1
|
Dai D, Li D, Zhang C. Unraveling Nanomaterials in Biomimetic Mineralization of Dental Hard Tissue: Focusing on Advantages, Mechanisms, and Prospects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405763. [PMID: 39206945 DOI: 10.1002/advs.202405763] [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: 05/26/2024] [Revised: 07/31/2024] [Indexed: 09/04/2024]
Abstract
The demineralization of dental hard tissue imposes considerable health and economic burdens worldwide, but an optimal method that can repair both the chemical composition and complex structures has not been developed. The continuous development of nanotechnology has created new opportunities for the regeneration and repair of dental hard tissue. Increasingly studies have reported that nanomaterials (NMs) can induce and regulate the biomimetic mineralization of dental hard tissue, but few studies have examined how they are involved in the different stages, let alone the relevant mechanisms of action. Besides their nanoscale dimensions and excellent designability, NMs play a corresponding role in the function of the raw materials for mineralization, mineralized microenvironment, mineralization guidance, and the function of mineralized products. This review comprehensively summarizes the advantages of NMs and examines the specific mineralization mechanisms. Design strategies to promote regeneration and repair are summarized according to the application purpose of NMs in the oral cavity, and limitations and development directions in dental hard tissue remineralization are proposed. This review can provide a theoretical basis to understand the interaction between NMs and the remineralization of dental hard tissue, thereby optimizing design strategy, rational development, and clinical application of NMs in the field of remineralization.
Collapse
Affiliation(s)
- Danni Dai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Dan Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Chao Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
2
|
Ballester-Servera C, Alonso J, Taurón M, Rotllán N, Rodríguez C, Martínez-González J. Lysyl oxidase expression in smooth muscle cells determines the level of intima calcification in hypercholesterolemia-induced atherosclerosis. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:286-298. [PMID: 38402026 DOI: 10.1016/j.arteri.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/26/2024]
Abstract
INTRODUCTION Cardiovascular calcification is an important public health issue with an unmeet therapeutic need. We had previously shown that lysyl oxidase (LOX) activity critically influences vascular wall smooth muscle cells (VSMCs) and valvular interstitial cells (VICs) calcification by affecting extracellular matrix remodeling. We have delved into the participation of LOX in atherosclerosis and vascular calcification, as well as in the mineralization of the aortic valve. METHODS Immunohistochemical and expression studies were carried out in human atherosclerotic lesions and experimental models, valves from patients with aortic stenosis, VICs, and in a genetically modified mouse model that overexpresses LOX in CMLV (TgLOXCMLV). Hyperlipemia and atherosclerosis was induced in mice through the administration of adeno-associated viruses encoding a PCSK9 mutated form (AAV-PCSK9D374Y) combined with an atherogenic diet. RESULTS LOX expression is increased in the neointimal layer of atherosclerotic lesions from human coronary arteries and in VSMC-rich regions of atheromas developed both in the brachiocephalic artery of control (C57BL/6J) animals transduced with PCSK9D374Y and in the aortic root of ApoE-/- mice. In TgLOXCMLV mice, PCSK9D374Y transduction did not significantly alter the enhanced aortic expression of genes involved in matrix remodeling, inflammation, oxidative stress and osteoblastic differentiation. Likewise, LOX transgenesis did not alter the size or lipid content of atherosclerotic lesions in the aortic arch, brachiocephalic artery and aortic root, but exacerbated calcification. Among lysyl oxidase isoenzymes, LOX is the most expressed member of this family in highly calcified human valves, colocalizing with RUNX2 in VICs. The lower calcium deposition and decreased RUNX2 levels triggered by the overexpression of the nuclear receptor NOR-1 in VICs was associated with a reduction in LOX. CONCLUSIONS Our results show that LOX expression is increased in atherosclerotic lesions, and that overexpression of this enzyme in VSMC does not affect the size of the atheroma or its lipid content, but it does affect its degree of calcification. Further, these data suggest that the decrease in calcification driven by NOR-1 in VICs would involve a reduction in LOX. These evidences support the interest of LOX as a therapeutic target in cardiovascular calcification.
