1
|
Giovanetti K, Tuma RB, Sant'Ana Pegorin Brasil G, Miranda MCR, Borges FA, Tanaka JL, Burd BS, Cortellazzi KL, Guerra NB, Mussagy CU, Floriano JF, Dos Santos LS, de Melo Silva W, Cao W, Herculano RD, Caria PHF. β-Tricalcium phosphate incorporated natural rubber latex membranes for calvarial bone defects: Physicochemical, in vitro and in vivo assessment. Int J Biol Macromol 2024; 282:137328. [PMID: 39515716 DOI: 10.1016/j.ijbiomac.2024.137328] [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: 08/06/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Natural rubber latex membrane (NRL) is a biocompatible macromolecule that stimulates angiogenesis and promotes bone repair. Similarly, β-tricalcium phosphate (β-TCP) is an osteoconductive and osteoinductive bioceramic widely used as a bone substitute. Here, we investigated the combined use of these biomaterials in the guided bone regeneration process for calvarial defects in rats. Physicochemical characterization was performed to evaluate the interaction between β-TCP and NRL. Membrane toxicity was assessed using MC3T3 osteoblasts culture and in vivo assays with Caenorhabditis elegans. Lastly, NRL membranes, NRL incorporated with β-TCP membranes (NRL-β-TCP), and a periosteum-only (control group) were tested on rodents. MC3T3 cells adhered to membranes, preserving their morphology and intercellular connections. NRL-β-TCP membranes demonstrated no toxicity in larvae, which maintained their sinusoidal wave shape. Tests results on rodents revealed statistical difference between the groups at 60 days post-operation. NRL-β-TCP (56.1 ± 14.0 %) had an average 1.48-fold higher than the control group (38.0 ± 9.1 %), with tissue production and bone remodeling. Our qualitative histological analyses revealed that membranes significantly accelerated bone formation without any signs of inflammatory reactions. We conclude that NRL-β-TCP has potential to be used for flat bone regeneration, with osteoconductive properties, being a cheap, biocompatible, and effective occlusive barrier.
Collapse
Affiliation(s)
- Karina Giovanetti
- Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Rafael Brull Tuma
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Giovana Sant'Ana Pegorin Brasil
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Matheus Carlos Romeiro Miranda
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo (USP), Diadema, SP, Brazil
| | - Felipe Azevedo Borges
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Jean Lucas Tanaka
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Betina Sayeg Burd
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Karine Laura Cortellazzi
- Departmentof Social Dentistry, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Nayrim Brizuela Guerra
- School of Science, São Paulo State University (UNESP), Bauru, São Paulo 17033-360, Brazil
| | - Cassamo Ussemane Mussagy
- Escuela de Agronomía, Facultad de Ciencias Agronómicas y de los Alimentos, Pontificia Universidad Católica de Valparaíso, Chile
| | - Juliana Ferreira Floriano
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil; National Heart and Lung Institute, Imperial College London, London, UK
| | - Lindomar Soares Dos Santos
- Faculty of Philosophy, Sciences and Languages of Ribeirão Preto, University of São Paulo (USP), 3900 Bandeirantes Avenue, Ribeirão Preto, SP 14.040-901, Brazil
| | - William de Melo Silva
- Institute of Biotechnology, São Paulo State University (UNESP), University Avenue 3780, 18610-034, Botucatu, Brazil
| | - Wei Cao
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Rondinelli Donizetti Herculano
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA.
| | | |
Collapse
|
2
|
Jiang X, Jian Y, Zhang Y, Zhong J, Li Q, Wang X, Jia X, Wu X, Zhao K, Yao Y. Dual-Mode Release of IL-4 and TCP from a PGA-SF Core-Shell Electrospinning Scaffold for Enhanced Bone Regeneration through Synergistic Immunoregulation and Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58148-58167. [PMID: 39279657 DOI: 10.1021/acsami.4c08996] [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: 09/18/2024]
Abstract
The successful filling of bone defects remains challenging due to the incongruity between bone graft materials and the dynamic process of bone healing. Developing multifunctional materials matching the dynamic process of bone healing offers a viable solution to the current dilemma. Lines of evidence have shown that engineering osteoimmunomodulatory biomaterials can modulate the function of immune cells and thus promote bone regeneration. Herein, we utilized silk fibroin (SF) and polyglycolic acid (PGA) to create a PGA-SF core-shell fibrous scaffold, incorporating interleukin-4 (IL-4) and tricalcium phosphate (TCP) as a codelivery system (PGA/TCP-SF/IL-4), aiming to achieve an initial rapid release of IL-4 and sustained release of TCP. The PGA/TCP-SF/IL-4 scaffold mimicked the native bone structure and showed superior tenacity in the wetting regime. In vitro studies demonstrated that the PGA/TCP-SF/IL-4 scaffold significantly reduced the inflammatory response by upregulating the M2 macrophages, created a favorable microenvironment for osteogenesis, and facilitated osteogenic differentiation and mineralization. Implantation of the PGA/TCP-SF/IL-4 scaffold into the rat skull defect model notably increased the formation of new bones. IL-4 and TCP acted synergistically in attenuating inflammation and enhancing osteogenic differentiation. Overall, this multifunctional scaffold comprehensively considers the various demands in the bone defect region, which might have a significant potential for application in bone reconstruction.
Collapse
Affiliation(s)
- Xiao Jiang
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Yutao Jian
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Yuan Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Juan Zhong
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Qiulan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Xiaodong Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Xiaoshi Jia
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Xiangnan Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Ke Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| | - Yitong Yao
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510275, China
| |
Collapse
|
3
|
Mills EG, Abbara A, Dhillo WS, Comninos AN. Interactions between kisspeptin and bone: Cellular mechanisms, clinical evidence, and future potential. Ann N Y Acad Sci 2024; 1540:47-60. [PMID: 39269749 DOI: 10.1111/nyas.15213] [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] [Indexed: 09/15/2024]
Abstract
The neuropeptide kisspeptin and its cognate receptor have been extensively studied in reproductive physiology, with diverse and well-established functions, including as an upstream regulator of pubertal onset, reproductive hormone secretion, and sexual behavior. Besides classical reproduction, both kisspeptin and its receptor are extensively expressed in bone-resorbing osteoclasts and bone-forming osteoblasts, which putatively permits direct bone effects. Accordingly, this sets the scene for recent compelling findings derived from in vitro experiments through to in vivo and clinical studies revealing prominent regulatory interactions for kisspeptin signaling in bone metabolism, as well as certain oncological aspects of bone metabolism. Herein, we comprehensively examine the experimental evidence obtained to date supporting the interaction between kisspeptin and bone. A comprehensive understanding of this emerging facet of kisspeptin biology is fundamental to exploiting the future therapeutic potential of kisspeptin-based medicines as a novel strategy for treating bone-related disorders.
Collapse
Affiliation(s)
- Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
- Endocrine Bone Unit, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
4
|
Zhong J, Huang W, Ahmad R, Chen J, Wu C, Hu J, Zheng K, Swain MV, Li Q. A Soft-Tissue Driven Bone Remodeling Algorithm for Mandibular Residual Ridge Resorption Based on Patient CT Image Data. Adv Healthc Mater 2024; 13:e2400091. [PMID: 38722148 DOI: 10.1002/adhm.202400091] [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: 01/09/2024] [Revised: 05/04/2024] [Indexed: 09/03/2024]
Abstract
The role of the biomechanical stimulation generated from soft tissue has not been well quantified or separated from the self-regulated hard tissue remodeling governed by Wolff's Law. Prosthodontic overdentures, commonly used to restore masticatory functions, can cause localized ischemia and inflammation as they often compress patients' oral mucosa and impede local circulation. This biomechanical stimulus in mucosa is found to accelerate the self-regulated residual ridge resorption (RRR), posing ongoing clinical challenges. Based on the dedicated long-term clinical datasets, this work develops an in-silico framework with a combination of techniques, including advanced image post-processing, patient-specific finite element models and unsupervised machine learning Self-Organizing map algorithm, to identify the soft tissue induced RRR and quantitatively elucidate the governing relationship between the RRR and hydrostatic pressure in mucosa. The proposed governing equation has not only enabled a predictive simulation for RRR as showcased in this study, providing a biomechanical basis for optimizing prosthodontic treatments, but also extended the understanding of the mechanobiological responses in the soft-hard tissue interfaces and the role in bone remodeling.
Collapse
Affiliation(s)
- Jingxiao Zhong
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Wenwei Huang
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Rohana Ahmad
- Faculty of Dentistry and Integrative Pharmacogenomics Institute, Universiti Teknologi MARA, Selangor, 40450, Malaysia
| | - Junning Chen
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QJ, UK
| | - Chi Wu
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Jingrui Hu
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QJ, UK
| | - Keke Zheng
- Institute for Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Michael V Swain
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
5
|
Waletzko-Hellwig J, Sass JO, Bader R, Frerich B, Dau M. Evaluation of Integrity of Allogeneic Bone Processed with High Hydrostatic Pressure: A Pilot Animal Study. Biomater Res 2024; 28:0067. [PMID: 39148817 PMCID: PMC11325089 DOI: 10.34133/bmr.0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 07/29/2024] [Indexed: 08/17/2024] Open
Abstract
Processing of bone allografts with strong acids and γ-sterilization results in decreased biomechanical properties and reduction in osteogenecity and osteoconductivity. High hydrostatic pressure (HHP) treatment could be a gentle alternative to processing techniques usually applied. HHP is known to induce devitalization of cancellous bone while preserving biomechanical stability and molecules that induce cell differentiation. Here, a specific HHP protocol for devitalization of cancellous bone was applied to rabbit femoral bone. Allogeneic bone cylinders were subsequently implanted into a defect in the lateral condyles of rabbit femora and were compared to autologous bone grafts. Analysis of bone integration 4 and 12 weeks postoperatively revealed no differences between autografts and HHP-treated allografts regarding the expression of genes characteristic for bone remodeling, showing expression niveous comparable to original bone cylinder. Furthermore, biomechanical properties were evaluated 12 weeks postoperatively. Autografts and HHP-treated allografts both showed a yield strength ranging between 2 and 2.5 MPa and an average bone mass density of 250 mg/cm2. Furthermore, histological analysis of the region of interest revealed a rate of 5 to 10% BPM-2 and approximately 40% osteocalcin-positive staining, with no marked differences between allografts and autografts demonstrating comparable matrix deposition in the graft region. A suitable graft integrity was pointed out by μCT imaging in both groups, supporting the biomechanical data. In summary, the integrity of HHP-treated cancellous bone allografts showed similar results to untreated autografts. Hence, HHP treatment may represent a gentle and effective alternative to existing processing techniques for bone allografts.
Collapse
Affiliation(s)
- Janine Waletzko-Hellwig
- Department of Oral, Maxillofacial and Plastic Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, 18057 Rostock, Germany
| | - Jan-Oliver Sass
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, 18057 Rostock, Germany
| | - Rainer Bader
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, 18057 Rostock, Germany
| | - Bernhard Frerich
- Department of Oral, Maxillofacial and Plastic Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michael Dau
- Department of Oral, Maxillofacial and Plastic Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
6
|
Luo S, Zhang C, Xiong W, Song Y, Wang Q, Zhang H, Guo S, Yang S, Liu H. Advances in electroactive biomaterials: Through the lens of electrical stimulation promoting bone regeneration strategy. J Orthop Translat 2024; 47:191-206. [PMID: 39040489 PMCID: PMC11261049 DOI: 10.1016/j.jot.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/16/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
The regenerative capacity of bone is indispensable for growth, given that accidental injury is almost inevitable. Bone regenerative capacity is relevant for the aging population globally and for the repair of large bone defects after osteotomy (e.g., following removal of malignant bone tumours). Among the many therapeutic modalities proposed to bone regeneration, electrical stimulation has attracted significant attention owing to its economic convenience and exceptional curative effects, and various electroactive biomaterials have emerged. This review summarizes the current knowledge and progress regarding electrical stimulation strategies for improving bone repair. Such strategies range from traditional methods of delivering electrical stimulation via electroconductive materials using external power sources to self-powered biomaterials, such as piezoelectric materials and nanogenerators. Electrical stimulation and osteogenesis are related via bone piezoelectricity. This review examines cell behaviour and the potential mechanisms of electrostimulation via electroactive biomaterials in bone healing, aiming to provide new insights regarding the mechanisms of bone regeneration using electroactive biomaterials. The translational potential of this article This review examines the roles of electroactive biomaterials in rehabilitating the electrical microenvironment to facilitate bone regeneration, addressing current progress in electrical biomaterials and the mechanisms whereby electrical cues mediate bone regeneration. Interactions between osteogenesis-related cells and electroactive biomaterials are summarized, leading to proposals regarding the use of electrical stimulation-based therapies to accelerate bone healing.
