), can affect cell metabolism and lead to changes of cell state. Considering the growing interests in cell metabolic regulation, advanced materials have the potential to help a larger population in overcoming bone defects.
Collapse
Affiliation(s)
- Zhengyang Kang
- Department of Orthopedics, The Second People’s Hospital of Panyu Guangzhou, Guangzhou, China
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Bin Wu
- Department of Orthopedics, The Second People’s Hospital of Panyu Guangzhou, Guangzhou, China
| | - Luhui Zhang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinzhi Liang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dong Guo
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shuai Yuan
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Denghui Xie
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangxi Key Laboratory of Bone and Joint Degeneration Diseases, Youjiang Medical University For Nationalities, Baise, China
| |
Collapse
|
16
|
Cai H, Ao Z, Tian C, Wu Z, Kaurich C, Chen Z, Gu M, Hohmann AG, Mackie K, Guo F. Engineering human spinal microphysiological systems to model opioid-induced tolerance. Bioact Mater 2023; 22:482-490. [PMID: 36330161 PMCID: PMC9618681 DOI: 10.1016/j.bioactmat.2022.10.007] [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: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
pioids are commonly used for treating chronic pain. However, with continued use, they may induce tolerance and/or hyperalgesia, which limits therapeutic efficacy. The human mechanisms of opioid-induced tolerance and hyperalgesia are significantly understudied, in part, because current models cannot fully recapitulate human pathology. Here, we engineered novel human spinal microphysiological systems (MPSs) integrated with plug-and-play neural activity sensing for modeling human nociception and opioid-induced tolerance. Each spinal MPS consists of a flattened human spinal cord organoid derived from human stem cells and a 3D printed organoid holder device for plug-and-play neural activity measurement. We found that the flattened organoid design of MPSs not only reduces hypoxia and necrosis in the organoids, but also promotes their neuron maturation, neural activity, and functional development. We further demonstrated that prolonged opioid exposure resulted in neurochemical correlates of opioid tolerance and hyperalgesia, as measured by altered neural activity, and downregulation of μ-opioid receptor expression of human spinal MPSs. The MPSs are scalable, cost-effective, easy-to-use, and compatible with commonly-used well-plates, thus allowing plug-and-play measurements of neural activity. We believe the MPSs hold a promising translational potential for studying human pain etiology, screening new treatments, and validating novel therapeutics for human pain medicine.
Collapse
Affiliation(s)
- Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Connor Kaurich
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| | - Zi Chen
- Department of Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, 02115, United States
| | - Mingxia Gu
- Division of Pulmonary Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, United States
- University of Cincinnati School of Medicine, Cincinnati, OH, 45229, United States
| | - Andrea G. Hohmann
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, United States
| |
Collapse
|
17
|
Hosseini FS, Abedini AA, Chen F, Whitfield T, Ude CC, Laurencin CT. Oxygen-Generating Biomaterials for Translational Bone Regenerative Engineering. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50721-50741. [PMID: 36988393 DOI: 10.1021/acsami.2c20715] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Successful regeneration of critical-size defects remains one of the significant challenges in regenerative engineering. These large-scale bone defects are difficult to regenerate and are often reconstructed with matrices that do not provide adequate oxygen levels to stem cells involved in the regeneration process. Hypoxia-induced necrosis predominantly occurs in the center of large matrices since the host tissue's local vasculature fails to provide sufficient nutrients and oxygen. Indeed, utilizing oxygen-generating materials can overcome the central hypoxic region, induce tissue in-growth, and increase the quality of life for patients with extensive tissue damage. This article reviews recent advances in oxygen-generating biomaterials for translational bone regenerative engineering. We discussed different oxygen-releasing and delivery methods, fabrication methods for oxygen-releasing matrices, biology, oxygen's role in bone regeneration, and emerging new oxygen delivery methods that could potentially be used for bone regenerative engineering.
Collapse
Affiliation(s)
- Fatemeh S Hosseini
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, Connecticut 06030, United States
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut 06030, United States
| | - Amir Abbas Abedini
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Feiyang Chen
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
| | - Taraje Whitfield
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, Connecticut 06030, United States
| | - Chinedu C Ude
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut 06030, United States
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, Connecticut 06030, United States
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut 06030, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
18
|
Xu B, Ye J, Fan BS, Wang X, Zhang JY, Song S, Song Y, Jiang WB, Wang X, Yu JK. Protein-spatiotemporal partition releasing gradient porous scaffolds and anti-inflammatory and antioxidant regulation remodel tissue engineered anisotropic meniscus. Bioact Mater 2023; 20:194-207. [PMID: 35702607 PMCID: PMC9160676 DOI: 10.1016/j.bioactmat.2022.05.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 01/02/2023] Open
Abstract
Meniscus is a wedge-shaped fibrocartilaginous tissue, playing important roles in maintaining joint stability and function. Meniscus injuries are difficult to heal and frequently progress into structural breakdown, which then leads to osteoarthritis. Regeneration of heterogeneous tissue engineering meniscus (TEM) continues to be a scientific and translational challenge. The morphology, tissue architecture, mechanical strength, and functional applications of the cultivated TEMs have not been able to meet clinical needs, which may due to the negligent attention on the importance of microenvironment in vitro and in vivo. Herein, we combined the 3D (three-dimensional)-printed gradient porous scaffolds, spatiotemporal partition release of growth factors, and anti-inflammatory and anti-oxidant microenvironment regulation of Ac2-26 peptide to prepare a versatile meniscus composite scaffold with heterogeneous bionic structures, excellent biomechanical properties and anti-inflammatory and anti-oxidant effects. By observing the results of cell activity and differentiation, and biomechanics under anti-inflammatory and anti-oxidant microenvironments in vitro, we explored the effects of anti-inflammatory and anti-oxidant microenvironments on construction of regional and functional heterogeneous TEM via the growth process regulation, with a view to cultivating a high-quality of TEM from bench to bedside. A polycaprolactone meniscus scaffold with the gradient porous architecture. Spatiotemporal partition release of two growth factors to promote heterogeneous phenotypes. Anti-inflammatory and antioxidant regulation by Ac2-26 peptide. Scaffold with biomimetic morphology, biomechanics, heterogeneity of native meniscus.