Collapse
MESH Headings
- Animals
- Humans
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Protein-Lysine 6-Oxidase/metabolism
- Protein-Lysine 6-Oxidase/genetics
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Disease Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Hypercholesterolemia/complications
- Mice, Inbred C57BL
- Aortic Valve Stenosis/pathology
- Aortic Valve Stenosis/metabolism
- Aortic Valve Stenosis/genetics
- Aortic Valve/pathology
- Aortic Valve/metabolism
- Male
- Proprotein Convertase 9/genetics
- Proprotein Convertase 9/metabolism
- Mice, Transgenic
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Diet, Atherogenic/adverse effects
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, España
| | - Noemí Rotllán
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud CarlosIII, Madrid, España
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España.
| |
Collapse
|
3
|
Yoon H, Park Y, Kwak JG, Lee J. Collagen structures of demineralized bone paper direct mineral metabolism. JBMR Plus 2024; 8:ziae080. [PMID: 38989259 PMCID: PMC11235081 DOI: 10.1093/jbmrpl/ziae080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Bone is a dynamic mineralized tissue that undergoes continuous turnover throughout life. While the general mechanism of bone mineral metabolism is documented, the role of underlying collagen structures in regulating osteoblastic mineral deposition and osteoclastic mineral resorption remains an active research area, partly due to the lack of biomaterial platforms supporting accurate and analytical investigation. The recently introduced osteoid-inspired demineralized bone paper (DBP), prepared by 20-μm thin sectioning of demineralized bovine compact bone, holds promise in addressing this challenge as it preserves the intrinsic bony collagen structure and retains semi-transparency. Here, we report on the impact of collagen structures on modulating osteoblast and osteoclast-driven bone mineral metabolism using vertical and transversal DBPs that exhibit a uniaxially aligned and a concentric ring collagen structure, respectively. Translucent DBP reveals these collagen structures and facilitates longitudinal tracking of mineral deposition and resorption under brightfield microscopy for at least 3 wk. Genetically labeled primary osteogenic cells allow fluorescent monitoring of these cellular processes. Osteoblasts adhere and proliferate following the underlying collagen structures of DBPs. Osteoblastic mineral deposition is significantly higher in vertical DBP than in transversal DBP. Spatiotemporal analysis reveals notably more osteoblast adhesion and faster mineral deposition in vascular regions than in bone regions. Subsequent osteoclastic resorption follows these mineralized collagen structures, directing distinct trench and pit-type resorption patterns. In vertical DBP, trench-type resorption occurs at an 80% frequency, whereas transversal DBP shows 35% trench-type and 65% pit-type resorption. Our studies substantiate the importance of collagen structures in regulating mineral metabolism by osteogenic cells. DBP is expected to serve as an enabling biomaterial platform for studying various aspects of cellular and extracellular bone remodeling biology.