Collapse
Affiliation(s)
- Songyang Luo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, 110001, China
| | - Chengshuo Zhang
- Hepatobiliary Surgery Department, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Hospital of Shihezi Medical University, Shihezi, 832000, China
| | - Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, 110001, China
| | - Hangzhou Zhang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang Sports Medicine Clinical Medical Research Center, Shenyang, 110001, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, 110001, China
| | - Huanye Liu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, 110001, China
| |
Collapse
|
7
|
Vasireddi N, Neitzke CC, Chandi SK, Cororaton AD, Driscoll DA, Sculco PK, Chalmers BP, Gausden EB. Early Periprosthetic Femur Fractures After Primary Cementless Total Hip Arthroplasty: High Risk of Periprosthetic Joint Infection and Subsequent Reoperation. J Arthroplasty 2024; 39:1083-1087.e1. [PMID: 37871864 DOI: 10.1016/j.arth.2023.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/06/2023] [Accepted: 10/14/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Periprosthetic femur fracture (PFF) following total hip arthroplasty (THA) is a leading cause of early reoperation. The objective of this study was to compare rates of periprosthetic joint infection (PJI) and reoperation following PFFs occurring early postoperatively to those that occurred late. METHODS We retrospectively identified 173 consecutive surgically managed PFFs following primary THA. Cases were categorized as "early" if they occurred within 90 days of THA (n = 117) or "late" if they occurred following the initial 90 days (n = 56). Mean age at time of PFF was 68 years (range, 26 to 96) and 60% were women. Mean body mass index was 29 (range, 16 to 52). Mean follow-up was 2 years (range, 0 to 13). Kaplan-Meier survival analysis estimated cumulative incidences of PJI and reoperation. RESULTS Early PFFs had higher 2-year cumulative incidence of PJI (11% versus 0%, P < .001) and reoperation (24% versus 13%, P = .110). Following early PFF, 27 patients required reoperation (ie, 13 for PJI, 5 for instability, 2 for re-fracture, 2 for painful hardware, 2 for non-union, 1 for adverse local tissue reaction, 1 for aseptic loosening, and 1 for leg-length discrepancy). Following late PFF, 5 patients required reoperation (ie, 3 for instability, 1 for re-fracture, and 1 for non-union). CONCLUSIONS There are greater incidences of PJIs and overall reoperations following early PFFs compared to late PFFs after THA. In addition to focusing efforts on prevention of early PFFs, surgeons should consider antiseptic interventions to mitigate the increased risk of PJI after treatment of early PFF.
Collapse
Affiliation(s)
- Nikhil Vasireddi
- Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Colin C Neitzke
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Sonia K Chandi
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Agnes D Cororaton
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Daniel A Driscoll
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Peter K Sculco
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Brian P Chalmers
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Elizabeth B Gausden
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| |
Collapse
|
8
|
Meisterhans M, Dimitriou D, Fasser MR, Hoch A, Jud L, Zingg PO. Influence of offset on osseointegration in cementless total hip arthroplasty: A finite element study. J Orthop Res 2024. [PMID: 38376065 DOI: 10.1002/jor.25808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/21/2024]
Abstract
Early aseptic loosening is caused by deficient osteointegration of the femoral stem due to increased micromotions and represents a common mode of failure in uncemented total hip arthroplasty (THA). This study hypothesized that a higher femoral offset, a smaller stem size and obesity increase femoral micromotion, potentially resulting in early aseptic loosening. A finite element analysis was conducted based on computed tomography segmented model of four patients who received a THA with a triple-tapered straight stem (Size 1, 3, 6). The influence of femoral stem offset (short neck, standard, lateral), head length (S to XXL), femoral anteversion and obesity during daily activities of fast walking and stair climbing was analyzed. The micromotions for the femoral stem zones were compared to a threshold representing a value above which only partial osseointegration is expected. The minimum femoral offset configuration compared to the maximum offset configuration (short neck stem, S head vs. lateral stem, XXL head) leads to a relative mean micromotion increase of 24% for the upper stem zone. Increasing the body weight (body mass index 30-35 kg/m2 ) increases the micromotion by 20% for all stem zones. The obese population recorded threshold-exceeding micromotions for stem sizes 1 and 3 for all offset configurations during stair climbing. Higher femoral offset, a smaller stem size, and higher loading due to obesity lead to an increase in micromotion between the prosthesis and proximal femur and represent a risk configuration for impaired osseointegration of a triple-tapered straight stem, especially when these three factors are present simultaneously.
Collapse
Affiliation(s)
- Michel Meisterhans
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Dimitris Dimitriou
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Marie-Rosa Fasser
- Institute of Biomechanics, Balgrist Campus, ETH Zurich, Zurich, Switzerland
- Spine Biomechanics, Department of Orthopedic Surgery, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Armando Hoch
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Lukas Jud
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Patrick O Zingg
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Wang W, Zheng X, Wang H, Zuo B, Chen S, Li J. Mechanical Unloading Promotes Osteoclastic Differentiation and Bone Resorption by Modulating the MSC Secretome to Favor Inflammation. Cell Transplant 2024; 33:9636897241236584. [PMID: 38501500 PMCID: PMC10953070 DOI: 10.1177/09636897241236584] [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: 10/22/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
Aging, space flight, and prolonged bed rest have all been linked to bone loss, and no effective treatments are clinically available at present. Here, with the rodent hindlimb unloading (HU) model, we report that the bone marrow (BM) microenvironment was significantly altered, with an increased number of myeloid cells and elevated inflammatory cytokines. In such inflammatory BM, the osteoclast-mediated bone resorption was greatly enhanced, leading to a shifted bone remodeling balance that ultimately ends up with disuse-induced osteoporosis. Using Piezo1 conditional knockout (KO) mice (Piezo1fl/fl;LepRCre), we proved that lack of mechanical stimuli on LepR+ mesenchymal stem cells (MSCs) is the main reason for the pathological BM inflammation. Mechanically, the secretome of MSCs was regulated by mechanical stimuli. Inadequate mechanical load leads to increased production of inflammatory cytokines, such as interleukin (IL)-1α, IL-6, macrophage colony-stimulating factor 1 (M-CSF-1), and so on, which promotes monocyte proliferation and osteoclastic differentiation. Interestingly, transplantation of 10% cyclic mechanical stretch (CMS)-treated MSCs into HU animals significantly alleviated the BM microenvironment and rebalanced bone remodeling. In summary, our research revealed a new mechanism underlying mechanical unloading-induced bone loss and suggested a novel stem cell-based therapy to potentially prevent disuse-induced osteoporosis.
Collapse
Affiliation(s)
- Wanyuji Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Xueling Zheng
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Hehe Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Bin Zuo
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sisi Chen
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Li
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
10
|
de Carvalho Moreira LMC, de Sousa Silva ABA, de Araújo Medeiros K, Oshiro Júnior JA, da Silva DTC, de Lima Damasceno BPG. Effectiveness In Vivo and In Vitro of Polymeric Nanoparticles as a Drug Release System in the Treatment of Leishmaniasis. Curr Med Chem 2024; 31:286-307. [PMID: 36683370 DOI: 10.2174/0929867330666230120163543] [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: 06/20/2022] [Revised: 10/13/2022] [Accepted: 11/08/2022] [Indexed: 01/24/2023]
Abstract
Leishmaniasis is a neglected disease caused by the parasite of the genus Leishmania. Current treatment regimens are obsolete and cause several side effects, promoting poor patient compliance, in addition to the vast majority already having the potential for resistance. Therefore, polymeric nanoparticles emerge as one of the viable alternatives to overcome existing limitations, through passive or active vectorization. This review aims to summarize the latest studies of polymeric nanoparticles as an alternative treatment for leishmaniasis. In the first section, the main pharmacokinetic and pharmacodynamic challenges of current drugs are reported. The second section details how nanoparticles with and without functionalization are efficient in the treatment of leishmaniasis, discussing the characteristics of the polymer in the formulation. In this way, polymeric nanoparticles can improve the physicochemical properties of leishmanicidal drugs, improving solubility and stability, as well as improve the release of these drugs, directly or indirectly reaching monocytes/macrophages. 64.28% drugs were focused on the treatment of visceral leishmaniasis, and 28.57% on cutaneous leishmaniasis. The most chosen polymers in the literature are chitosan (35.71%) and PLGA (35.71%), the others represented 14.30% drugs, with all able to manage the drug release and increase the in vitro and/or in vivo efficacy of the original molecule. However, there are several barriers for these nanoformulations to cross laboratory research and is necessary more in-depth studies about the metabolites and degradation pathways of the polymers used in the formulations and plasma proteomics studies.
Collapse
Affiliation(s)
- Lívia Maria Coelho de Carvalho Moreira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | | | - Kaline de Araújo Medeiros
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | - João Augusto Oshiro Júnior
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | - Dayanne Tomaz Casimiro da Silva
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | - Bolívar Ponciano Goulart de Lima Damasceno
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| |
Collapse
|
11
|
Rojas-Rojas L, Tozzi G, Guillén-Girón T. A Comprehensive Mechanical Characterization of Subject-Specific 3D Printed Scaffolds Mimicking Trabecular Bone Architecture Biomechanics. Life (Basel) 2023; 13:2141. [PMID: 38004281 PMCID: PMC10672154 DOI: 10.3390/life13112141] [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: 09/27/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
This study presents a polymeric scaffold designed and manufactured to mimic the structure and mechanical compressive characteristics of trabecular bone. The morphological parameters and mechanical behavior of the scaffold were studied and compared with trabecular bone from bovine iliac crest. Its mechanical properties, such as modulus of elasticity and yield strength, were studied under a three-step monotonic compressive test. Results showed that the elastic modulus of the scaffold was 329 MPa, and the one for trabecular bone reached 336 MPa. A stepwise dynamic compressive test was used to assess the behavior of samples under various loading regimes. With microcomputed tomography (µCT), a three-dimensional reconstruction of the samples was obtained, and their porosity was estimated as 80% for the polymeric scaffold and 88% for trabecular bone. The full-field strain distribution of the samples was measured using in situ µCT mechanics and digital volume correlation (DVC). This provided information on the local microdeformation mechanism of the scaffolds when compared to that of the tissue. The comprehensive results illustrate the potential of the fabricated scaffolds as biomechanical templates for in vitro studies. Furthermore, there is potential for extending this structure and fabrication methodology to incorporate suitable biocompatible materials for both in vitro and in vivo clinical applications.
Collapse
Affiliation(s)
- Laura Rojas-Rojas
- Materials Science and Engineering School, Tecnológico de Costa Rica, Cartago 30109, Costa Rica;
| | - Gianluca Tozzi
- School of Engineering, University of Greenwich, Chatham ME4 4TB, UK;
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth PO1 3DJ, UK
| | - Teodolito Guillén-Girón
- Materials Science and Engineering School, Tecnológico de Costa Rica, Cartago 30109, Costa Rica;
| |
Collapse
|
12
|
Han X, Saiding Q, Cai X, Xiao Y, Wang P, Cai Z, Gong X, Gong W, Zhang X, Cui W. Intelligent Vascularized 3D/4D/5D/6D-Printed Tissue Scaffolds. NANO-MICRO LETTERS 2023; 15:239. [PMID: 37907770 PMCID: PMC10618155 DOI: 10.1007/s40820-023-01187-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023]
Abstract
Blood vessels are essential for nutrient and oxygen delivery and waste removal. Scaffold-repairing materials with functional vascular networks are widely used in bone tissue engineering. Additive manufacturing is a manufacturing technology that creates three-dimensional solids by stacking substances layer by layer, mainly including but not limited to 3D printing, but also 4D printing, 5D printing and 6D printing. It can be effectively combined with vascularization to meet the needs of vascularized tissue scaffolds by precisely tuning the mechanical structure and biological properties of smart vascular scaffolds. Herein, the development of neovascularization to vascularization to bone tissue engineering is systematically discussed in terms of the importance of vascularization to the tissue. Additionally, the research progress and future prospects of vascularized 3D printed scaffold materials are highlighted and presented in four categories: functional vascularized 3D printed scaffolds, cell-based vascularized 3D printed scaffolds, vascularized 3D printed scaffolds loaded with specific carriers and bionic vascularized 3D printed scaffolds. Finally, a brief review of vascularized additive manufacturing-tissue scaffolds in related tissues such as the vascular tissue engineering, cardiovascular system, skeletal muscle, soft tissue and a discussion of the challenges and development efforts leading to significant advances in intelligent vascularized tissue regeneration is presented.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, People's Republic of China
| | - Yi Xiao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xuan Gong
- University of Texas Southwestern Medical Center, Dallas, TX, 75390-9096, USA
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China.
| | - Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
13
|
Zhong J, Shibata Y, Wu C, Watanabe C, Chen J, Zheng K, Hu J, Swain MV, Li Q. Functional non-uniformity of periodontal ligaments tunes mechanobiological stimuli across soft- and hard-tissue interfaces. Acta Biomater 2023; 170:240-249. [PMID: 37634832 DOI: 10.1016/j.actbio.2023.08.047] [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: 04/10/2023] [Revised: 08/09/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
The bone-periodontal ligament-tooth (BPT) complex is a unique mechanosensing soft-/hard-tissue interface, which governs the most rapid bony homeostasis in the body responding to external loadings. While the correlation between such loading and alveolar bone remodelling has been widely recognised, it has remained challenging to investigate the transmitted mechanobiological stimuli across such embedded soft-/hard-tissue interfaces of the BPT complex. Here, we propose a framework combining three distinct bioengineering techniques (i, ii, and iii below) to elucidate the innate functional non-uniformity of the PDL in tuning mechanical stimuli to the surrounding alveolar bone. The biphasic PDL mechanical properties measured via nanoindentation, namely the elastic moduli of fibres and ground substance at the sub-tissue level (i), were used as the input parameters in an image-based constitutive modelling framework for finite element simulation (ii). In tandem with U-net deep learning, the Gaussian mixture method enabled the comparison of 5195 possible pseudo-microstructures versus the innate non-uniformity of the PDL (iii). We found that the balance between hydrostatic pressure in PDL and the strain energy in the alveolar bone was maintained within a specific physiological range. The innate PDL microstructure ensures the transduction of favourable mechanobiological stimuli, thereby governing alveolar bone homeostasis. Our outcomes expand current knowledge of the PDL's mechanobiological roles and the proposed framework can be adopted to a broad range of similar soft-/hard- tissue interfaces, which may impact future tissue engineering, regenerative medicine, and evaluating therapeutic strategies. STATEMENT OF SIGNIFICANCE: A combination of cutting-edge technologies, including dynamic nanomechanical testing, high-resolution image-based modelling and machine learning facilitated computing, was used to elucidate the association between the microstructural non-uniformity and biomechanical competence of periodontal ligaments (PDLs). The innate PDL fibre network regulates mechanobiological stimuli, which govern alveolar bone remodelling, in different tissues across the bone-PDL-tooth (BPT) interfaces. These mechanobiological stimuli within the BPT are tuned within a physiological range by the non-uniform microstructure of PDLs, ensuring functional tissue homeostasis. The proposed framework in this study is also applicable for investigating the structure-function relationship in broader types of fibrous soft-/hard- tissue interfaces.