Collapse
|
19
|
Luo G, Wosinski P, Salazar-Noratto GE, Bensidhoum M, Bizios R, Marashi SA, Potier E, Sheng P, Petite H. Glucose Metabolism: Optimizing Regenerative Functionalities of Mesenchymal Stromal Cells Postimplantation. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:47-61. [PMID: 35754335 DOI: 10.1089/ten.teb.2022.0063] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mesenchymal stromal cells (MSCs) are considered promising candidates for regenerative medicine applications. Their clinical performance postimplantation, however, has been disappointing. This lack of therapeutic efficacy is most likely due to suboptimal formulations of MSC-containing material constructs. Tissue engineers, therefore, have developed strategies addressing/incorporating optimized cell, microenvironmental, biochemical, and biophysical cues/stimuli to enhance MSC-containing construct performance. Such approaches have had limited success because they overlooked that maintenance of MSC viability after implantation for a sufficient time is necessary for MSCs to develop their regenerative functionalities fully. Following a brief overview of glucose metabolism and regulation in MSCs, the present literature review includes recent pertinent findings that challenge old paradigms and notions. We hereby report that glucose is the primary energy substrate for MSCs, provides precursors for biomass generation, and regulates MSC functions, including proliferation and immunosuppressive properties. More importantly, glucose metabolism is central in controlling in vitro MSC expansion, in vivo MSC viability, and MSC-mediated angiogenesis postimplantation when addressing MSC-based therapies. Meanwhile, in silico models are highlighted for predicting the glucose needs of MSCs in specific regenerative medicine settings, which will eventually enable tissue engineers to design viable and potent tissue constructs. This new knowledge should be incorporated into developing novel effective MSC-based therapies. Impact statement The clinical use of mesenchymal stromal cells (MSCs) has been unsatisfactory due to the inability of MSCs to survive and be functional after implantation for sufficient periods to mediate directly or indirectly a successful regenerative tissue response. The present review summarizes the endeavors in the past, but, most importantly, reports the latest findings that elucidate underlying mechanisms and identify glucose metabolism as the crucial parameter in MSC survival and the subsequent functions pertinent to new tissue formation of importance in tissue regeneration applications. These latest findings justify further basic research and the impetus for developing new strategies to improve the modalities and efficacy of MSC-based therapies.
Collapse
Affiliation(s)
- Guotian Luo
- Université Paris Cité, CNRS, INSERM, B3OA, Paris, France.,École Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Pauline Wosinski
- Université Paris Cité, CNRS, INSERM, B3OA, Paris, France.,École Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Giuliana E Salazar-Noratto
- Université Paris Cité, CNRS, INSERM, B3OA, Paris, France.,École Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Morad Bensidhoum
- Université Paris Cité, CNRS, INSERM, B3OA, Paris, France.,École Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Rena Bizios
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Sayed-Amir Marashi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Esther Potier
- Université Paris Cité, CNRS, INSERM, B3OA, Paris, France.,École Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hervé Petite
- Université Paris Cité, CNRS, INSERM, B3OA, Paris, France.,École Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| |
Collapse
|
20
|
Recent Trends in the Development of Polyphosphazenes for Bio-applications. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00278-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
21
|
Major R, Wilczek G, Więcek J, Gawlikowski M, Plutecka H, Kasperkiewicz K, Kot M, Pomorska M, Ostrowski R, Kopernik M. Hemocompatibile Thin Films Assessed under Blood Flow Shear Forces. Molecules 2022; 27:molecules27175696. [PMID: 36080463 PMCID: PMC9458224 DOI: 10.3390/molecules27175696] [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: 06/20/2022] [Revised: 08/23/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to minimize the risk of life-threatening thromboembolism in the ventricle through the use of a new biomimetic heart valve based on metal-polymer composites. Finite volume element simulations of blood adhesion to the material were carried out, encompassing radial flow and the cone and plane test together with determination of the effect of boundary conditions. Both tilt-disc and bicuspid valves do not have optimized blood flow due to their design based on rigid valve materials (leaflet made of pyrolytic carbon). The main objective was the development of materials with specific properties dedicated to contact with blood. Materials were evaluated by dynamic tests using blood, concentrates, and whole human blood. Hemostability tests under hydrodynamic conditions were related to the mechanical properties of thin-film materials obtained from tribological tests. The quality of the coatings was high enough to avoid damage to the coating even as they were exposed up to maximum loading. Analysis towards blood concentrates of the hydrogenated carbon sample and the nitrogen-doped hydrogenated carbon sample revealed that the interaction of the coating with erythrocytes was the strongest. Hemocompatibility evaluation under hydrodynamic conditions confirmed very good properties of the developed coatings.