Collapse
Affiliation(s)
- Hyejin Yoon
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, United States
| | - Jun-Goo Kwak
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Jungwoo Lee
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, United States
| |
Collapse
|
4
|
Wan Z, Bai X, Wang X, Guo X, Wang X, Zhai M, Fu Y, Liu Y, Zhang P, Zhang X, Yang R, Liu Y, Lv L, Zhou Y. Mgp High-Expressing MSCs Orchestrate the Osteoimmune Microenvironment of Collagen/Nanohydroxyapatite-Mediated Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308986. [PMID: 38588510 PMCID: PMC11187922 DOI: 10.1002/advs.202308986] [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: 11/23/2023] [Revised: 03/22/2024] [Indexed: 04/10/2024]
Abstract
Activating autologous stem cells after the implantation of biomaterials is an important process to initiate bone regeneration. Although several studies have demonstrated the mechanism of biomaterial-mediated bone regeneration, a comprehensive single-cell level transcriptomic map revealing the influence of biomaterials on regulating the temporal and spatial expression patterns of mesenchymal stem cells (MSCs) is still lacking. Herein, the osteoimmune microenvironment is depicted around the classical collagen/nanohydroxyapatite-based bone repair materials via combining analysis of single-cell RNA sequencing and spatial transcriptomics. A group of functional MSCs with high expression of matrix Gla protein (Mgp) is identified, which may serve as a pioneer subpopulation involved in bone repair. Remarkably, these Mgp high-expressing MSCs (MgphiMSCs) exhibit efficient osteogenic differentiation potential and orchestrate the osteoimmune microenvironment around implanted biomaterials, rewiring the polarization and osteoclastic differentiation of macrophages through the Mdk/Lrp1 ligand-receptor pair. The inhibition of Mdk/Lrp1 activates the pro-inflammatory programs of macrophages and osteoclastogenesis. Meanwhile, multiple immune-cell subsets also exhibit close crosstalk between MgphiMSCs via the secreted phosphoprotein 1 (SPP1) signaling pathway. These cellular profiles and interactions characterized in this study can broaden the understanding of the functional MSC subpopulations at the early stage of biomaterial-mediated bone regeneration and provide the basis for materials-designed strategies that target osteoimmune modulation.
Collapse
Affiliation(s)
- Zhuqing Wan
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiaoqiang Bai
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xin Wang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiaodong Guo
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xu Wang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Mo Zhai
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yang Fu
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yunsong Liu
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Ping Zhang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiao Zhang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Ruili Yang
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
- Department of OrthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
| | - Yan Liu
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
- Department of OrthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
| | - Longwei Lv
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yongsheng Zhou
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| |
Collapse
|
5
|
Ma Z, Zeng P, Zhai T, Zhao Y, Liang H. In Situ Mitochondrial Biomineralization for Drug-Free Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310218. [PMID: 38315577 DOI: 10.1002/adma.202310218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/13/2024] [Indexed: 02/07/2024]
Abstract
The common clinical chemotherapy often brings serious side effects to patients, mainly due to the off-target and leakage of toxic drugs. However, this is fatal for some specific clinical tumors, such as brain tumors and neuroma. This study performs a drug-free approach by encapsulating black phosphorus (BP) and calcium peroxide (CaO2) in liposomes with surface-modified triphenylphosphonium (BCLT) to develop mitochondria targeting calcification for cancer therapy without damaging normal cells. BCLT preferentially accumulates inside tumor mitochondria and then is activated by near-infrared (NIR) laser irradiation to produce abundant PO4 3- and Ca2+ to accelerate in situ mitochondrial mineralization, leading to mitochondrial dysfunction and cancer cell death. More importantly, both PO4 3- and Ca2+ are essential components of metabolism in the body, and random gradient diffusion or premature leakage does not cause damage to adjacent normal cells. This achievement promises to be an alternative to conventional chemotherapy in clinical practice for many specific tumor types.