Collapse
Affiliation(s)
- Jingxiao Zhong
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Yo Shibata
- Department of Biomaterials and Engineering, Showa University School of Dentistry, Tokyo, Japan
| | - Chi Wu
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Chie Watanabe
- Department of Biomaterials and Engineering, Showa University School of Dentistry, Tokyo, Japan
| | - Junning Chen
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Keke Zheng
- Institute for Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, UK
| | - Jingrui Hu
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Michael V Swain
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
14
|
Agarwal T, Chiesa I, Costantini M, Lopamarda A, Tirelli MC, Borra OP, Varshapally SVS, Kumar YAV, Koteswara Reddy G, De Maria C, Zhang LG, Maiti TK. Chitosan and its derivatives in 3D/4D (bio) printing for tissue engineering and drug delivery applications. Int J Biol Macromol 2023; 246:125669. [PMID: 37406901 DOI: 10.1016/j.ijbiomac.2023.125669] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/19/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Tissue engineering research has undergone to a revolutionary improvement, thanks to technological advancements, such as the introduction of bioprinting technologies. The ability to develop suitable customized biomaterial inks/bioinks, with excellent printability and ability to promote cell proliferation and function, has a deep impact on such improvements. In this context, printing inks based on chitosan and its derivatives have been instrumental. Thus, the current review aims at providing a comprehensive overview on chitosan-based materials as suitable inks for 3D/4D (bio)printing and their applicability in creating advanced drug delivery platforms and tissue engineered constructs. Furthermore, relevant strategies to improve the mechanical and biological performances of this biomaterial are also highlighted.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India.
| | - Irene Chiesa
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland.
| | - Anna Lopamarda
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | | | - Om Prakash Borra
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | | | | | - G Koteswara Reddy
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Carmelo De Maria
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy.
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of technology Kharagpur, West Bengal 721302, India
| |
Collapse
|
15
|
Akiyama N, Patel KD, Jang EJ, Shannon MR, Patel R, Patel M, Perriman AW. Tubular nanomaterials for bone tissue engineering. J Mater Chem B 2023; 11:6225-6248. [PMID: 37309580 DOI: 10.1039/d3tb00905j] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanomaterial composition, morphology, and mechanical performance are critical parameters for tissue engineering. Within this rapidly expanding space, tubular nanomaterials (TNs), including carbon nanotubes (CNTs), titanium oxide nanotubes (TNTs), halloysite nanotubes (HNTs), silica nanotubes (SiNTs), and hydroxyapatite nanotubes (HANTs) have shown significant potential across a broad range of applications due to their high surface area, versatile surface chemistry, well-defined mechanical properties, excellent biocompatibility, and monodispersity. These include drug delivery vectors, imaging contrast agents, and scaffolds for bone tissue engineering. This review is centered on the recent developments in TN-based biomaterials for structural tissue engineering, with a strong focus on bone tissue regeneration. It includes a detailed literature review on TN-based orthopedic coatings for metallic implants and composite scaffolds to enhance in vivo bone regeneration.
Collapse
Affiliation(s)
- Naomi Akiyama
- Department of Chemical Engineering, The Cooper Union of the Advancement of Science and Art, New York City, NY 10003, USA
| | - Kapil D Patel
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Eun Jo Jang
- Nano Science and Engineering (NSE), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, Yeonsu-gu, Incheon 21983, South Korea
| | - Mark R Shannon
- Bristol Composites Institute (BCI), University of Bristol, Bristol, BS8 1UP, UK
| | - Rajkumar Patel
- Energy and Environmental Science and Engineering (EESE), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, Yeonsu-gu, Incheon 21983, South Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea.
| | - Adam Willis Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
16
|
Kim MK, Paek K, Woo SM, Kim JA. Bone-on-a-Chip: Biomimetic Models Based on Microfluidic Technologies for Biomedical Applications. ACS Biomater Sci Eng 2023. [PMID: 37183366 DOI: 10.1021/acsbiomaterials.3c00066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
With the increasing importance of preclinical evaluation of newly developed drugs or treatments, in vitro organ or disease models are necessary. Although various organ-specific on-chip (organ-on-a-chip, or OOC) systems have been developed as emerging in vitro models, bone-on-a-chip (BOC) systems that recapitulate the bone microenvironment have been less developed or reviewed compared with other OOCs. The bone is one of the most dynamic organs and undergoes continuous remodeling throughout its lifetime. The aging population is growing worldwide, and healthcare costs are rising rapidly. Since in vitro BOC models that recapitulate native bone niches and pathological features can be important for studying the underlying mechanism of orthopedic diseases and predicting drug responses in preclinical trials instead of in animals, the development of biomimetic BOCs with high efficiency and fidelity will be accelerated further. Here, we review recently engineered BOCs developed using various microfluidic technologies and investigate their use to model the bone microenvironment. We have also explored various biomimetic strategies based on biological, geometrical, and biomechanical cues for biomedical applications of BOCs. Finally, we addressed the limitations and challenging issues of current BOCs that should be overcome to obtain more acceptable BOCs in the biomedical and pharmaceutical industries.
Collapse
Affiliation(s)
- Min Kyeong Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Kyurim Paek
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Program in Biomicro System Technology, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Mi Woo
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Republic of Korea
| |
Collapse
|
17
|
Sultan N, Jayash SN. Evaluation of osteogenic potential of demineralized dentin matrix hydrogel for bone formation. BMC Oral Health 2023; 23:247. [PMID: 37118728 PMCID: PMC10148431 DOI: 10.1186/s12903-023-02928-w] [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: 12/31/2022] [Accepted: 03/31/2023] [Indexed: 04/30/2023] Open
Abstract
OBJECTIVES Dentin, the bulk material of the tooth, resemble the bone's chemical composition and is considered a valuable bone substitute. In the current study, we assessed the cytotoxicity and osteogenic potential of demineralized dentin matrix (DDM) in comparison to HA nanoparticles (n-HA) on bone marrow mesenchymal stem cells (BMMSCs) using a hydrogel formulation. MATERIALS AND METHODS Human extracted teeth were minced into particles and treated via chemical demineralization using ethylene diamine tetra-acetic acid solution (EDTA) to produce DDM particles. DDM and n-HA particles were added to the sodium alginate then, the combination was dripped into a 5% (w/v) calcium chloride solution to obtain DDM hydrogel (DDMH) or nano-hydroxyapatite hydrogel (NHH). The particles were evaluated by dynamic light scattering (DLS) and the hydrogels were evaluated via scanning electron microscope (SEM). BMMSCs were treated with different hydrogel concentrations (25%, 50%, 75% and neat/100%) and cell viability was evaluated using MTT assay after 72 h of culture. Collagen-I (COL-I) gene expression was studied with real-time quantitative polymerase chain reaction (RT-qPCR) after 3 weeks of culture and alkaline phosphatase (ALP) activity was assessed using enzyme-linked immune sorbent assay (ELISA) over 7th, 10th, 14th and 21st days of culture. BMMSCs seeded in a complete culture medium were used as controls. One-way ANOVA was utilized to measure the significant differences in the tested groups. RESULTS DLS measurements revealed that DDM and n-HA particles had negative values of zeta potential. SEM micrographs showed a porous microstructure of the tested hydrogels. The viability results revealed that 100% concentrations of either DDMH or NHH were cytotoxic to BMMSCs after 72 h of culture. However, the cytotoxicity of 25% and 50% concentrations of DDMH were not statistically significant compared to the control group. RT-qPCR showed that COL-I gene expression was significantly upregulated in BMMSCs cultured with 50% DDMH compared to all other treated or control groups (P < 0.01). ELISA analysis revealed that ALP level was significantly increased in the groups treated with 50% DDMH compared to 50% NHH after 21 days in culture (P < 0.001). CONCLUSION The injectable hydrogel containing demineralized dentin matrix was successfully formulated. DDMH has a porous structure and has been shown to provide a supporting matrix for the viability and differentiation of BMMSCs. A 50% concentration of DDMH was revealed to be not cytotoxic to BMMSCs and may have a great potential to promote bone formation ability.
Collapse
Affiliation(s)
- Nessma Sultan
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.
| | | |
Collapse
|
18
|
Cheema U. Position Paper Progress in the development of biomimetic engineered human tissues. J Tissue Eng 2023; 14:20417314221145663. [PMID: 36874985 PMCID: PMC9974615 DOI: 10.1177/20417314221145663] [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: 06/21/2022] [Accepted: 11/28/2022] [Indexed: 03/07/2023] Open
Abstract
Tissue engineering (TE) is the multi-disciplinary approach to building 3D human tissue equivalents in the laboratory. The advancement of medical sciences and allied scientific disciplines have aspired to engineer human tissues for three decades. To date there is limited use of TE tissues/organs as replacement body parts in humans. This position paper outlines advances in engineering of specific tissues and organs with tissue-specific challenges. This paper outlines the technologies most successful for engineering tissues and key areas of advancement.
Collapse
Affiliation(s)
- Umber Cheema
- Division of Surgery and interventional science, UCL Centre for 3D models of Health and Disease, Fitzrovia
| |
Collapse
|
19
|
Alamán‐Díez P, García‐Gareta E, Arruebo M, Pérez MÁ. A bone-on-a-chip collagen hydrogel-based model using pre-differentiated adipose-derived stem cells for personalized bone tissue engineering. J Biomed Mater Res A 2023; 111:88-105. [PMID: 36205241 PMCID: PMC9828068 DOI: 10.1002/jbm.a.37448] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/24/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023]
Abstract
Mesenchymal stem cells have contributed to the continuous progress of tissue engineering and regenerative medicine. Adipose-derived stem cells (ADSC) possess many advantages compared to other origins including easy tissue harvesting, self-renewal potential, and fast population doubling time. As multipotent cells, they can differentiate into osteoblastic cell linages. In vitro bone models are needed to carry out an initial safety assessment in the study of novel bone regeneration therapies. We hypothesized that 3D bone-on-a-chip models containing ADSC could closely recreate the physiological bone microenvironment and promote differentiation. They represent an intermedium step between traditional 2D-in vitro and in vivo experiments facilitating the screening of therapeutic molecules while saving resources. Herein, we have differentiated ADSC for 7 and 14 days and used them to fabricate in vitro bone models by embedding the pre-differentiated cells in a 3D collagen matrix placed in a microfluidic chip. Osteogenic markers such as alkaline phosphatase activity, calcium mineralization, changes on cell morphology, and expression of specific proteins (bone sialoprotein 2, dentin matrix acidic phosphoprotein-1, and osteocalcin) were evaluated to determine cell differentiation potential and evolution. This is the first miniaturized 3D-in vitro bone model created from pre-differentiated ADSC embedded in a hydrogel collagen matrix which could be used for personalized bone tissue engineering.