Collapse
Affiliation(s)
- Roman Major
- Institute of Metallurgy and Materials Science, Polish Academy of Sciences, 25 Reymonta St., 30-059 Cracow, Poland
| | - Grażyna Wilczek
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia, Bankowa St. 9, 40-007 Katowice, Poland
| | - Justyna Więcek
- Institute of Metallurgy and Materials Science, Polish Academy of Sciences, 25 Reymonta St., 30-059 Cracow, Poland
| | - Maciej Gawlikowski
- Faculty of Biomedical Engineering, Department of Biosensors and Processing of Biomedical Signals, Silesian University of Technology, Roosevelt St. 40, 41-800 Zabrze, Poland
| | - Hanna Plutecka
- Division of Molecular Biology and Clinical Genetics, Faculty of Medicine, Jagiellonian University Medical College, Skawinska St. 8, 31-066 Cracow, Poland
| | - Katarzyna Kasperkiewicz
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Jagiellońska St., 2840-032 Katowice, Poland
| | - Marcin Kot
- Faculty of Mechanical Engineering and Robotics, AGH University of Science and Technology, Al. Mickiewicza 30, 30-059 Cracow, Poland
| | - Małgorzata Pomorska
- Institute of Metallurgy and Materials Science, Polish Academy of Sciences, 25 Reymonta St., 30-059 Cracow, Poland
| | - Roman Ostrowski
- Institute of Optoelectronics, Military University of Technology, Gen. S. Kaliskiego St. 2, 00-908 Warsaw, Poland
| | - Magdalena Kopernik
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, Al. Mickiewicza 30, 30-059 Cracow, Poland
- Correspondence: ; Tel.: +48-12-617-51-26
| |
Collapse
|
22
|
Tan X, Gerhard E, Wang Y, Tran RT, Xu H, Yan S, Rizk EB, Armstrong AD, Zhou Y, Du J, Bai X, Yang J. Development of Biodegradable Osteopromotive Citrate-Based Bone Putty. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203003. [PMID: 35717669 PMCID: PMC9463100 DOI: 10.1002/smll.202203003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Indexed: 05/30/2023]
Abstract
The burden of bone fractures demands development of effective biomaterial solutions, while additional acute events such as noncompressible bleeding further motivate the search for multi-functional implants to avoid complications including osseous hemorrhage, infection, and nonunion. Bone wax has been widely used in orthopedic bleeding control due to its simplicity of use and conformation to irregular defects; however, its nondegradability results in impaired bone healing, risk of infection, and significant inflammatory responses. Herein, a class of intrinsically fluorescent, osteopromotive citrate-based polymer/hydroxyapatite (HA) composites (BPLP-Ser/HA) as a highly malleable press-fit putty is designed. BPLP-Ser/HA putty displays mechanics replicating early nonmineralized bone (initial moduli from ≈2-500 kPa), hydration induced mechanical strengthening in physiological conditions, tunable degradation rates (over 2 months), low swelling ratios (<10%), clotting and hemostatic sealing potential (resistant to blood pressure for >24 h) and significant adhesion to bone (≈350-550 kPa). Simultaneously, citrate's bioactive properties result in antimicrobial (≈100% and 55% inhibition of S. aureus and E. coli) and osteopromotive effects. Finally, BPLP-Ser/HA putty demonstrates in vivo regeneration in a critical-sized rat calvaria model equivalent to gold standard autograft. BPLP-Ser/HA putty represents a simple, off-the-shelf solution to the combined challenges of acute wound management and subsequent bone regeneration.
Collapse
Affiliation(s)
- Xinyu Tan
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
- Academy of Orthopedics, Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, 510280, China
| | - Ethan Gerhard
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yuqi Wang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Richard T. Tran
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hui Xu
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Su Yan
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Elias B. Rizk
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - April D. Armstrong
- Department of Orthopaedics and Rehabilitation, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Yuxiao Zhou
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jing Du
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Academy of Orthopedics, Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, 510280, China
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
23
|
Ordoño J, Pérez-Amodio S, Ball K, Aguirre A, Engel E. The generation of a lactate-rich environment stimulates cell cycle progression and modulates gene expression on neonatal and hiPSC-derived cardiomyocytes. BIOMATERIALS ADVANCES 2022; 139:213035. [PMID: 35907761 PMCID: PMC11061846 DOI: 10.1016/j.bioadv.2022.213035] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
In situ tissue engineering strategies are a promising approach to activate the endogenous regenerative potential of the cardiac tissue helping the heart to heal itself after an injury. However, the current use of complex reprogramming vectors for the activation of reparative pathways challenges the easy translation of these therapies into the clinic. Here, we evaluated the response of mouse neonatal and human induced pluripotent stem cell-derived cardiomyocytes to the presence of exogenous lactate, thus mimicking the metabolic environment of the fetal heart. An increase in cardiomyocyte cell cycle activity was observed in the presence of lactate, as determined through Ki67 and Aurora-B kinase. Gene expression and RNA-sequencing data revealed that cardiomyocytes incubated with lactate showed upregulation of BMP10, LIN28 or TCIM in tandem with downregulation of GRIK1 or DGKK among others. Lactate also demonstrated a capability to modulate the production of inflammatory cytokines on cardiac fibroblasts, reducing the production of Fas, Fraktalkine or IL-12p40, while stimulating IL-13 and SDF1a. In addition, the generation of a lactate-rich environment improved ex vivo neonatal heart culture, by affecting the contractile activity and sarcomeric structures and inhibiting epicardial cell spreading. Our results also suggested a common link between the effect of lactate and the activation of hypoxia signaling pathways. These findings support a novel use of lactate in cardiac tissue engineering, modulating the metabolic environment of the heart and thus paving the way to the development of lactate-releasing platforms for in situ cardiac regeneration.
Collapse
Affiliation(s)
- Jesús Ordoño
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain; CIBER Bioengineering, Biomaterials and Nanotechnology, Spain
| | - Soledad Pérez-Amodio
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain; CIBER Bioengineering, Biomaterials and Nanotechnology, Spain; IMEM-BRT Group, Dpt. Material Science and Engineering, Universitat Politecnica de Catalunya (UPC), Barcelona, Spain
| | - Kristen Ball
- Regenerative Biology and cell Reprogramming Laboratory, Institute for Quantitative Health Sciences and Engineering (IQ), Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, MI, USA
| | - Aitor Aguirre
- Regenerative Biology and cell Reprogramming Laboratory, Institute for Quantitative Health Sciences and Engineering (IQ), Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, MI, USA
| | - Elisabeth Engel
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain; CIBER Bioengineering, Biomaterials and Nanotechnology, Spain; IMEM-BRT Group, Dpt. Material Science and Engineering, Universitat Politecnica de Catalunya (UPC), Barcelona, Spain.