Collapse
Affiliation(s)
- Zhaoyu Ma
- Department of Urology, Union Hospital, Tongji Medical College, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Pei Zeng
- Department of Urology, Union Hospital, Tongji Medical College, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Tianyou Zhai
- Department of Urology, Union Hospital, Tongji Medical College, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Huageng Liang
- Department of Urology, Union Hospital, Tongji Medical College, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
6
|
Liu Y, Lin S, Xu Z, Wu Y, Wang G, Yang G, Cao L, Chang H, Zhou M, Jiang X. High-Performance Hydrogel-Encapsulated Engineered Exosomes for Supporting Endoplasmic Reticulum Homeostasis and Boosting Diabetic Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309491. [PMID: 38380490 PMCID: PMC11077675 DOI: 10.1002/advs.202309491] [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: 12/06/2023] [Revised: 02/09/2024] [Indexed: 02/22/2024]
Abstract
The regeneration of bone defects in diabetic patients still faces challenges, as the intrinsic healing process is impaired by hyperglycemia. Inspired by the discovery that the endoplasmic reticulum (ER) is in a state of excessive stress and dysfunction under hyperglycemia, leading to osteogenic disorder, a novel engineered exosome is proposed to modulate ER homeostasis for restoring the function of mesenchymal stem cells (MSCs). The results indicate that the constructed engineered exosomes efficiently regulate ER homeostasis and dramatically facilitate the function of MSCs in the hyperglycemic niche. Additionally, the underlying therapeutic mechanism of exosomes is elucidated. The results reveal that exosomes can directly provide recipient cells with SHP2 for the activation of mitophagy and elimination of mtROS, which is the immediate cause of ER dysfunction. To maximize the therapeutic effect of engineered exosomes, a high-performance hydrogel with self-healing, bioadhesive, and exosome-conjugating properties is applied to encapsulate the engineered exosomes for in vivo application. In vivo, evaluation in diabetic bone defect repair models demonstrates that the engineered exosomes delivering hydrogel system intensively enhance osteogenesis. These findings provide crucial insight into the design and biological mechanism of ER homeostasis-based tissue-engineering strategies for diabetic bone regeneration.
Collapse
Affiliation(s)
- Yulan Liu
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Sihan Lin
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Zeqian Xu
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Yuqiong Wu
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Guifang Wang
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Guangzheng Yang
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Lingyan Cao
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Haishuang Chang
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200125China
| | - Mingliang Zhou
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| | - Xinquan Jiang
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyShanghai Engineering Research Center of Advanced Dental Technology and MaterialsShanghai200125China
| |
Collapse
|
7
|
Zhang DG, Pan YJ, Chen BQ, Lu XC, Xu QX, Wang P, Kankala RK, Jiang NN, Wang SB, Chen AZ. Protein-guided biomimetic nanomaterials: a versatile theranostic nanoplatform for biomedical applications. NANOSCALE 2024; 16:1633-1649. [PMID: 38168813 DOI: 10.1039/d3nr05495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Over the years, bioinspired mineralization-based approaches have been applied to synthesize multifunctional organic-inorganic nanocomposites. These nanocomposites can address the growing demands of modern biomedical applications. Proteins, serving as vital biological templates, play a pivotal role in the nucleation and growth processes of various organic-inorganic nanocomposites. Protein-mineralized nanomaterials (PMNMs) have attracted significant interest from researchers due to their facile and convenient preparation, strong physiological activity, stability, impressive biocompatibility, and biodegradability. Nevertheless, few comprehensive reviews have expounded on the progress of these nanomaterials in biomedicine. This article systematically reviews the principles and strategies for constructing nanomaterials using protein-directed biomineralization and biomimetic mineralization techniques. Subsequently, we focus on their recent applications in the biomedical field, encompassing areas such as bioimaging, as well as anti-tumor, anti-bacterial, and anti-inflammatory therapies. Furthermore, we discuss the challenges encountered in practical applications of these materials and explore their potential in future applications. This review aspired to catalyze the continued development of these bioinspired nanomaterials in drug development and clinical diagnosis, ultimately contributing to the fields of precision medicine and translational medicine.