Collapse
Affiliation(s)
- Pilar Alamán‐Díez
- Multiscale in Mechanical and Biological Engineering, Aragón Institute of Engineering Research (I3A), Aragón Institute of Healthcare Research (IIS Aragón)University of ZaragozaZaragozaSpain
| | - Elena García‐Gareta
- Multiscale in Mechanical and Biological Engineering, Aragón Institute of Engineering Research (I3A), Aragón Institute of Healthcare Research (IIS Aragón)University of ZaragozaZaragozaSpain,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental InstituteUniversity College LondonLondonUK
| | - Manuel Arruebo
- Aragón Institute of Nanoscience and Materials (INMA), Consejo Superior de Investigaciones Científicas (CSIC)University of ZaragozaZaragozaSpain,Department of Chemical EngineeringUniversity of ZaragozaZaragozaSpain
| | - María Ángeles Pérez
- Multiscale in Mechanical and Biological Engineering, Aragón Institute of Engineering Research (I3A), Aragón Institute of Healthcare Research (IIS Aragón)University of ZaragozaZaragozaSpain
| |
Collapse
|
20
|
Kohli N, Theodoridis K, Hall TAG, Sanz-Pena I, Gaboriau DCA, van Arkel RJ. Bioreactor analyses of tissue ingrowth, ongrowth and remodelling around implants: An alternative to live animal testing. Front Bioeng Biotechnol 2023; 11:1054391. [PMID: 36890911 PMCID: PMC9986429 DOI: 10.3389/fbioe.2023.1054391] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction: Preclinical assessment of bone remodelling onto, into or around novel implant technologies is underpinned by a large live animal testing burden. The aim of this study was to explore whether a lab-based bioreactor model could provide similar insight. Method: Twelve ex vivo trabecular bone cylinders were extracted from porcine femora and were implanted with additively manufactured stochastic porous titanium implants. Half were cultured dynamically, in a bioreactor with continuous fluid flow and daily cyclic loading, and half in static well plates. Tissue ongrowth, ingrowth and remodelling around the implants were evaluated with imaging and mechanical testing. Results: For both culture conditions, scanning electron microscopy (SEM) revealed bone ongrowth; widefield, backscatter SEM, micro computed tomography scanning, and histology revealed mineralisation inside the implant pores; and histology revealed woven bone formation and bone resorption around the implant. The imaging evidence of this tissue ongrowth, ingrowth and remodelling around the implant was greater for the dynamically cultured samples, and the mechanical testing revealed that the dynamically cultured samples had approximately three times greater push-through fixation strength (p < 0.05). Discussion: Ex vivo bone models enable the analysis of tissue remodelling onto, into and around porous implants in the lab. While static culture conditions exhibited some characteristics of bony adaptation to implantation, simulating physiological conditions with a bioreactor led to an accelerated response.
Collapse
Affiliation(s)
- Nupur Kohli
- Biomechanics Group, Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | - Konstantinos Theodoridis
- Biomechanics Group, Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | - Thomas A G Hall
- Biomechanics Group, Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | - Inigo Sanz-Pena
- Biomechanics Group, Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | - David C A Gaboriau
- FILM, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Richard J van Arkel
- Biomechanics Group, Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Mertz EL, Makareeva E, Mirigian LS, Leikin S. Bone Formation in 2D Culture of Primary Cells. JBMR Plus 2022; 7:e10701. [PMID: 36699640 PMCID: PMC9850442 DOI: 10.1002/jbm4.10701] [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: 07/21/2022] [Revised: 09/15/2022] [Accepted: 10/16/2022] [Indexed: 11/13/2022] Open
Abstract
Relevance of mineralized nodules in two-dimensional (2D) osteoblast/osteocyte cultures to bone biology, pathology, and engineering is a decades old question, but a comprehensive answer appears to be still wanting. Bone-like cells, extracellular matrix (ECM), and mineral were all reported but so were non-bone-like ones. Many studies described seemingly bone-like cell-ECM structures based on similarity to few select bone features in vivo, yet no studies examined multiple bone features simultaneously and none systematically studied all types of structures coexisting in the same culture. Here, we report such comprehensive analysis of 2D cultures based on light and electron microscopies, Raman microspectroscopy, gene expression, and in situ messenger RNA (mRNA) hybridization. We demonstrate that 2D cultures of primary cells from mouse calvaria do form bona fide bone. Cells, ECM, and mineral within it exhibit morphology, structure, ultrastructure, composition, spatial-temporal gene expression pattern, and growth consistent with intramembranous ossification. However, this bone is just one of at least five different types of cell-ECM structures coexisting in the same 2D culture, which vary widely in their resemblance to bone and ability to mineralize. We show that the other two mineralizing structures may represent abnormal (disrupted) bone and cartilage-like structure with chondrocyte-to-osteoblast transdifferentiation. The two nonmineralizing cell-ECM structures may mimic periosteal cambium and pathological, nonmineralizing osteoid. Importantly, the most commonly used culture conditions (10mM β-glycerophosphate) induce artificial mineralization of all cell-ECM structures, which then become barely distinguishable. We therefore discuss conditions and approaches promoting formation of bona fide bone and simple means for distinguishing it from the other cell-ECM structures. Our findings may improve osteoblast differentiation and function analyses based on 2D cultures and extend applications of these cultures to general bone biology and tissue engineering research. Published 2022. This article is a U.S. Government work and is in the public domain in the USA. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Edward L. Mertz
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Elena Makareeva
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Lynn S. Mirigian
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Sergey Leikin
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
22
|
Carter SSD, Atif AR, Diez-Escudero A, Grape M, Ginebra MP, Tenje M, Mestres G. A microfluidic-based approach to investigate the inflammatory response of macrophages to pristine and drug-loaded nanostructured hydroxyapatite. Mater Today Bio 2022; 16:100351. [PMID: 35865408 PMCID: PMC9294551 DOI: 10.1016/j.mtbio.2022.100351] [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: 04/10/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 11/28/2022] Open
Abstract
The in vitro biological characterization of biomaterials is largely based on static cell cultures. However, for highly reactive biomaterials such as calcium-deficient hydroxyapatite (CDHA), this static environment has limitations. Drastic alterations in the ionic composition of the cell culture medium can negatively affect cell behavior, which can lead to misleading results or data that is difficult to interpret. This challenge could be addressed by a microfluidics-based approach (i.e. on-chip), which offers the opportunity to provide a continuous flow of cell culture medium and a potentially more physiologically relevant microenvironment. The aim of this work was to explore microfluidic technology for its potential to characterize CDHA, particularly in the context of inflammation. Two different CDHA substrates (chemically identical, but varying in microstructure) were integrated on-chip and subsequently evaluated. We demonstrated that the on-chip environment can avoid drastic ionic alterations and increase protein sorption, which was reflected in cell studies with RAW 264.7 macrophages. The cells grown on-chip showed a high cell viability and enhanced proliferation compared to cells maintained under static conditions. Whereas no clear differences in the secretion of tumor necrosis factor alpha (TNF-α) were found, variations in cell morphology suggested a more anti-inflammatory environment on-chip. In the second part of this study, the CDHA substrates were loaded with the drug Trolox. We showed that it is possible to characterize drug release on-chip and moreover demonstrated that Trolox affects the TNF-α secretion and morphology of RAW 264.7 cells. Overall, these results highlight the potential of microfluidics to evaluate (bioactive) biomaterials, both in pristine form and when drug-loaded. This is of particular interest for the latter case, as it allows the biological characterization and assessment of drug release to take place under the same dynamic in vitro environment.
Collapse
Affiliation(s)
- Sarah-Sophia D Carter
- Division of Biomedical Engineering, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22, Uppsala, Sweden
| | - Abdul-Raouf Atif
- Division of Biomedical Engineering, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22, Uppsala, Sweden
| | - Anna Diez-Escudero
- Ortholab, Department of Surgical Sciences-Orthopaedics, Uppsala University, Uppsala, 751 85, Sweden
| | - Maja Grape
- Division of Biomedical Engineering, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22, Uppsala, Sweden
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Departament de Ciència i Enginyeria de Materials, Universitat Politècnica de Catalunya (UPC), 08930, Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, 08930, Barcelona, Spain.,Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10-12, 08028, Barcelona, Spain
| | - Maria Tenje
- Division of Biomedical Engineering, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22, Uppsala, Sweden
| | - Gemma Mestres
- Division of Biomedical Engineering, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22, Uppsala, Sweden
| |
Collapse
|
23
|
Smits A, Annaert P, Cavallaro G, De Cock PAJG, de Wildt SN, Kindblom JM, Lagler FB, Moreno C, Pokorna P, Schreuder MF, Standing JF, Turner MA, Vitiello B, Zhao W, Weingberg AM, Willmann R, van den Anker J, Allegaert K. Current knowledge, challenges and innovations in developmental pharmacology: A combined conect4children Expert Group and European Society for Developmental, Perinatal and Paediatric Pharmacology White Paper. Br J Clin Pharmacol 2022; 88:4965-4984. [PMID: 34180088 PMCID: PMC9787161 DOI: 10.1111/bcp.14958] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/22/2021] [Accepted: 05/30/2021] [Indexed: 12/30/2022] Open
Abstract
Developmental pharmacology describes the impact of maturation on drug disposition (pharmacokinetics, PK) and drug effects (pharmacodynamics, PD) throughout the paediatric age range. This paper, written by a multidisciplinary group of experts, summarizes current knowledge, and provides suggestions to pharmaceutical companies, regulatory agencies and academicians on how to incorporate the latest knowledge regarding developmental pharmacology and innovative techniques into neonatal and paediatric drug development. Biological aspects of drug absorption, distribution, metabolism and excretion throughout development are summarized. Although this area made enormous progress during the last two decades, remaining knowledge gaps were identified. Minimal risk and burden designs allow for optimally informative but minimally invasive PK sampling, while concomitant profiling of drug metabolites may provide additional insight in the unique PK behaviour in children. Furthermore, developmental PD needs to be considered during drug development, which is illustrated by disease- and/or target organ-specific examples. Identifying and testing PD targets and effects in special populations, and application of age- and/or population-specific assessment tools are discussed. Drug development plans also need to incorporate innovative techniques such as preclinical models to study therapeutic strategies, and shift from sequential enrolment of subgroups, to more rational designs. To stimulate appropriate research plans, illustrations of specific PK/PD-related as well as drug safety-related challenges during drug development are provided. The suggestions made in this joint paper of the Innovative Medicines Initiative conect4children Expert group on Developmental Pharmacology and the European Society for Developmental, Perinatal and Paediatric Pharmacology, should facilitate all those involved in drug development.
Collapse
Affiliation(s)
- Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Neonatal intensive Care unit, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Giacomo Cavallaro
- Neonatal intensive care unit, Fondazione IRCCS Ca' Grande Ospedale Maggiore Policlinico, Milan, Italy
| | - Pieter A J G De Cock
- Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium.,Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium.,Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
| | - Saskia N de Wildt
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pharmacology and Toxicology, Radboud Institute Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jenny M Kindblom
- Pediatric Clinical Research Center, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Florian B Lagler
- Institute for Inherited Metabolic Diseases and Department of Pediatrics, Paracelsus Medical University, Clinical Research Center Salzburg, Salzburg, Austria
| | - Carmen Moreno
- Institute of Psychiatry and Mental Health, Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Paula Pokorna
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Physiology and Pharmacology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Amalia Children's Hospital, Nijmegen, the Netherlands
| | - Joseph F Standing
- UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Mark A Turner
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool Health Partners, Liverpool, UK
| | - Benedetto Vitiello
- Division of Child and Adolescent Neuropsychiatry, Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, China.,Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | | | | | - John van den Anker
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Department of Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
24
|
Wang P, Wang X. Mimicking the native bone regenerative microenvironment for in situ repair of large physiological and pathological bone defects. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
25
|
Alipour S, Nour S, Attari SM, Mohajeri M, Kianersi S, Taromian F, Khalkhali M, Aninwene GE, Tayebi L. A review on in vitro/ in vivo response of additively manufactured Ti-6Al-4V alloy. J Mater Chem B 2022; 10:9479-9534. [PMID: 36305245 DOI: 10.1039/d2tb01616h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bone replacement using porous and solid metallic implants, such as Ti-alloy implants, is regarded as one of the most practical therapeutic approaches in biomedical engineering. The bone is a complex tissue with various mechanical properties based on the site of action. Patient-specific Ti-6Al-4V constructs may address the key needs in bone treatment for having customized implants that mimic the complex structure of the natural tissue and diminish the risk of implant failure. This review focuses on the most promising methods of fabricating such patient-specific Ti-6Al-4V implants using additive manufacturing (AM) with a specific emphasis on the popular subcategory, which is powder bed fusion (PBF). Characteristics of the ideal implant to promote optimized tissue-implant interactions, as well as physical, mechanical/chemical treatments and modifications will be discussed. Accordingly, such investigations will be classified into 3B-based approaches (Biofunctionality, Bioactivity, and Biostability), which mainly govern native body response and ultimately the success in implantation.