| |
Collapse
|
24
|
Hong IS. Enhancing Stem Cell-Based Therapeutic Potential by Combining Various Bioengineering Technologies. Front Cell Dev Biol 2022; 10:901661. [PMID: 35865629 PMCID: PMC9294278 DOI: 10.3389/fcell.2022.901661] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/17/2022] [Indexed: 12/05/2022] Open
Abstract
Stem cell-based therapeutics have gained tremendous attention in recent years due to their wide range of applications in various degenerative diseases, injuries, and other health-related conditions. Therapeutically effective bone marrow stem cells, cord blood- or adipose tissue-derived mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and more recently, induced pluripotent stem cells (iPSCs) have been widely reported in many preclinical and clinical studies with some promising results. However, these stem cell-only transplantation strategies are hindered by the harsh microenvironment, limited cell viability, and poor retention of transplanted cells at the sites of injury. In fact, a number of studies have reported that less than 5% of the transplanted cells are retained at the site of injury on the first day after transplantation, suggesting extremely low (<1%) viability of transplanted cells. In this context, 3D porous or fibrous national polymers (collagen, fibrin, hyaluronic acid, and chitosan)-based scaffold with appropriate mechanical features and biocompatibility can be used to overcome various limitations of stem cell-only transplantation by supporting their adhesion, survival, proliferation, and differentiation as well as providing elegant 3-dimensional (3D) tissue microenvironment. Therefore, stem cell-based tissue engineering using natural or synthetic biomimetics provides novel clinical and therapeutic opportunities for a number of degenerative diseases or tissue injury. Here, we summarized recent studies involving various types of stem cell-based tissue-engineering strategies for different degenerative diseases. We also reviewed recent studies for preclinical and clinical use of stem cell-based scaffolds and various optimization strategies.
Collapse
Affiliation(s)
- In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Seongnam, South Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Seongnam, South Korea
- *Correspondence: In-Sun Hong,
| |
Collapse
|
25
|
Zhong J, Wang H, Yang K, Wang H, Duan C, Ni N, An L, Luo Y, Zhao P, Gou Y, Sheng S, Shi D, Chen C, Wagstaff W, Hendren-Santiago B, Haydon RC, Luu HH, Reid RR, Ho SH, Ameer GA, Shen L, He TC, Fan J. Reversibly immortalized keratinocytes (iKera) facilitate re-epithelization and skin wound healing: Potential applications in cell-based skin tissue engineering. Bioact Mater 2022; 9:523-540. [PMID: 34820586 PMCID: PMC8581279 DOI: 10.1016/j.bioactmat.2021.07.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Skin injury is repaired through a multi-phase wound healing process of tissue granulation and re-epithelialization. Any failure in the healing process may lead to chronic non-healing wounds or abnormal scar formation. Although significant progress has been made in developing novel scaffolds and/or cell-based therapeutic strategies to promote wound healing, effective management of large chronic skin wounds remains a clinical challenge. Keratinocytes are critical to re-epithelialization and wound healing. Here, we investigated whether exogenous keratinocytes, in combination with a citrate-based scaffold, enhanced skin wound healing. We first established reversibly immortalized mouse keratinocytes (iKera), and confirmed that the iKera cells expressed keratinocyte markers, and were responsive to UVB treatment, and were non-tumorigenic. In a proof-of-principle experiment, we demonstrated that iKera cells embedded in citrate-based scaffold PPCN provided more effective re-epithelialization and cutaneous wound healing than that of either PPCN or iKera cells alone, in a mouse skin wound model. Thus, these results demonstrate that iKera cells may serve as a valuable skin epithelial source when, combining with appropriate biocompatible scaffolds, to investigate cutaneous wound healing and skin regeneration.
Collapse
Affiliation(s)
- Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hao Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Ke Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- The Pediatric Research Institute, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Huifeng Wang
- Biomedical Engineering Department, Northwestern University, Evanston, IL, 60208, USA
| | - Chongwen Duan
- Biomedical Engineering Department, Northwestern University, Evanston, IL, 60208, USA
| | - Na Ni
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Liqin An
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yetao Luo
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Piao Zhao
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shiyan Sheng
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Bryce Hendren-Santiago
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL, 60208, USA
| | - Sherwin H. Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Guillermo A. Ameer
- Biomedical Engineering Department, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL, 60208, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60616, USA
| | - Le Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL, 60208, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| |
Collapse
|
26
|
Yan Z, Chen C, Rosso G, Qian Y, Fan C. Two-Dimensional Nanomaterials for Peripheral Nerve Engineering: Recent Advances and Potential Mechanisms. Front Bioeng Biotechnol 2021; 9:746074. [PMID: 34820361 PMCID: PMC8606639 DOI: 10.3389/fbioe.2021.746074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/19/2021] [Indexed: 01/19/2023] Open
Abstract
Peripheral nerve tissues possess the ability to regenerate within artificial nerve scaffolds, however, despite the advance of biomaterials that support nerve regeneration, the functional nerve recovery remains unsatisfactory. Importantly, the incorporation of two-dimensional nanomaterials has shown to significantly improve the therapeutic effect of conventional nerve scaffolds. In this review, we examine whether two-dimensional nanomaterials facilitate angiogenesis and thereby promote peripheral nerve regeneration. First, we summarize the major events occurring after peripheral nerve injury. Second, we discuss that the application of two-dimensional nanomaterials for peripheral nerve regeneration strategies by facilitating the formation of new vessels. Then, we analyze the mechanism that the newly-formed capillaries directionally and metabolically support neuronal regeneration. Finally, we prospect that the two-dimensional nanomaterials should be a potential solution to long range peripheral nerve defect. To further enhance the therapeutic effects of two-dimensional nanomaterial, strategies which help remedy the energy deficiency after peripheral nerve injury could be a viable solution.