Collapse
Affiliation(s)
- Da-Gui Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Yu-Jing Pan
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Biao-Qi Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Xiao-Chang Lu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Qin-Xi Xu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Pei Wang
- Jiangxi Provincial Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Ranjith Kumar Kankala
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ni-Na Jiang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Shi-Bin Wang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ai-Zheng Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
8
|
Surman F, Asadikorayem M, Weber P, Weber D, Zenobi-Wong M. Ionically annealed zwitterionic microgels for bioprinting of cartilaginous constructs. Biofabrication 2024; 16:025004. [PMID: 38176081 DOI: 10.1088/1758-5090/ad1b1f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/04/2024] [Indexed: 01/06/2024]
Abstract
Foreign body response (FBR) is a pervasive problem for biomaterials used in tissue engineering. Zwitterionic hydrogels have emerged as an effective solution to this problem, due to their ultra-low fouling properties, which enable them to effectively inhibit FBRin vivo. However, no versatile zwitterionic bioink that allows for high resolution extrusion bioprinting of tissue implants has thus far been reported. In this work, we introduce a simple, novel method for producing zwitterionic microgel bioink, using alginate methacrylate (AlgMA) as crosslinker and mechanical fragmentation as a microgel fabrication method. Photocrosslinked hydrogels made of zwitterionic carboxybetaine acrylamide (CBAA) and sulfobetaine methacrylate (SBMA) are mechanically fragmented through meshes with aperture diameters of 50 and 90µm to produce microgel bioink. The bioinks made with both microgel sizes showed excellent rheological properties and were used for high-resolution printing of objects with overhanging features without requiring a support structure or support bath. The AlgMA crosslinker has a dual role, allowing for both primary photocrosslinking of the bulk hydrogel as well as secondary ionic crosslinking of produced microgels, to quickly stabilize the printed construct in a calcium bath and to produce a microporous scaffold. Scaffolds showed ∼20% porosity, and they supported viability and chondrogenesis of encapsulated human primary chondrocytes. Finally, a meniscus model was bioprinted, to demonstrate the bioink's versatility at printing large, cell-laden constructs which are stable for furtherin vitroculture to promote cartilaginous tissue production. This easy and scalable strategy of producing zwitterionic microgel bioink for high resolution extrusion bioprinting allows for direct cell encapsulation in a microporous scaffold and has potential forin vivobiocompatibility due to the zwitterionic nature of the bioink.
Collapse
Affiliation(s)
- František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, 8032 Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
9
|
Ballester-Servera C, Alonso J, Cañes L, Vázquez-Sufuentes P, Puertas-Umbert L, Fernández-Celis A, Taurón M, Rodríguez-Sinovas A, López-Andrés N, Rodríguez C, Martínez-González J. Lysyl oxidase-dependent extracellular matrix crosslinking modulates calcification in atherosclerosis and aortic valve disease. Biomed Pharmacother 2023; 167:115469. [PMID: 37729730 DOI: 10.1016/j.biopha.2023.115469] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Extracellular matrix (ECM) is an active player in cardiovascular calcification (CVC), a major public health issue with an unmet need for effective therapies. Lysyl oxidase (LOX) conditions ECM biomechanical properties; thus, we hypothesized that LOX might impact on mineral deposition in calcific aortic valve disease (CAVD) and atherosclerosis. LOX was upregulated in calcified valves from two cohorts of CAVD patients. Strong LOX immunostaining was detected surrounding calcified foci in calcified human valves and atherosclerotic lesions colocalizing with RUNX2 on valvular interstitial cells (VICs) or vascular smooth muscle cells (VSMCs). Both LOX secretion and organized collagen deposition were enhanced in calcifying VICs exposed to osteogenic media. β-aminopropionitrile (BAPN), an inhibitor of LOX, attenuated collagen deposition and calcification. VICs seeded onto decellularized matrices from BAPN-treated VICs calcified less than cells cultured onto control scaffolds; instead, VICs exposed to conditioned media from cells over-expressing LOX or cultured onto LOX-crosslinked matrices calcified more. Atherosclerosis was induced in WT and transgenic mice that overexpress LOX in VSMC (TgLOXVSMC) by AAV-PCSK9D374Y injection and high-fat feeding. In atherosclerosis-challenged TgLOXVSMC mice both atherosclerosis burden and calcification assessed by near-infrared fluorescence (NIRF) imaging were higher than in WT mice. These animals also exhibited larger calcified areas in atherosclerotic lesions from aortic arches and brachiocephalic arteries. Moreover, LOX transgenesis exacerbated plaque inflammation, and increased VSMC cellularity, the rate of RUNX2-positive cells and both connective tissue content and collagen cross-linking. Our findings highlight the relevance of LOX in CVC and postulate this enzyme as a potential therapeutic target for CVC.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Paula Vázquez-Sufuentes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lídia Puertas-Umbert
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed, IdiSNA, UPNA, Hospital Universitario de Navarra (HUN), Pamplona, Spain
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Diseases Research Group, Vall d'Hebron University Hospital and Research Institute, Barcelona, Spain
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed, IdiSNA, UPNA, Hospital Universitario de Navarra (HUN), Pamplona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain.