Collapse
Affiliation(s)
- Saeid Alipour
- Department of Materials Science and Engineering, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Shirin Nour
- Tissue Engineering Group, Department of Biomedical Engineering, University of Melbourne, VIC 3010, Australia.,Polymer Science Group, Department of Chemical Engineering, University of Melbourne, VIC 3010, Australia
| | - Seyyed Morteza Attari
- Department of Material Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, TX, USA
| | - Sogol Kianersi
- CÚRAM, SFI Centre for Research in Medical Devices, Biomedical Sciences, University of Galway, Galway, Ireland
| | - Farzaneh Taromian
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mohammadparsa Khalkhali
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - George E Aninwene
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA.,California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, California, USA
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, Wisconsin, USA.
| |
Collapse
|
26
|
Kumar A, Sakhare K, Bhattacharya D, Chattopadhyay R, Parikh P, Narayan KP, Mukherjee A. Communication in non-communicable diseases (NCDs) and role of immunomodulatory nutraceuticals in their management. Front Nutr 2022; 9:966152. [PMID: 36211513 PMCID: PMC9532975 DOI: 10.3389/fnut.2022.966152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Conveyance of pathogens between organisms causes communicable diseases. On the other hand, a non-communicable disease (NCD) was always thought to have no causative transmissible infective agents. Today, this clear distinction is increasingly getting blurred and NCDs are found to be associated with some transmissible components. The human microbiota carries a congregation of microbes, the majority and the most widely studied being bacteria in the gut. The adult human gut harbors ginormous inhabitant microbes, and the microbiome accommodates 150-fold more genes than the host genome. Microbial communities share a mutually beneficial relationship with the host, especially with respect to host physiology including digestion, immune responses, and metabolism. This review delineates the connection between environmental factors such as infections leading to gut dysbiosis and NCDs and explores the evidence regarding possible causal link between them. We also discuss the evidence regarding the value of appropriate therapeutic immunomodulatory nutritional interventions to reduce the development of such diseases. We behold such immunomodulatory effects have the potential to influence in various NCDs and restore homeostasis. We believe that the beginning of the era of microbiota-oriented personalized treatment modalities is not far away.
Collapse
Affiliation(s)
- Abhiram Kumar
- Esperer Onco Nutrition Pvt. Ltd., Mumbai, India
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | | | - Purvish Parikh
- Department of Clinical Haematology, Mahatma Gandhi Medical College and Hospital, Jaipur, India
| | - Kumar P. Narayan
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
- *Correspondence: Kumar P. Narayan,
| | | |
Collapse
|
27
|
Negrescu AM, Mitran V, Draghicescu W, Popescu S, Pirvu C, Ionascu I, Soare T, Uzun S, Croitoru SM, Cimpean A. TiO2 Nanotubes Functionalized with Icariin for an Attenuated In Vitro Immune Response and Improved In Vivo Osseointegration. J Funct Biomater 2022; 13:jfb13020043. [PMID: 35466225 PMCID: PMC9036299 DOI: 10.3390/jfb13020043] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/30/2022] Open
Abstract
Due to their superior mechanical and chemical properties, titanium (Ti) and its alloys have been widely used as orthopedic implantable devices. However, their bioinertness represents a limitation, which can be overcome by employing various surface modifications, such as TiO2 nanotube (TNT) fabrication via electrochemical anodization. Anodic TNTs present tunable dimensions and unique structures, turning them into feasible drug delivery platforms. In the present work, TNTs were loaded with icariin (Ica) through an adhesive intermediate layer of polydopamine (DP), and their in vitro and in vivo biological performance was evaluated. The successful fabrication of the modified surfaces was verified by scanning electron microscopy (SEM), atomic force microscopy (AFM), Fourier transform infrared spectroscopy (FTIR), and contact angle measurements (CA), while the in vitro release of Ica was evaluated via UV-VIS spectrophotometry. In terms of in vitro behaviour, comparative studies on RAW 264.7 macrophages demonstrated that the TNT substrates, especially TNT-DP-Ica, elicited a lower inflammatory response compared to the Ti support. Moreover, the in vivo implantation studies evinced generation of a reduced fibrotic capsule around this implant and increased thickness of the newly formed bone tissue at 1 month and 3 months post-implantation, respectively. Overall, our results indicate that the controlled release of Ica from TNT surfaces could result in an improved osseointegration process.
Collapse
Affiliation(s)
- Andreea-Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (V.M.)
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (V.M.)
| | - Wanda Draghicescu
- Faculty of Chemical Engineering and Biotechnology, University Politehnica of Bucharest, 1-7 Polizu, 011061 Bucharest, Romania; (W.D.); (S.P.); (C.P.)
| | - Simona Popescu
- Faculty of Chemical Engineering and Biotechnology, University Politehnica of Bucharest, 1-7 Polizu, 011061 Bucharest, Romania; (W.D.); (S.P.); (C.P.)
| | - Cristian Pirvu
- Faculty of Chemical Engineering and Biotechnology, University Politehnica of Bucharest, 1-7 Polizu, 011061 Bucharest, Romania; (W.D.); (S.P.); (C.P.)
- Faculty of Medical Engineering, University Politehnica of Bucharest, 1-7 Polizu, 011061 Bucharest, Romania
| | - Iuliana Ionascu
- Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 105 Spl. Independentei, 050097 Bucharest, Romania; (I.I.); (T.S.); (S.U.)
| | - Teodoru Soare
- Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 105 Spl. Independentei, 050097 Bucharest, Romania; (I.I.); (T.S.); (S.U.)
| | - Seralp Uzun
- Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 105 Spl. Independentei, 050097 Bucharest, Romania; (I.I.); (T.S.); (S.U.)
| | - Sorin Mihai Croitoru
- Machines and Manufacturing Systems Department, University Politehnica of Bucharest, 313 Spl. Independentei, 060042 Bucharest, Romania;
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (V.M.)
- Correspondence: ; Tel.: +40-21-318-1575 (ext. 106)
| |
Collapse
|
28
|
Tanaka M, Izumiya M, Haniu H, Ueda K, Ma C, Ueshiba K, Ideta H, Sobajima A, Uchiyama S, Takahashi J, Saito N. Current Methods in the Study of Nanomaterials for Bone Regeneration. NANOMATERIALS 2022; 12:nano12071195. [PMID: 35407313 PMCID: PMC9000656 DOI: 10.3390/nano12071195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022]
Abstract
Nanomaterials show great promise as bone regeneration materials. They can be used as fillers to strengthen bone regeneration scaffolds, or employed in their natural form as carriers for drug delivery systems. A variety of experiments have been conducted to evaluate the osteogenic potential of bone regeneration materials. In vivo, such materials are commonly tested in animal bone defect models to assess their bone regeneration potential. From an ethical standpoint, however, animal experiments should be minimized. A standardized in vitro strategy for this purpose is desirable, but at present, the results of studies conducted under a wide variety of conditions have all been evaluated equally. This review will first briefly introduce several bone regeneration reports on nanomaterials and the nanosize-derived caveats of evaluations in such studies. Then, experimental techniques (in vivo and in vitro), types of cells, culture media, fetal bovine serum, and additives will be described, with specific examples of the risks of various culture conditions leading to erroneous conclusions in biomaterial analysis. We hope that this review will create a better understanding of the evaluation of biomaterials, including nanomaterials for bone regeneration, and lead to the development of versatile assessment methods that can be widely used in biomaterial development.
Collapse
Affiliation(s)
- Manabu Tanaka
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Chuang Ma
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Koki Ueshiba
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hirokazu Ideta
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Atsushi Sobajima
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
- Department of Orthopedics (Lower Limbs), Social Medical Care Corporation Hosei-kai Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, Nagano 390-8601, Japan
| | - Shigeharu Uchiyama
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
| | - Jun Takahashi
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
| |
Collapse
|
29
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
30
|
Abstract
Bone is an outstanding, well-designed composite. It is constituted by a multi-level structure wherein its properties and behavior are dependent on its composition and structural organization at different length scales. The combination of unique mechanical properties with adaptive and self-healing abilities makes bone an innovative model for the future design of synthetic biomimetic composites with improved performance in bone repair and regeneration. However, the relation between structure and properties in bone is very complex. In this review article, we intend to describe the hierarchical organization of bone on progressively greater scales and present the basic concepts that are fundamental to understanding the arrangement-based mechanical properties at each length scale and their influence on bone’s overall structural behavior. The need for a better understanding of bone’s intricate composite structure is also highlighted.
Collapse
|
31
|
Zheng Y, Gao A, Bai J, Liao Q, Wu Y, Zhang W, Guan M, Tong L, Geng D, Zhao X, Chu PK, Wang H. A programmed surface on polyetheretherketone for sequentially dictating osteoimmunomodulation and bone regeneration to achieve ameliorative osseointegration under osteoporotic conditions. Bioact Mater 2022; 14:364-376. [PMID: 35386814 PMCID: PMC8964985 DOI: 10.1016/j.bioactmat.2022.01.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Polyetheretherketone (PEEK) is a desirable alternative to conventional biomedical metals for orthopedic implants due to the excellent mechanical properties. However, the inherent bioinertness of PEEK contributes to inferior osseointegration of PEEK implants, especially under pathological conditions of osteoporosis. Herein, a programmed surface is designed and fabricated on PEEK to dictate osteoimmunomodulation and bone regeneration sequentially. A degradable hybrid coating consisting of poly(lactide-co-glycolide) and alendronate (ALN) loaded nano-hydroxyapatite is deposited on PEEK and then interleukin-4 (IL-4) is grafted onto the outer surface of the hybrid coating with the aid of N2 plasma immersion ion implantation and subsequent immersion in IL-4 solution. Dominant release of IL-4 together with ALN and Ca2+ during the first few days synergistically mitigates the early acute inflammatory reactions and creates an osteoimmunomodulatory microenvironment that facilitates bone regeneration. Afterwards, slow and sustained delivery of ALN and Ca2+ in the following weeks boosts osteogenesis and suppresses osteoclastogenesis simultaneously, consequently ameliorating bone-implant osseointegration even under osteoporotic conditions. By taking into account the different phases in bone repair, this strategy of constructing advanced bone implants with sequential functions provides customizable and clinically viable therapy to osteoporotic patients. A programmed surface is designed and fabricated on PEEK to dictate osteoimmunomodulation and bone regeneration sequentially. A degradable coating consisting ALN loaded nano-HA is deposited on PEEK, with IL-4 being grafted onto the outmost surface. Dominant release of IL-4 together with ALN and Ca2+ synergistically mitigates the early acute inflammatory reactions. Slow and sustained delivery of ALN and Ca2+ boosts osteogenesis and suppresses osteoclastogenesis simultaneously. Sequential regulation of peri-implant biological responses is achieved to match the dynamic process of bone regeneration.
Collapse
Affiliation(s)
- Yanyan Zheng
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China
| | - Ang Gao
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qing Liao
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuzheng Wu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Wei Zhang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Min Guan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liping Tong
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Corresponding author
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Corresponding author
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Paul K. Chu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Corresponding author
| |
Collapse
|
32
|
Luo Q, Li X, Zhong W, Cao W, Zhu M, Wu A, Chen W, Ye Z, Han Q, Natarajan D, Pathak JL, Zhang Q. Dicalcium silicate-induced mitochondrial dysfunction and autophagy-mediated macrophagic inflammation promotes osteogenic differentiation of BMSCs. Regen Biomater 2021; 9:rbab075. [PMID: 35480858 PMCID: PMC9039510 DOI: 10.1093/rb/rbab075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 11/14/2022] Open
Abstract
Dicalcium silicate (Ca2SiO4, C2S) has osteogenic potential but induces macrophagic inflammation. Mitochondrial function plays a vital role in macrophage polarization and macrophagic inflammation. The mitochondrial function of C2S-treated macrophages is still unclear. This study hypothesized: (i) the C2S modulates mitochondrial function and autophagy in macrophages to regulate macrophagic inflammation, and (ii) C2S-induced macrophagic inflammation regulates osteogenesis. We used RAW264.7 cells as a model of macrophage. The C2S (75-150 μg/ml) extract was used to analyze the macrophagic mitochondrial function and macrophage-mediated effect on osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (BMSCs). The results showed that C2S extract (150 μg/ml) induced TNF-α, IL-1β and IL-6 production in macrophages. C2S extract (150 μg/ml) enhanced reactive oxygen species level and intracellular calcium level but reduced mitochondrial membrane potential and ATP production. TEM images showed reduced mitochondrial abundance and altered the mitochondrial morphology in C2S (150 μg/ml)-treated macrophages. Protein level expression of PINK1, Parkin, Beclin1 and LC3 was upregulated but TOMM20 was downregulated. mRNA sequencing and KEGG analysis showed that C2S-induced differentially expressed mRNAs in macrophages were mainly distributed in the essential signaling pathways involved in mitochondrial function and autophagy. The conditioned medium from C2S-treated macrophage robustly promoted osteogenic differentiation in BMSCs. In conclusion, our results indicate mitochondrial dysfunction and autophagy as the possible mechanism of C2S-induced macrophagic inflammation. The promotion of osteogenic differentiation of BMSCs by the C2S-induced macrophagic inflammation suggests the potential application of C2S in developing immunomodulatory bone grafts.