Collapse
Affiliation(s)
- Zhiwen Yan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China.,Youth Science and Technology Innovation Studio, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Chen
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Gonzalo Rosso
- Max Planck Institute for the Science of Light, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.,Institute of Physiology II, University of Münster, Münster, Germany
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China.,Youth Science and Technology Innovation Studio, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China.,Youth Science and Technology Innovation Studio, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Han P, Vaquette C, Abdal-hay A, Ivanovski S. The Mechanosensing and Global DNA Methylation of Human Osteoblasts on MEW Fibers. NANOMATERIALS 2021; 11:nano11112943. [PMID: 34835707 PMCID: PMC8621030 DOI: 10.3390/nano11112943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/02/2023]
Abstract
Cells interact with 3D fibrous platform topography via a nano-scaled focal adhesion complex, and more research is required on how osteoblasts sense and respond to random and aligned fibers through nano-sized focal adhesions and their downstream events. The present study assessed human primary osteoblast cells’ sensing and response to random and aligned medical-grade polycaprolactone (PCL) fibrous 3D scaffolds fabricated via the melt electrowriting (MEW) technique. Cells cultured on a tissue culture plate (TCP) were used as 2D controls. Compared to 2D TCP, 3D MEW fibrous substrates led to immature vinculin focal adhesion formation and significantly reduced nuclear localization of the mechanosensor-yes-associated protein (YAP). Notably, aligned MEW fibers induced elongated cell and nucleus shape and highly activated global DNA methylation of 5-methylcytosine, 5-hydroxymethylcytosine, and N-6 methylated deoxyadenosine compared to the random fibers. Furthermore, although osteogenic markers (osterix-OSX and bone sialoprotein-BSP) were significantly enhanced in PCL-R and PCL-A groups at seven days post-osteogenic differentiation, calcium deposits on all seeded samples did not show a difference after normalizing for DNA content after three weeks of osteogenic induction. Overall, our study linked 3D extracellular fiber alignment to nano-focal adhesion complex, nuclear mechanosensing, DNA epigenetics at an early point (24 h), and longer-term changes in osteoblast osteogenic differentiation.
Collapse
Affiliation(s)
- Pingping Han
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, School of Dentistry, The University of Queensland, Brisbane, QLD 4006, Australia;
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia; (C.V.); (A.A.-h.)
| | - Cedryck Vaquette
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia; (C.V.); (A.A.-h.)
| | - Abdalla Abdal-hay
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia; (C.V.); (A.A.-h.)
- Department of Mechanical Engineering, Faculty of Engineering, South Valley University, Qena 83523, Egypt
| | - Sašo Ivanovski
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, School of Dentistry, The University of Queensland, Brisbane, QLD 4006, Australia;
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia; (C.V.); (A.A.-h.)
- Correspondence:
| |
Collapse
|
28
|
Sthijns MMJPE, van Blitterswijk CA, LaPointe VLS. Synthetic Materials that Affect the Extracellular Matrix via Cellular Metabolism and Responses to a Metabolic State. Front Bioeng Biotechnol 2021; 9:742132. [PMID: 34708025 PMCID: PMC8542861 DOI: 10.3389/fbioe.2021.742132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
In regenerative medicine and tissue engineering, many materials are developed to mimic the extracellular matrix (ECM). However, these ECM-mimicking materials do not yet completely recapitulate the diversity and complexity of biological tissue-specific ECM. In this review, an alternative strategy is proposed to generate ECM, namely synthesizing a material that functions as a drug delivery system, releasing molecules that target cellular metabolic pathways and thereby stimulate the local cells to create their own ECM. This is based on the fact that ECM synthesis, modification, composition, signaling, stiffness, and degradation are modulated by cellular metabolism. Metabolism can be targeted at different levels, ranging from modulating the availability of substrates or co-factors to regulating the activity of essential transcription factors. Depending on the drug of interest, its characteristics, mechanism of action, cellular target, and application, a different drug delivery system should be designed. Metabolic drugs modulating the ECM require cellular uptake for their function, therefore reversible linkers are recommended. Preferably the metabolic modulators are only released when needed, which will be upon a specific metabolic state, a change in ECM stiffness, or ECM remodeling. Therefore, reversible linkers that respond to an environmental stimulus could be incorporated. All in all, a novel strategy is suggested to develop a tissue-specific ECM by generating a synthetic material that releases metabolic molecules modulating the ECM. Various ways to modulate the ECM properties via the metabolism are reviewed and guidelines for the development of these materials are provided.
Collapse
Affiliation(s)
- Mireille M J P E Sthijns
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands.,Department of Food Innovation and Health at the Centre of Healthy Eating and Food Innovation, Maastricht University, Maastricht, Netherlands
| | - Clemens A van Blitterswijk
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
29
|
Siddiqui Z, Sarkar B, Kim KK, Kumar A, Paul R, Mahajan A, Grasman JM, Yang J, Kumar VA. Self-assembling Peptide Hydrogels Facilitate Vascularization in Two-Component Scaffolds. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 422:130145. [PMID: 34054331 PMCID: PMC8158327 DOI: 10.1016/j.cej.2021.130145] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
One of the major constraints against using polymeric scaffolds as tissue-regenerative matrices is a lack of adequate implant vascularization. Self-assembling peptide hydrogels can sequester small molecules and biological macromolecules, and they can support infiltrating cells in vivo. Here we demonstrate the ability of self-assembling peptide hydrogels to facilitate angiogenic sprouting into polymeric scaffolds after subcutaneous implantation. We constructed two-component scaffolds that incorporated microporous polymeric scaffolds and viscoelastic nanoporous peptide hydrogels. Nanofibrous hydrogels modified the biocompatibility and vascular integration of polymeric scaffolds with microscopic pores (pore diameters: 100-250 μm). In spite of similar amphiphilic sequences, charges, secondary structures, and supramolecular nanostructures, two soft hydrogels studied herein had different abilities to aid implant vascularization, but had similar levels of cellular infiltration. The functional difference of the peptide hydrogels was predicted by the difference in the bioactive moieties inserted into the primary sequences of the peptide monomers. Our study highlights the utility of soft supramolecular hydrogels to facilitate host-implant integration and control implant vascularization in biodegradable polyester scaffolds in vivo. Our study provides useful tools in designing multi-component regenerative scaffolds that recapitulate vascularized architectures of native tissues.