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
| |
Collapse
|
10
|
Zhu J, Li Z, Zou Y, Lu G, Ronca A, D’Amora U, Liang J, Fan Y, Zhang X, Sun Y. Advanced application of collagen-based biomaterials in tissue repair and restoration. JOURNAL OF LEATHER SCIENCE AND ENGINEERING 2022. [DOI: 10.1186/s42825-022-00102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AbstractIn tissue engineering, bioactive materials play an important role, providing structural support, cell regulation and establishing a suitable microenvironment to promote tissue regeneration. As the main component of extracellular matrix, collagen is an important natural bioactive material and it has been widely used in scientific research and clinical applications. Collagen is available from a wide range of animal origin, it can be produced by synthesis or through recombinant protein production systems. The use of pure collagen has inherent disadvantages in terms of physico-chemical properties. For this reason, a processed collagen in different ways can better match the specific requirements as biomaterial for tissue repair. Here, collagen may be used in bone/cartilage regeneration, skin regeneration, cardiovascular repair and other fields, by following different processing methods, including cross-linked collagen, complex, structured collagen, mineralized collagen, carrier and other forms, promoting the development of tissue engineering. This review summarizes a wide range of applications of collagen-based biomaterials and their recent progress in several tissue regeneration fields. Furthermore, the application prospect of bioactive materials based on collagen was outlooked, aiming at inspiring more new progress and advancements in tissue engineering research.
Graphical Abstract
Collapse
|
11
|
Chang R, Liu Y, Zhang Y, Zhang S, Han B, Chen F, Chen Y. Phosphorylated and Phosphonated Low-Complexity Protein Segments for Biomimetic Mineralization and Repair of Tooth Enamel. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103829. [PMID: 34978158 PMCID: PMC8867149 DOI: 10.1002/advs.202103829] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/18/2021] [Indexed: 05/03/2023]
Abstract
Biomimetic mineralization based on self-assembly has made great progress, providing bottom-up strategies for the construction of new organic-inorganic hybrid materials applied in the treatment of hard tissue defects. Herein, inspired by the cooperative effects of key components in biomineralization microenvironments, a new type of biocompatible peptide scaffold based on flexibly self-assembling low-complexity protein segments (LCPSs) containing phosphate or phosphonate groups is developed. These LCPSs can retard the transformation of amorphous calcium phosphate into hydroxyapatite (HAP), leading to merged mineralization structures. Moreover, the application of phosphonated LCPS over phosphorylated LCPS can prevent hydrolysis by phosphatases that are enriched in extracellular mineralization microenvironments. After being coated on the etched tooth enamel, these LCPSs facilitate the growth of HAP to generate new enamel layers comparable to the natural layers and mitigate the adhesion of Streptococcus mutans. In addition, they can effectively stimulate the differentiation pathways of osteoblasts. These results shed light on the potential biomedical applications of two LCPSs in hard tissue repair.