Collapse
Affiliation(s)
- Qianting Luo
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen 529030, China
| | - Xingyang Li
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wenchao Zhong
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wei Cao
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Department of Oral Cell Biology, Academic Centre of Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam 1081LA, The Netherlands
| | - Mingjing Zhu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Antong Wu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wanyi Chen
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Zhitong Ye
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qiao Han
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Duraipandy Natarajan
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Janak L Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| |
Collapse
|
33
|
Pinho LC, Alves MM, Colaço B, Fernandes MH, Santos C. Green-Synthesized Magnesium Hydroxide Nanoparticles Induced Osteoblastic Differentiation in Bone Co-Cultured Cells. Pharmaceuticals (Basel) 2021; 14:ph14121281. [PMID: 34959683 PMCID: PMC8704631 DOI: 10.3390/ph14121281] [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: 11/05/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023] Open
Abstract
In this work, magnesium hydroxide NPs were synthesized using water (Mg(OH)2 NPs) or a rose hip (RH) extract (Mg(OH)2RH NPs) and tested for the bone cells’ effects in co-cultured osteoblastic and osteoclastic cells, using a Transwell® insert system, allowing reciprocal cell paracrine interactions. Behavior of each cell population was characterized for typical phenotype markers, at days 1 and 6. Cell cultures treated with osteogenic/osteoclastogenic inducers were used as positive control of cell differentiation. The NPs presented a round shape morphology with an average diameter ~90 nm (Mg(OH)2 NPs) and below 10 nm (Mg(OH)2RH NPs. Both NPs induced osteoblastic and osteoclastic behavior similarly to that observed in induced osteoblastic and osteoclastic cultures (positive controls). Differences between the two types of particles were evident at the gene expression level. Compared to Mg(OH)2 NPs, the green-synthesized NPs greatly increased the expression of osteoblastic genes coding for the early markers ALP and collagen type 1 and the later transcription factor osterix, while decreasing the expression of osteoclastogenic genes, namely the essential transcription factor NFATC1, TRAP and the genes coding for the functional markers CA2 and CTSK. Overall, a positive added effect could be hypothesized for Mg(OH)2RH NPs with potential usefulness to promote bone formation in regenerative applications.
Collapse
Affiliation(s)
- Laura Costa Pinho
- Department of Animal Science, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal;
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal;
- Correspondence: (L.C.P.); (C.S.)
| | - Marta M. Alves
- CQE Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal;
| | - Bruno Colaço
- Department of Animal Science, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal;
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
- CECAV—Animal and Veterinary Research Centre UTAD, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Maria Helena Fernandes
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal;
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
| | - Catarina Santos
- EST Setúbal, CDP2T, Instituto Politécnico de Setúbal, Campus IPS, 2910-761 Setúbal, Portugal
- Correspondence: (L.C.P.); (C.S.)
| |
Collapse
|
34
|
Yang W, Li HY, Wu YF, Mi RJ, Liu WZ, Shen X, Lu YX, Jiang YH, Ma MJ, Shen HY. ac4C acetylation of RUNX2 catalyzed by NAT10 spurs osteogenesis of BMSCs and prevents ovariectomy-induced bone loss. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:135-147. [PMID: 34513300 PMCID: PMC8413676 DOI: 10.1016/j.omtn.2021.06.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 06/25/2021] [Indexed: 12/16/2022]
Abstract
N-acetyltransferase 10 (NAT10) is the key enzyme for N4-acetylcytidine (ac4C) modification of mRNA, which participates in various cellular processes and is related to many diseases. Here, we explore the relationships among osteoblast differentiation, NAT10, and ac4C, and we found that NAT0 expression and the ac4C level of total RNA were decreased in the bone tissues of bilateral ovariectomized (OVX) mice and osteoporosis patients. Adenoviruses overexpressing NAT10 reversed bone loss, and Remodelin, an NAT10 inhibitor, enhanced the loss of bone mass in OVX mice. Moreover, bone marrow-derived mesenchymal stem cells (BMSCs) with low-level ac4C modification formed fewer calcium nodules in vitro with NAT10 silencing, whereas BMSCs with high-level ac4C modification formed more calcium nodules with NAT10 overexpression. Moreover, we demonstrated that the ac4C level of runt-related transcription factor 2 (RUNX2) mRNA was increased after BMSCs were cultured in osteogenic medium (OM) and decreased after NAT10 silencing. The RUNX2 mRNA half-life and protein expression decreased after silencing NAT10 in BMSCs. Therefore, NAT10-based ac4C modification promotes the osteogenic differentiation of BMSCs by regulating the RUNX2 ac4C level. Because abnormal levels of NAT10 are probably one of the mechanisms responsible for osteoporosis, NAT10 is a new potential therapeutic target for this disease.
Collapse
Affiliation(s)
- W Yang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - H Y Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - Y F Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, People's Republic of China
| | - R J Mi
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, People's Republic of China
| | - W Z Liu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - X Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - Y X Lu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, People's Republic of China
| | - Y H Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - M J Ma
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - H Y Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| |
Collapse
|
35
|
Tomazela L, Cruz MAE, Nascimento LA, Fagundes CC, da Veiga MAMS, Zamarioli A, Bottini M, Ciancaglini P, Brassesco MS, Engel EE, Ramos AP. Fabrication and characterization of a bioactive polymethylmethacrylate-based porous cement loaded with strontium/calcium apatite nanoparticles. J Biomed Mater Res A 2021; 110:812-826. [PMID: 34783455 DOI: 10.1002/jbm.a.37330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 11/06/2022]
Abstract
Polymethylmethacrylate (PMMA)-based cements are used for bone reparation due to their biocompatibility, suitable mechanical properties, and mouldability. However, these materials suffer from high exothermic polymerization and poor bioactivity, which can cause the formation of fibrous tissue around the implant and aseptic loosening. Herein, we tackled these problems by adding Sr2+ -substituted hydroxyapatite nanoparticles (NPs) and a porogenic compound to the formulations, thus creating a microenvironment suitable for the proliferation of osteoblasts. The NPs resembled the structure of the bone's apatite and enabled the controlled release of Sr2+ . Trends in the X-ray patterns and infrared spectra confirmed that Sr2+ replaced Ca2+ in the whole composition range of the NPs. The inclusion of an effervescent additive reduced the polymerization temperature and lead to the formation of highly porous cement exhibiting mechanical properties comparable to the trabecular bone. The formation of an opened and interconnected matrix allowed osteoblasts to penetrate the cement structure. Most importantly, the gas formation confined the NPs at the surface of the pores, guaranteeing the controlled delivery of Sr2+ within a concentration sufficient to maintain osteoblast viability. Additionally, the cement was able to form apatite when immersed into simulated body fluids, further increasing its bioactivity. Therefore, we offer a formulation of PMMA cement with improved in vitro performance supported by enhanced bioactivity, increased osteoblast viability and deposition of mineralized matrix assigned to the loading with Sr2+ -substituted hydroxyapatite NPs and the creation of an interconnected porous structure. Altogether, our results hold promise for enhanced bone reparation guided by PMMA cements.
Collapse
Affiliation(s)
- Larissa Tomazela
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Marcos Antônio Eufrásio Cruz
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Larissa Aine Nascimento
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Cecilia C Fagundes
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ariane Zamarioli
- Departamento de Ortopedia e Anestesiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Maria Sol Brassesco
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Edgard E Engel
- Departamento de Ortopedia e Anestesiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Ren J, Kohli N, Sharma V, Shakouri T, Keskin-Erdogan Z, Saifzadeh S, Brierly GI, Knowles JC, Woodruff MA, García-Gareta E. Poly-ε-Caprolactone/Fibrin-Alginate Scaffold: A New Pro-Angiogenic Composite Biomaterial for the Treatment of Bone Defects. Polymers (Basel) 2021; 13:3399. [PMID: 34641215 PMCID: PMC8512525 DOI: 10.3390/polym13193399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022] Open
Abstract
We hypothesized that a composite of 3D porous melt-electrowritten poly-ɛ-caprolactone (PCL) coated throughout with a porous and slowly biodegradable fibrin/alginate (FA) matrix would accelerate bone repair due to its angiogenic potential. Scanning electron microscopy showed that the open pore structure of the FA matrix was maintained in the PCL/FA composites. Fourier transform infrared spectroscopy and differential scanning calorimetry showed complete coverage of the PCL fibres by FA, and the PCL/FA crystallinity was decreased compared with PCL. In vitro cell work with osteoprogenitor cells showed that they preferentially bound to the FA component and proliferated on all scaffolds over 28 days. A chorioallantoic membrane assay showed more blood vessel infiltration into FA and PCL/FA compared with PCL, and a significantly higher number of bifurcation points for PCL/FA compared with both FA and PCL. Implantation into a rat cranial defect model followed by microcomputed tomography, histology, and immunohistochemistry after 4- and 12-weeks post operation showed fast early bone formation at week 4, with significantly higher bone formation for FA and PCL/FA compared with PCL. However, this phenomenon was not extrapolated to week 12. Therefore, for long-term bone regeneration, tuning of FA degradation to ensure syncing with new bone formation is likely necessary.
Collapse
Affiliation(s)
- Jiongyu Ren
- Faculty of Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia; (J.R.); (G.I.B.); (M.A.W.)
| | - Nupur Kohli
- Regenerative Biomaterials Group, The RAFT Institute & The Griffin Institute, Northwick Park & Saint Mark’s Hospital, London HA1 3UJ, UK; (N.K.); (V.S.)
- Department of Mechanical Engineering, Imperial College London, London SW7 1AL, UK
| | - Vaibhav Sharma
- Regenerative Biomaterials Group, The RAFT Institute & The Griffin Institute, Northwick Park & Saint Mark’s Hospital, London HA1 3UJ, UK; (N.K.); (V.S.)
| | - Taleen Shakouri
- Division of Biomaterials & Tissue Engineering, Eastman Dental Institute, University College London, Rowland Hill Street, London NW3 2PF, UK; (T.S.); (Z.K.-E.); (J.C.K.)
| | - Zalike Keskin-Erdogan
- Division of Biomaterials & Tissue Engineering, Eastman Dental Institute, University College London, Rowland Hill Street, London NW3 2PF, UK; (T.S.); (Z.K.-E.); (J.C.K.)
| | - Siamak Saifzadeh
- Medical Engineering Research Facility, Queensland University of Technology, Brisbane, QLD 4059, Australia;
| | - Gary I. Brierly
- Faculty of Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia; (J.R.); (G.I.B.); (M.A.W.)
| | - Jonathan C. Knowles
- Division of Biomaterials & Tissue Engineering, Eastman Dental Institute, University College London, Rowland Hill Street, London NW3 2PF, UK; (T.S.); (Z.K.-E.); (J.C.K.)
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Maria A. Woodruff
- Faculty of Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia; (J.R.); (G.I.B.); (M.A.W.)
| | - Elena García-Gareta
- Regenerative Biomaterials Group, The RAFT Institute & The Griffin Institute, Northwick Park & Saint Mark’s Hospital, London HA1 3UJ, UK; (N.K.); (V.S.)
- Division of Biomaterials & Tissue Engineering, Eastman Dental Institute, University College London, Rowland Hill Street, London NW3 2PF, UK; (T.S.); (Z.K.-E.); (J.C.K.)
| |
Collapse
|
37
|
Liu W, Yu M, Chen F, Wang L, Ye C, Chen Q, Zhu Q, Xie D, Shao M, Yang L. A novel delivery nanobiotechnology: engineered miR-181b exosomes improved osteointegration by regulating macrophage polarization. J Nanobiotechnology 2021; 19:269. [PMID: 34493305 PMCID: PMC8424816 DOI: 10.1186/s12951-021-01015-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/25/2021] [Indexed: 12/18/2022] Open
Abstract
Background Many patients suffer from implant loosening after the implantation of titanium alloy caused by immune response to the foreign bodies and this could inhibit the following osteogenesis, which could possibly give rise to aseptic loosening and poor osteointegration while there is currently no appropriate solution in clinical practice. Exosome (Exo) carrying miRNA has been proven to be a suitable nanocarrier for solving this problem. In this study, we explored whether exosomes overexpressing miR-181b (Exo-181b) could exert beneficial effect on promoting M2 macrophage polarization, thus inhibiting inflammation as well as promoting osteogenesis and elaborated the underlying mechanism in vitro. Furthermore, we aimed to find whether Exo-181b could enhance osteointegration. Results In vitro, we firstly verified that Exo-181b significantly enhanced M2 polarization and inhibited inflammation by suppressing PRKCD and activating p-AKT. Then, in vivo, we verified that Exo-181b enhanced M2 polarization, reduced the inflammatory response and enhanced osteointegration. Also, we verified that the enhanced M2 polarization could indirectly promote the migration and osteogenic differentiation by secreting VEGF and BMP-2 in vitro. Conclusions Exo-181b could suppress inflammatory response by promoting M2 polarization via activating PRKCD/AKT signaling pathway, which further promoting osteogenesis in vitro and promote osteointegration in vivo. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01015-y.