Collapse
Affiliation(s)
- Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Arjun Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Reshma Paul
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Aryan Mahajan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Jonathan M. Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Jian Yang
- Department of Biomedical Engineering, Huck Institutes of The Life Sciences, Materials Research Institute, Pennsylvania State University, University Park, PA, USA
| | - Vivek A. Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
- Department of Chemical & Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
30
|
Chen L, Yu Q, Jia Y, Xu M, Wang Y, Wang J, Wen T, Wang L. Micro-and-nanometer topological gradient of block copolymer fibrous scaffolds towards region-specific cell regulation. J Colloid Interface Sci 2021; 606:248-260. [PMID: 34390992 DOI: 10.1016/j.jcis.2021.08.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022]
Abstract
Regulating cell behavior and function by surface topography has drawn significant attention in tissue engineering. Herein, a gradient fibrous scaffold comprising anisotropic aligned fibers and isotropic annealed fibers was developed to provide a controllable direction of cell migration, adhesion, and spreading. The electrospun aligned fibers were engraved to create surface gradients with micro-and-nanometer roughness through block copolymer (BCP) self-assembly induced by selective solvent vapor annealing (SVA). The distinct manipulation of cell behavior by annealed fibrous scaffolds with tailored self-assembled nanostructure and welded fibrous microstructure has been illustrated by in situ/ex situ small angle X-ray scattering (SAXS), scanning electron microscopy (SEM), atomic force microscopy (AFM) and in vitro cell culture. Further insights into the effect of integrated gradient fibrous scaffold were gained at the level of protein expression. From the perspective of gradient topology, this region-specific scaffold based on BCP fibers shows the prospect of guiding cell migration, adhesion and spreading and provides a generic method for designing biomaterials for tissue-engineering.
Collapse
Affiliation(s)
- Lei Chen
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Qianqian Yu
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Yifan Jia
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Mengmeng Xu
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Yingying Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jing Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Tao Wen
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Linge Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
31
|
Metabolomics in Bone Research. Metabolites 2021; 11:metabo11070434. [PMID: 34357328 PMCID: PMC8303949 DOI: 10.3390/metabo11070434] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Identifying the changes in endogenous metabolites in response to intrinsic and extrinsic factors has excellent potential to obtain an understanding of cells, biofluids, tissues, or organisms' functions and interactions with the environment. The advantages provided by the metabolomics strategy have promoted studies in bone research fields, including an understanding of bone cell behaviors, diagnosis and prognosis of diseases, and the development of treatment methods such as implanted biomaterials. This review article summarizes the metabolism changes during osteogenesis, osteoclastogenesis, and immunoregulation in hard tissue. The second section of this review is dedicated to describing and discussing metabolite changes in the most relevant bone diseases: osteoporosis, bone injuries, rheumatoid arthritis, and osteosarcoma. We consolidated the most recent finding of the metabolites and metabolite pathways affected by various bone disorders. This collection can serve as a basis for future metabolomics-driven bone research studies to select the most relevant metabolites and metabolic pathways. Additionally, we summarize recent metabolic studies on metabolomics for the development of bone disease treatment including biomaterials for bone engineering. With this article, we aim to provide a comprehensive summary of metabolomics in bone research, which can be helpful for interdisciplinary researchers, including material engineers, biologists, and clinicians.
Collapse
|
32
|
Claudio-Rizo JA, Escobedo-Estrada N, Carrillo-Cortes SL, Cabrera-Munguía DA, Flores-Guía TE, Becerra-Rodriguez JJ. Highly absorbent hydrogels comprised from interpenetrated networks of alginate-polyurethane for biomedical applications. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:70. [PMID: 34117933 PMCID: PMC8197714 DOI: 10.1007/s10856-021-06544-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/31/2021] [Indexed: 06/12/2023]
Abstract
Developing new approaches to improve the swelling, degradation rate, and mechanical properties of alginate hydrogels without compromising their biocompatibility for biomedical applications represents a potential area of research. In this work, the generation of interpenetrated networks (IPN) comprised from alginate-polyurethane in an aqueous medium is proposed to design hydrogels with tailored properties for biomedical applications. Aqueous polyurethane (PU) dispersions can crosslink and interpenetrate alginate chains, forming amide bonds that allow the structure and water absorption capacity of these novel hydrogels to be regulated. In this sense, this work focuses on studying the relation of the PU concentration on the properties of these hydrogels. The results indicate that the crosslinking of the alginate with PU generates IPN hydrogels with a crystalline structure characterized by a homogeneous smooth surface with high capacity to absorb water, tailoring the degradation rate, thermal decomposition, and storage module, not altering the native biocompatibility of alginate, providing character to inhibit the growth of E. coli and increasing also its hemocompatibility. The IPN hydrogels that include 20 wt.% of PU exhibit a reticulation index of 46 ± 4%, swelling capacity of 545 ± 13% at 7 days of incubation at physiological pH, resistance to both acidic and neutral hydrolytic degradation, mechanical improvement of 91 ± 1%, and no cytotoxicity for monocytes and fibroblasts growing for up to 72 h of incubation. These results indicate that these novel hydrogels can be used for successful biomedical applications in the design of wound healing dressings.