Collapse
Affiliation(s)
- Rong Chang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
| | - Yang‐Jia Liu
- Central LaboratoryPeking University Hospital of StomatologyBeijing100081China
| | - Yun‐Lai Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
| | - Shi‐Ying Zhang
- Central LaboratoryPeking University Hospital of StomatologyBeijing100081China
| | - Bei‐Bei Han
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
| | - Feng Chen
- Central LaboratoryPeking University Hospital of StomatologyBeijing100081China
| | - Yong‐Xiang Chen
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
| |
Collapse
|
12
|
Chen Y, Wang Y, Luo SC, Zheng X, Kankala RK, Wang SB, Chen AZ. Advances in Engineered Three-Dimensional (3D) Body Articulation Unit Models. Drug Des Devel Ther 2022; 16:213-235. [PMID: 35087267 PMCID: PMC8789231 DOI: 10.2147/dddt.s344036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/24/2021] [Indexed: 12/19/2022] Open
Abstract
Indeed, the body articulation units, commonly referred to as body joints, play significant roles in the musculoskeletal system, enabling body flexibility. Nevertheless, these articulation units suffer from several pathological conditions, such as osteoarthritis (OA), rheumatoid arthritis (RA), ankylosing spondylitis, gout, and psoriatic arthritis. There exist several treatment modalities based on the utilization of anti-inflammatory and analgesic drugs, which can reduce or control the pathophysiological symptoms. Despite the success, these treatment modalities suffer from major shortcomings of enormous cost and poor recovery, limiting their applicability and requiring promising strategies. To address these limitations, several engineering strategies have been emerged as promising solutions in fabricating the body articulation as unit models towards local articulation repair for tissue regeneration and high-throughput screening for drug development. In this article, we present challenges related to the selection of biomaterials (natural and synthetic sources), construction of 3D articulation models (scaffold-free, scaffold-based, and organ-on-a-chip), architectural designs (microfluidics, bioprinting, electrospinning, and biomineralization), and the type of culture conditions (growth factors and active peptides). Then, we emphasize the applicability of these articulation units for emerging biomedical applications of drug screening and tissue repair/regeneration. In conclusion, we put forward the challenges and difficulties for the further clinical application of the in vitro 3D articulation unit models in terms of the long-term high activity of the models.
Collapse
Affiliation(s)
- Ying Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People’s Republic of China
| | - Ying Wang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, Guangdong, People’s Republic of China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510080, Guangdong, People’s Republic of China
| | - Sheng-Chang Luo
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People’s Republic of China
| | - Xiang Zheng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People’s Republic of China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People’s Republic of China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People’s Republic of China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People’s Republic of China
| |
Collapse
|
13
|
Enzymatic Approach in Calcium Phosphate Biomineralization: A Contribution to Reconcile the Physicochemical with the Physiological View. Int J Mol Sci 2021; 22:ijms222312957. [PMID: 34884758 PMCID: PMC8657759 DOI: 10.3390/ijms222312957] [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: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/22/2023] Open
Abstract
Biomineralization is the process by which organisms produce hard inorganic matter from soft tissues with outstanding control of mineral deposition in time and space. For this purpose, organisms deploy a sophisticated "toolkit" that has resulted in significant evolutionary innovations, for which calcium phosphate (CaP) is the biomineral selected for the skeleton of vertebrates. While CaP mineral formation in aqueous media can be investigated by studying thermodynamics and kinetics of phase transitions in supersaturated solutions, biogenic mineralization requires coping with the inherent complexity of biological systems. This mainly includes compartmentalization and homeostatic processes used by organisms to regulate key physiological factors, including temperature, pH and ion concentration. A detailed analysis of the literature shows the emergence of two main views describing the mechanism of CaP biomineralization. The first one, more dedicated to the study of in vivo systems and supported by researchers in physiology, often involves matrix vesicles (MVs). The second one, more investigated by the physicochemistry community, involves collagen intrafibrillar mineralization particularly through in vitro acellular models. Herein, we show that there is an obvious need in the biological systems to control both where and when the mineral forms through an in-depth survey of the mechanism of CaP mineralization. This necessity could gather both communities of physiologists and physicochemists under a common interest for an enzymatic approach to better describe CaP biomineralization. Both homogeneous and heterogeneous enzymatic catalyses are conceivable for these systems, and a few preliminary promising results on CaP mineralization for both types of enzymatic catalysis are reported in this work. Through them, we aim to describe the relevance of our point of view and the likely findings that could be obtained when adding an enzymatic approach to the already rich and creative research field dealing with CaP mineralization. This complementary approach could lead to a better understanding of the biomineralization mechanism and inspire the biomimetic design of new materials.
Collapse
|