Collapse
Affiliation(s)
- Wei Liu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Muyu Yu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Medical Centre of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Key Clinic Centre of Metabolism Disease, Shanghai Institute for Diabetes, Shanghai, China
| | - Feng Chen
- Department of Orthopaedics, Shanghai Fengxian Central Hospital, Branch of the Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 201400, People's Republic of China.,College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Longqing Wang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Cheng Ye
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Qing Chen
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Qi Zhu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Dong Xie
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Mingzhe Shao
- Department of Vascular Surgery, Multidisciplinary Collaboration Group of Diabetic Foot, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Lili Yang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China.
| |
Collapse
|
38
|
Wang D, Hou J, Xia C, Wei C, Zhu Y, Qian W, Qi S, Wu Y, Shi Y, Qin K, Wu L, Yin F, Chen Z, Li W. Multi-element processed pyritum mixed to β-tricalcium phosphate to obtain a 3D-printed porous scaffold: An option for treatment of bone defects. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112326. [PMID: 34474877 DOI: 10.1016/j.msec.2021.112326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/28/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Bone defects remain a challenging problem for doctors and patients in clinical practice. Processed pyritum is a traditional Chinese medicine that is often used to clinically treat bone fractures. It contains mainly Fe, Zn, Cu, Mn, and other elements. In this study, we added the extract of processed pyritum to β-tricalcium phosphate and produced a porous composite TPP (TCP/processed pyritum) scaffold using digital light processing (DLP) 3D printing technology. Scanning electron microscopy (SEM) analysis revealed that TPP scaffolds contained interconnected pore structures. When compared with TCP scaffolds (1.35 ± 0.15 MPa), TPP scaffolds (5.50 ± 0.24 MPa) have stronger mechanical strength and can effectively induce osteoblast proliferation, differentiation, and mineralization in vitro. Meanwhile, the in vivo study showed that the TPP scaffold had better osteogenic capacity than the TCP scaffold. Furthermore, the TPP scaffold had good biosafety after implantation. In summary, the TPP scaffold is a promising biomaterial for the clinical treatment of bone defects.
Collapse
Affiliation(s)
- Dan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Jingxia Hou
- Department of Pharmacy, Yongcheng City People's Hospital, Henan, Yongcheng 476600, PR China
| | - Chenjie Xia
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Chenxu Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Yuan Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Weiwei Qian
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Shuyang Qi
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Yu Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China; Department of Pharmacy, Nantong Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, Nantong 226000, PR China
| | - Yun Shi
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Kunming Qin
- School of Pharmacy, Jiangsu Ocean University, Jiangsu, Lianyungang 222005, PR China
| | - Li Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China
| | - Zhipeng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China.
| | - Weidong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing 210023, PR China; Nanjing University of Chinese Medicine, Jiangsu Key Laboratory of Chinese Medicine Processing, Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Jiangsu, Nanjing 210023, PR China.
| |
Collapse
|
39
|
Xie L, Wang G, Wu Y, Liao Q, Mo S, Ren X, Tong L, Zhang W, Guan M, Pan H, Chu PK, Wang H. Programmed surface on poly(aryl-ether-ether-ketone) initiating immune mediation and fulfilling bone regeneration sequentially. Innovation (N Y) 2021; 2:100148. [PMID: 34557785 PMCID: PMC8454576 DOI: 10.1016/j.xinn.2021.100148] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/26/2021] [Indexed: 12/22/2022] Open
Abstract
The immune responses are involved in every stage after implantation but the reported immune-regulated materials only work at the beginning without fully considering the different phases of bone healing. Here, poly(aryl-ether-ether-ketone) (PEEK) is coated with a programmed surface, which rapidly releases interleukin-10 (IL-10) in the first week and slowly delivers dexamethasone (DEX) up to 4 weeks. Owing to the synergistic effects of IL-10 and DEX, an aptly weak inflammation is triggered within the first week, followed by significant M2 polarization of macrophages and upregulation of the autophagy-related factors. The suitable immunomodulatory activities pave the way for osteogenesis and the steady release of DEX facilitates bone regeneration thereafter. The sequential immune-mediated process is also validated by an 8-week implementation on a rat model. This is the first attempt to construct implants by taking advantage of both immune-mediated modulation and sequential regulation spanning all bone regeneration phases, which provides insights into the fabrication of advanced biomaterials for tissue engineering and immunological therapeutics. A programed surface is designed and fabricated for immune-mediated osteogenesis The degradation of PTMC coating enables a sequential release of IL-10 and DEX Initially, osteoimmunomodulation is achieved by IL-10 and a small amount of DEX Afterwards, sustained release of DEX fosters the peri-implant bone regeneration
Collapse
Affiliation(s)
- Lingxia Xie
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Guomin Wang
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Yuzheng Wu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Qing Liao
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shi Mo
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Xiaoxue Ren
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liping Tong
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wei Zhang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Min Guan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
40
|
Mestres G, Carter SSD, Hailer NP, Diez-Escudero A. A practical guide for evaluating the osteoimmunomodulatory properties of biomaterials. Acta Biomater 2021; 130:115-137. [PMID: 34087437 DOI: 10.1016/j.actbio.2021.05.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022]
Abstract
Biomaterials offer a promising approach to repair bone defects. Whereas traditional studies predominantly focused on optimizing the osteogenic capacity of biomaterials, less focus has been on the immune response elicited by them. However, the immune and skeletal systems extensively interact, a concept which is referred to as 'osteoimmunology'. This realization has fuelled the development of biomaterials with favourable osteoimmunomodulatory (OIM) properties, aiming to modulate the immune response and to support bone regeneration, thereby affecting the success of an implant. Given the plethora of in vitro assays used to evaluate the OIM properties of biomaterials, it may be challenging to select the right methods to produce conclusive results. In this review, we aim to provide a comprehensive and practical guide for researchers interested in studying the OIM properties of biomaterials in vitro. After a concise overview of the concept of osteoimmunology, emphasis is put on the methodologies that are regularly used to evaluate the OIM properties of biomaterials. First, a description of the most commonly used cell types and cell culture media is provided. Second, typical experimental set-ups and their relevant characteristics are discussed. Third, a detailed overview of the generally used methodologies and readouts, including cell type-specific markers and time points of analysis, is given. Finally, we highlight the promise of advanced approaches, namely microarrays, bioreactors and microfluidic-based systems, and the potential that these may offer to the osteoimmunology field. STATEMENT OF SIGNIFICANCE: Osteoimmunology focuses on the connection and communication between the skeletal and immune systems. This interaction has been recognized to play an important role in the clinical success of biomaterials, which has resulted in an increasing amount of research on the osteoimmunomodulatory (OIM) properties of biomaterials. However, the amount of literature makes it challenging to extract the information needed to design experiments from beginning to end, and to compare obtained results to existing work. This article intends to serve as a guide for those aiming to learn more about the commonly used experimental approaches in the field. We cover early-stage choices, such as cell types and experimental set-ups, but also discuss specific assays, including cell markers and time points of analysis.
Collapse
Affiliation(s)
- Gemma Mestres
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden.
| | - Sarah-Sophia D Carter
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Nils P Hailer
- Ortholab, Department of Surgical Sciences-Orthopaedics, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Diez-Escudero
- Ortholab, Department of Surgical Sciences-Orthopaedics, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
41
|
Izumiya M, Haniu M, Ueda K, Ishida H, Ma C, Ideta H, Sobajima A, Ueshiba K, Uemura T, Saito N, Haniu H. Evaluation of MC3T3-E1 Cell Osteogenesis in Different Cell Culture Media. Int J Mol Sci 2021; 22:ijms22147752. [PMID: 34299372 PMCID: PMC8304275 DOI: 10.3390/ijms22147752] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 11/16/2022] Open
Abstract
Many biomaterials have been evaluated using cultured cells. In particular, osteoblast-like cells are often used to evaluate the osteocompatibility, hard-tissue-regeneration, osteoconductive, and osteoinductive characteristics of biomaterials. However, the evaluation of biomaterial osteogenesis-inducing capacity using osteoblast-like cells is not standardized; instead, it is performed under laboratory-specific culture conditions with different culture media. However, the effect of different media conditions on bone formation has not been investigated. Here, we aimed to evaluate the osteogenesis of MC3T3-E1 cells, one of the most commonly used osteoblast-like cell lines for osteogenesis evaluation, and assayed cell proliferation, alkaline phosphatase activity, expression of osteoblast markers, and calcification under varying culture media conditions. Furthermore, the various media conditions were tested in uncoated plates and plates coated with collagen type I and poly-L-lysine, highly biocompatible molecules commonly used as pseudobiomaterials. We found that the type of base medium, the presence or absence of vitamin C, and the freshness of the medium may affect biomaterial regeneration. We posit that an in vitro model that recapitulates in vivo bone formation should be established before evaluating biomaterials.
Collapse
Affiliation(s)
- Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Miyu Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Haruka Ishida
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Chuang Ma
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Hirokazu Ideta
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopaedic Surgery, School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Atsushi Sobajima
- Department of Orthopaedic Surgery, School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopedics (Lower Limbs), Social Medical Care Corporation Hosei-kai Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, Nagano 390-8601, Japan
| | - Koki Ueshiba
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Takeshi Uemura
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Correspondence: ; Tel.: +81-263-37-3555
| |
Collapse
|
42
|
Brassolatti P, Bossini PS, de Andrade ALM, Luna GLF, da Silva JV, Almeida-Lopes L, Napolitano MA, de Avó LRDS, Leal ÂMDO, Anibal FDF. Comparison of two different biomaterials in the bone regeneration (15, 30 and 60 days) of critical defects in rats. Acta Cir Bras 2021; 36:e360605. [PMID: 34287608 PMCID: PMC8291905 DOI: 10.1590/acb360605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
PURPOSE To evaluate and compare two types of different scaffolds in critical bone defects in rats. METHODS Seventy male Wistar rats (280 ± 20 grams) divided into three groups: control group (CG), untreated animals; biomaterial group 1 (BG1), animals that received the scaffold implanted hydroxyapatite (HA)/poly(lactic-co-glycolic) acid (PLGA); and biomaterial group 2 (BG2), animals that received the scaffolds HA/PLGA/Bleed. The critical bone defect was induced in the medial region of the skull calotte with the aid of an 8-mm-diameter trephine drill. The biomaterial was implanted in the form of 1.5 mm thick scaffolds, and samples were collected after 15, 30 and 60 days. Non-parametric Mann-Whitney test was used, with the significance level of 5% (p ≤ 0.05). RESULTS Histology revealed morphological and structural differences of the neoformed tissue between the experimental groups. Collagen-1 (Col-1) findings are consistent with the histological ones, in which BG2 presented the highest amount of fibers in its tissue matrix in all evaluated periods. In contrast, the results of receptor activator of nuclear factor kappa-Β ligand (Rank-L) immunoexpression were higher in BG2 in the periods of 30 and 60 days, indicating an increase of the degradation of the biomaterial and the remodeling activity of the bone. CONCLUSIONS The properties of the HA/PLGA/Bleed scaffold were superior when compared to the scaffold composed only by HA/PLGA.
Collapse
Affiliation(s)
- Patricia Brassolatti
- PhD in Biotechnology. Postgraduate Program in Evolutionary Genetics
and Molecular Biology – Department of Morphology and Pathology – Universidade
Federal de São Carlos – Sao Carlos (SP), Brazil
| | - Paulo Sérgio Bossini
- PhD in Physiotherapy. NUPEN - Research and Education Center in
Health Science and DMC Equipment Import and Export-Co. Ltda – Sao Carlos (SP),
Brazil
| | - Ana Laura Martins de Andrade
- PhD in Physiotherapy. Department of Physiotherapy – Universidade
Federal de São Carlos – Sao Carlos (SP), Brazil
| | - Genoveva Lourdes Flores Luna
- PhD in Biotechnology. Metabolic Endocrine Research Laboratory –
Department of Medicine – Universidade Federal University de São Carlos – Sao Carlos
(SP), Brazil
| | - Juliana Virginio da Silva
- Graduate student in Biotechnology. Institute of Physics of Sao
Carlos– Universidade de São Paulo – Sao Carlos (SP), Brazil
| | - Luciana Almeida-Lopes
- PhD in Science and Materials Engineering. NUPEN - Research and
Education Center in Health Science and DMC Equipment Import and Export-Co. Ltda –
Sao Carlos (SP), Brazil
| | | | | | | | - Fernanda de Freitas Anibal
- Associate Professor. Department of Morphology and Pathology –
Universidade Federal de São Carlos – Sao Carlos (SP), Brazil
| |
Collapse
|
43
|
Zankovic S, Seidenstuecker M, Prall WC, Loos J, Maderer F, Oberle M, Latorre SH, Schilling P, Riedel B, Bernstein A. A Method for the Evaluation of Early Osseointegration of Implant Materials Ex Vivo: Human Bone Organ Model. MATERIALS 2021; 14:ma14113001. [PMID: 34206040 PMCID: PMC8198050 DOI: 10.3390/ma14113001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 01/02/2023]
Abstract
In the present work, an ex vivo organ model using human bone (explant) was developed for the evaluation of the initial osseointegration behavior of implant materials. The model was tested with additive manufactured Ti6Al4V test substrates with different 3D geometries. Explants were obtained from patients who underwent total knee replacement surgery. The tibial plateaus were used within 24 h after surgery to harvest bone cylinders (BC) from the anterior side using hollow burrs. The BCs were brought into contact with the test substrate and inserted into an agarose mold, then covered with cell culture media and subjected to the external load of 500 g. Incubation was performed for 28 days. After 28d the test substrate was removed for further analysis. Cells grown out BC onto substrate were immunostained with DAPI and with an antibody against Collagen-I and alkaline phosphatase (ALP) for visualization and cell counting. We show that cells stayed alive for up to 28d in our organ model. The geometry of test substrates influences the number of cells grown onto substrate from BCs. The model presented here can be used for testing implant materials as an alternative for in vitro tests and animal models.