Collapse
Affiliation(s)
- Jesús A Claudio-Rizo
- Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Ing. J. Cárdenas Valdez S/N, República, 25280, Saltillo, Coahuila, México.
| | - Nallely Escobedo-Estrada
- Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Ing. J. Cárdenas Valdez S/N, República, 25280, Saltillo, Coahuila, México
| | - Sara L Carrillo-Cortes
- Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Ing. J. Cárdenas Valdez S/N, República, 25280, Saltillo, Coahuila, México
| | - Denis A Cabrera-Munguía
- Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Ing. J. Cárdenas Valdez S/N, República, 25280, Saltillo, Coahuila, México
| | - Tirso E Flores-Guía
- Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Ing. J. Cárdenas Valdez S/N, República, 25280, Saltillo, Coahuila, México
| | - Juan J Becerra-Rodriguez
- Universidad Politécnica de Pénjamo, Carretera Irapuato - La Piedad Km 44, Pénjamo, 36921, Guanajuato, México
| |
Collapse
|
33
|
Tang Z, Wang X, Yang J, Song X, Huang Y, Chen C, Yang H, Fu Z, Gong X, Chen G. Microconvex Dot-Featured Silk Fibroin Films for Promoting Human Umbilical Vein Endothelial Cell Angiogenesis via Enhancing the Expression of bFGF and VEGF. ACS Biomater Sci Eng 2021; 7:2420-2429. [PMID: 33878261 DOI: 10.1021/acsbiomaterials.0c01647] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Insufficient vascularization of grafts often leads to delayed tissue ingrowth and impaired tissue function in tissue engineering. The surface topography of grafts plays critical roles in angiogenesis. In the present study, we prepared silk fibroin (SF)-based microtopography films with the number of convex dots ranging from 37 to 4835/mm2. The convex dot-featured topography surfaces were characterized by scanning electron microscopy, a Profilm3D optical profilometer, atomic force microscopy, and a contact angle goniometer. The effect of microtopographic films on the proliferation, adhesion, and expression of angiogenic factors of human umbilical vein endothelial cells (HUVECs) was investigated. Our results demonstrated that the SF film surface with 2899 convex dots/mm2 significantly enhanced adhesion, viability, and levels of vascular endothelial growth factors and basic fibroblast growth factors of HUVECs and significantly downregulated the level of α-SMA in human aortic smooth muscle cells, indicating that the microtopographic films could promote angiogenesis. Furthermore, in vitro results showed that HUVEC proliferation was positively correlated with yes-associated protein (YAP) activation, suggesting that the enhanced angiogenesis was mediated via the YAP pathway. Finally, mice subcutaneous embedding model results indicated that the SF film surface with 2899 convex dots/mm2 could significantly enhance angiogenesis in vivo. Altogether, our results showed that the SF film surface with 2899 convex dots/mm2 promoted the angiogenesis of HUVECs and offered a novel angiogenesis-promoting strategy of implant surface design for tissue engineering.
Collapse
Affiliation(s)
- Zhexiong Tang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xin Wang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Junjun Yang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiongbo Song
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yang Huang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Hao Yang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhenlan Fu
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoyuan Gong
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Guangxing Chen
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
34
|
Chen Y, Huang J, Liu J, Wei Y, Yang X, Lei L, Chen L, Wu Y, Gou Z. Tuning filament composition and microstructure of 3D-printed bioceramic scaffolds facilitate bone defect regeneration and repair. Regen Biomater 2021; 8:rbab007. [PMID: 33738121 PMCID: PMC7955715 DOI: 10.1093/rb/rbab007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/03/2021] [Accepted: 01/17/2021] [Indexed: 12/24/2022] Open
Abstract
It is still a challenge to optimize the component distribution and microporous structures in scaffolds for tailoring biodegradation (ion releasing) and enhancing bone defect repair within an expected time stage. Herein, the core–shell-typed nonstoichiometric wollastonite (4% and 10% Mg-doping calcium silicate; CSiMg4, CSiMg10) macroporous scaffolds with microporous shells (adding ∼10 μm PS microspheres into shell-layer slurry) were fabricated via 3D printing. The initial mechanical properties and bio-dissolution (ion releasing) in vitro, and osteogenic capacity in vivo of the bioceramic scaffolds were evaluated systematically. It was shown that endowing high-density micropores in the sparingly dissolvable CSiMg10 or dissolvable CSiMg4 shell layer inevitably led to nearly 30% reduction of compressive strength, but such micropores could readily tune the ion release behaviour of the scaffolds (CSiMg4@CSiMg10 vs. CSiMg4@CSiMg10-p; CSiMg10@CSiMg4 vs. CSiMg10@CSiMg4-p). Based on the in rabbit femoral bone defect repair model, the 3D μCT reconstruction and histological observation demonstrated that the CSiMg4@CSiMg10-p scaffolds displayed markedly higher osteogenic capability than the other scaffolds after 12 weeks of implantation. It demonstrated that core–shell bioceramic 3D printing technique can be developed to fabricate single-phase or biphasic bioactive ceramic scaffolds with accurately tailored filament biodegradation for promoting bone defect regeneration and repair in some specific pathological conditions.
Collapse
Affiliation(s)
- Yi Chen
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jiaping Huang
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jiamei Liu
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yingming Wei
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xianyan Yang
- Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, China
| | - Lihong Lei
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lili Chen
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yanmin Wu
- Department of Stomotology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhongru Gou
- Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
35
|
Salihu R, Abd Razak SI, Ahmad Zawawi N, Rafiq Abdul Kadir M, Izzah Ismail N, Jusoh N, Riduan Mohamad M, Hasraf Mat Nayan N. Citric acid: A green cross-linker of biomaterials for biomedical applications. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110271] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
36
|
Amin Yavari S, Castenmiller SM, van Strijp JAG, Croes M. Combating Implant Infections: Shifting Focus from Bacteria to Host. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002962. [PMID: 32914481 DOI: 10.1002/adma.202002962] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Indexed: 05/06/2023]
Abstract
The widespread use of biomaterials to support or replace body parts is increasingly threatened by the risk of implant-associated infections. In the quest for finding novel anti-infective biomaterials, there generally has been a one-sided focus on biomaterials with direct antibacterial properties, which leads to excessive use of antibacterial agents, compromised host responses, and unpredictable effectiveness in vivo. This review sheds light on how host immunomodulation, rather than only targeting bacteria, can endow biomaterials with improved anti-infective properties. How antibacterial surface treatments are at risk to be undermined by biomaterial features that dysregulate the protection normally provided by critical immune cell subsets, namely, neutrophils and macrophages, is discussed. Accordingly, how the precise modification of biomaterial surface biophysical cues, or the incorporation of immunomodulatory drug delivery systems, can render biomaterials with the necessary immune-compatible and immune-protective properties to potentiate the host defense mechanisms is reviewed. Within this context, the protective role of host defense peptides, metallic particles, quorum sensing inhibitors, and therapeutic adjuvants is discussed. The highlighted immunomodulatory strategies may lay a foundation to develop anti-infective biomaterials, while mitigating the increasing threat of antibacterial drug resistance.
Collapse
Affiliation(s)
- Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Suzanne M Castenmiller
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| |
Collapse
|
37
|
Arrés M, Salama M, Rechena D, Paradiso P, Reis L, Alves MM, Botelho do Rego AM, Carmezim MJ, Vaz MF, Deus AM, Santos C. Surface and mechanical properties of a nanostructured citrate hydroxyapatite coating on pure titanium. J Mech Behav Biomed Mater 2020; 108:103794. [PMID: 32469718 DOI: 10.1016/j.jmbbm.2020.103794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/02/2020] [Accepted: 04/12/2020] [Indexed: 01/13/2023]
Abstract
The presence of a biomimetic HAP coating on titanium surface, which reduces the structural stiffness, is essential to improve implants biocompatibility and osteointegration. In this study, new citrate-HAP (cHAP) coatings were produced by a simple hydrothermal method on pure titanium (Ti) surface, without requiring any additional pretreatment on this metal surface. The formed cHAP coatings consisting of nanorod-like hydroxyapatite particles, conferred nanoroughness and wettability able to endow improved biological responses. Indeed, the presence of citrate species in the precipitate medium seems to be responsible for controlling the morphology of the new coatings. The presence of citrate groups on the surface of cHAP coatings, identified by chemical composition analysis, due to their implication in bone metabolism can additionally bring an add-value for bone implant applications. From a mechanical point of view, the Finite Element algorithm showing that cHAP coatings tend to decrease the mechanical stress at pure Ti, further favors these new coatings applicability. Overall, the simple and expedite strategy used to developed new biomimetic coatings of citrate-HAP resulted in improved physicochemical, morphological and mechanical properties of Ti, which can endeavor improved implantable materials in bone healing surgical procedures.
Collapse
Affiliation(s)
- Mar Arrés
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Mariana Salama
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Diogo Rechena
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Patrizia Paradiso
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Luis Reis
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Marta M Alves
- CQE, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Botelho do Rego
- CQFM (IN) and BSIRG (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Maria J Carmezim
- CQE, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; ESTSetubal, CDP2T, Instituto Politécnico de Setúbal, Setúbal, Portugal
| | - Maria Fátima Vaz
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; CQE, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Augusto M Deus
- CQE, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; CeFEMA, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Santos
- CQE, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; ESTSetubal, CDP2T, Instituto Politécnico de Setúbal, Setúbal, Portugal.
| |
Collapse
|
38
|
Ao Z, Cai H, Havert DJ, Wu Z, Gong Z, Beggs JM, Mackie K, Guo F. One-Stop Microfluidic Assembly of Human Brain Organoids To Model Prenatal Cannabis Exposure. Anal Chem 2020; 92:4630-4638. [PMID: 32070103 DOI: 10.1021/acs.analchem.0c00205] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Prenatal cannabis exposure (PCE) influences human brain development, but it is challenging to model PCE using animals and current cell culture techniques. Here, we developed a one-stop microfluidic platform to assemble and culture human cerebral organoids from human embryonic stem cells (hESC) to investigate the effect of PCE on early human brain development. By incorporating perfusable culture chambers, air-liquid interface, and one-stop protocol, this microfluidic platform can simplify the fabrication procedure and produce a large number of organoids (169 organoids per 3.5 cm × 3.5 cm device area) without fusion, as compared with conventional fabrication methods. These one-stop microfluidic assembled cerebral organoids not only recapitulate early human brain structure, biology, and electrophysiology but also have minimal size variation and hypoxia. Under on-chip exposure to the psychoactive cannabinoid, Δ-9-tetrahydrocannabinol (THC), cerebral organoids exhibited reduced neuronal maturation, downregulation of cannabinoid receptor type 1 (CB1) receptors, and impaired neurite outgrowth. Moreover, transient on-chip THC treatment also decreased spontaneous firing in these organoids. This one-stop microfluidic technique enables a simple, scalable, and repeatable organoid culture method that can be used not only for human brain organoids but also for many other human organoids including liver, kidney, retina, and tumor organoids. This technology could be widely used in modeling brain and other organ development, developmental disorders, developmental pharmacology and toxicology, and drug screening.
Collapse
Affiliation(s)
- Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Daniel J Havert
- Department of Physics, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhiyi Gong
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - John M Beggs
- Department of Physics, Indiana University, Bloomington, Indiana 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
39
|
Heng W, Bhavsar M, Han Z, Barker JH. Effects of Electrical Stimulation on Stem Cells. Curr Stem Cell Res Ther 2020; 15:441-448. [PMID: 31995020 DOI: 10.2174/1574888x15666200129154747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 11/22/2022]
Abstract
Recent interest in developing new regenerative medicine- and tissue engineering-based treatments has motivated researchers to develop strategies for manipulating stem cells to optimize outcomes in these potentially, game-changing treatments. Cells communicate with each other, and with their surrounding tissues and organs via electrochemical signals. These signals originate from ions passing back and forth through cell membranes and play a key role in regulating cell function during embryonic development, healing, and regeneration. To study the effects of electrical signals on cell function, investigators have exposed cells to exogenous electrical stimulation and have been able to increase, decrease and entirely block cell proliferation, differentiation, migration, alignment, and adherence to scaffold materials. In this review, we discuss research focused on the use of electrical stimulation to manipulate stem cell function with a focus on its incorporation in tissue engineering-based treatments.
Collapse
Affiliation(s)
- Wang Heng
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| | - Mit Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| | - Zhihua Han
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| | - John H Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|