Collapse
Affiliation(s)
- Sergej Zankovic
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (M.S.); (S.H.L.); (P.S.); (B.R.); (A.B.)
- Correspondence: ; Tel.: +49-761-270-26105
| | - Michael Seidenstuecker
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (M.S.); (S.H.L.); (P.S.); (B.R.); (A.B.)
| | - Wolf C. Prall
- Schoen Clinic Munich Harlaching, Teaching Hospital of Paracelsus Medical University Salzburg, 5026 Salzburg, Austria;
- Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University (LMU), Nussbaumstr. 20, 80336 Munich, Germany
| | - Johannes Loos
- FIT Additive Manufacturing Group, FIT Production GmbH, Am Grohberg 1, 92331 Lupburg, Germany; (J.L.); (F.M.)
| | - Franziska Maderer
- FIT Additive Manufacturing Group, FIT Production GmbH, Am Grohberg 1, 92331 Lupburg, Germany; (J.L.); (F.M.)
| | - Mike Oberle
- RKK Klinikum, St. Josefskrankenhaus, Klinik f Unfallchirurgie, Orthopaedie, Kinder-und Sporttraumatologie, Sautierstr. 1, 79104 Freiburg, Germany;
| | - Sergio H. Latorre
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (M.S.); (S.H.L.); (P.S.); (B.R.); (A.B.)
| | - Pia Schilling
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (M.S.); (S.H.L.); (P.S.); (B.R.); (A.B.)
| | - Bianca Riedel
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (M.S.); (S.H.L.); (P.S.); (B.R.); (A.B.)
| | - Anke Bernstein
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (M.S.); (S.H.L.); (P.S.); (B.R.); (A.B.)
| |
Collapse
|
44
|
The limit of tolerable micromotion for implant osseointegration: a systematic review. Sci Rep 2021; 11:10797. [PMID: 34031476 PMCID: PMC8144379 DOI: 10.1038/s41598-021-90142-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/04/2021] [Indexed: 02/04/2023] Open
Abstract
Much research effort is being invested into the development of porous biomaterials that enhance implant osseointegration. Large micromotions at the bone-implant interface impair this osseointegration process, resulting in fibrous capsule formation and implant loosening. This systematic review compiled all the in vivo evidence available to establish if there is a universal limit of tolerable micromotion for implant osseointegration. The protocol was registered with the International Prospective Register for Systematic Reviews (ID: CRD42020196686). Pubmed, Scopus and Web of Knowledge databases were searched for studies containing terms relating to micromotion and osseointegration. The mean value of micromotion for implants that osseointegrated was 32% of the mean value for those that did not (112 ± 176 µm versus 349 ± 231 µm, p < 0.001). However, there was a large overlap in the data ranges with no universal limit apparent. Rather, many factors were found to combine to affect the overall outcome including loading time, the type of implant and the material being used. The tables provided in this review summarise these factors and will aid investigators in identifying the most relevant micromotion values for their biomaterial and implant development research.
Collapse
|
45
|
Gao J, Su Y, Qin YX. Calcium phosphate coatings enhance biocompatibility and degradation resistance of magnesium alloy: Correlating in vitro and in vivo studies. Bioact Mater 2021; 6:1223-1229. [PMID: 33210020 PMCID: PMC7653207 DOI: 10.1016/j.bioactmat.2020.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 11/24/2022] Open
Abstract
Magnesium (Mg) and its alloys are promising biodegradable materials for orthopedic applications. However, one of the major problems is their rapid degradation rate with quick evolution of hydrogen gas. To overcome this problem, calcium phosphate (CaP) coatings have been used to improve the degradation resistance and the biocompatibility of Mg materials. This study focuses on the comparison and correlation of the in vitro and in vivo degradation and biocompatibility behaviors of these materials. A CaP coating consisting of dicalcium phosphate dihydrate (DCPD) was deposited on an AZ60 Mg alloy by the chemical conversion method. Then, the in vitro degradation testing including electrochemical and immersion tests, and in vivo implantation of the CaP coated Mg alloy were conducted to compare the degradation behaviors. Next, the in vitro cell behavior and in vivo bone tissue response were also compared on both uncoated and CaP-coated Mg samples. Data showed that the CaP coating provided the Mg alloy with significantly better biodegradation behavior and biocompatibility. The in vitro and in vivo biocompatibility tests exhibited good consistency while not the case for biodegradation. Results showed that the in vitro electrochemical test could be a quick screening tool for the biodegradation rate, while the in vitro immersion degradation rate was often 2-4 folds faster than the in vivo degradation rate.
Collapse
Affiliation(s)
- Julia Gao
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, United States
| | - Yingchao Su
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, United States
| | - Yi-Xian Qin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, United States
| |
Collapse
|
46
|
Chen S, Wang Q, Eltit F, Guo Y, Cox M, Wang R. An Ammonia-Induced Calcium Phosphate Nanostructure: A Potential Assay for Studying Osteoporosis and Bone Metastasis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17207-17219. [PMID: 33845570 DOI: 10.1021/acsami.1c00495] [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: 06/12/2023]
Abstract
Osteoclastic resorption of bones plays a central role in both osteoporosis and bone metastasis. A reliable in vitro assay that simulates osteoclastic resorption in vivo would significantly speed up the process of developing effective therapeutic solutions for those diseases. Here, we reported the development of a novel and robust nanostructured calcium phosphate coating with unique functions on the track-etched porous membrane by using an ammonia-induced mineralization (AiM) technique. The calcium phosphate coating uniformly covers one side of the PET membrane, enabling testing for osteoclastic resorption. The track-etched pores in the PET membrane allow calcium phosphate mineral pins to grow inside, which, on the one hand, enhances coating integration with a membrane substrate and, on the other hand, provides diffusion channels for delivering drugs from the lower chamber of a double-chamber cell culture system. The applications of the processed calcium phosphate coating were first demonstrated as a drug screening device by using alendronate, a widely used drug for osteoporosis. It was confirmed that the delivery of alendronate significantly decreased both the number of monocyte-differentiated osteoclasts and coating resorption. To demonstrate the application in studying bone metastasis, we delivered a PC3 prostate cancer-conditioned medium and confirmed that both the differentiation of monocytes into osteoclasts and the osteoclastic resorption of the calcium phosphate coating were significantly enhanced. This novel assay thus provides a new platform for studying osteoclastic activities and assessing drug efficacy in vitro.
Collapse
Affiliation(s)
- Sijia Chen
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Qiong Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Felipe Eltit
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Yubin Guo
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Michael Cox
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Rizhi Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
47
|
Jablonská E, Horkavcová D, Rohanová D, Brauer DS. A review of in vitro cell culture testing methods for bioactive glasses and other biomaterials for hard tissue regeneration. J Mater Chem B 2021; 8:10941-10953. [PMID: 33169773 DOI: 10.1039/d0tb01493a] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bioactive glasses are used to regenerate bone by a mechanism which involves surface degradation, the release of ions such as calcium, soluble silica and phosphate and the precipitation of a biomimetic apatite surface layer on the glass. One major area of bioactive glass research is the incorporation of therapeutically active ions to broaden the application range of these materials. When developing such new compositions, in vitro cell culture studies are a key part of their characterisation. However, parameters of cell culture studies vary widely, and depending on the intended use of bioactive glass compositions, different layouts, cell types and assays need to be used. The aim of this publication is to provide materials scientists, particularly those new to cell culture studies, with a tool for selecting the most appropriate assays to give insight into the properties of interest.
Collapse
Affiliation(s)
- Eva Jablonská
- Laboratory of Molecular Biology and Virology, Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague 6, Czech Republic.
| | - Diana Horkavcová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Dana Rohanová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Delia S Brauer
- Otto Schott Institute of Materials Research, Faculty of Chemistry and Earth Sciences, Friedrich Schiller University Jena, Fraunhoferstr. 6, 07743 Jena, Germany.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW One aim in bone tissue engineering is to develop human cell-based, 3D in vitro bone models to study bone physiology and pathology. Due to the heterogeneity of cells among patients, patient's own cells are needed to be obtained, ideally, from one single cell source. This review attempts to identify the appropriate cell sources for development of such models. RECENT FINDINGS Bone marrow and peripheral blood are considered as suitable sources for extraction of osteoblast/osteocyte and osteoclast progenitor cells. Recent studies on these cell sources have shown no significant differences between isolated progenitor cells. However, various parameters such as medium composition affect the cell's proliferation and differentiation potential which could make the peripheral blood-derived stem cells superior to the ones from bone marrow. Peripheral blood can be considered a suitable source for osteoblast/osteocyte and osteoclast progenitor cells, being less invasive for the patient. However, more investigations are needed focusing on extraction and differentiation of both cell types from the same donor sample of peripheral blood.
Collapse
Affiliation(s)
- Sana Ansari
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
49
|
Negrescu AM, Necula MG, Gebaur A, Golgovici F, Nica C, Curti F, Iovu H, Costache M, Cimpean A. In Vitro Macrophage Immunomodulation by Poly(ε-caprolactone) Based-Coated AZ31 Mg Alloy. Int J Mol Sci 2021; 22:ijms22020909. [PMID: 33477539 PMCID: PMC7831122 DOI: 10.3390/ijms22020909] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Due to its excellent bone-like mechanical properties and non-toxicity, magnesium (Mg) and its alloys have attracted great interest as biomaterials for orthopaedic applications. However, their fast degradation rate in physiological environments leads to an acute inflammatory response, restricting their use as biodegradable metallic implants. Endowing Mg-based biomaterials with immunomodulatory properties can help trigger a desired immune response capable of supporting a favorable healing process. In this study, electrospun poly(ε-caprolactone) (PCL) fibers loaded with coumarin (CM) and/or zinc oxide nanoparticles (ZnO) were used to coat the commercial AZ31 Mg alloy as single and combined formulas, and their effects on the macrophage inflammatory response and osteoclastogenic process were investigated by indirect contact studies. Likewise, the capacity of the analyzed samples to generate reactive oxygen species (ROS) has been investigated. The data obtained by attenuated total reflection Fourier-transform infrared (FTIR-ATR) and X-ray photoelectron spectroscopy (XPS) analyses indicate that AZ31 alloy was perfectly coated with the PCL fibers loaded with CM and ZnO, which had an important influence on tuning the release of the active ingredient. Furthermore, in terms of degradation in phosphate-buffered saline (PBS) solution, the PCL-ZnO- and secondary PCL-CM-ZnO-coated samples exhibited the best corrosion behaviour. The in vitro results showed the PCL-CM-ZnO and, to a lower extent, PCL-ZnO coated sample exhibited the best behaviour in terms of inflammatory response and receptor activator of nuclear factor kappa-B ligand (RANKL)-mediated differentiation of RAW 264.7 macrophages into osteoclasts. Altogether, the results obtained suggest that the coating of Mg alloys with fibrous PCL containing CM and/or ZnO can constitute a feasible strategy for biomedical applications.
Collapse
Affiliation(s)
- Andreea-Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Madalina-Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Adi Gebaur
- Advance Polymer Materials Group, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh. Polizu 17, 011061 Bucharest, Romania; (A.G.); (F.C.); (H.I.)
| | - Florentina Golgovici
- Department of General Chemistry, Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania;
| | - Cristina Nica
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Filis Curti
- Advance Polymer Materials Group, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh. Polizu 17, 011061 Bucharest, Romania; (A.G.); (F.C.); (H.I.)
| | - Horia Iovu
- Advance Polymer Materials Group, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh. Polizu 17, 011061 Bucharest, Romania; (A.G.); (F.C.); (H.I.)
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
- Correspondence: ; Tel.: +40-21-318-1575 (ext. 106)
| |
Collapse
|
50
|
Safina I, Alghazali KM, Childress L, Griffin C, Hashoosh A, Kannarpady G, Watanabe F, Bourdo SE, Dings RPM, Biris AS, Vang KB. Dendritic cell biocompatibility of ether-based urethane films. J Appl Toxicol 2021; 41:1456-1466. [PMID: 33417269 DOI: 10.1002/jat.4136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
The use of synthetic materials for biomedical applications is ever expanding. One of the major requirements for these materials is biocompatibility, which includes prevention of immune system responses. Due to the inherent complexity of their structural composition, the polyurethane (PU) family of polymers is being used in a variety of medical applications, from soft and hard tissue scaffolds to intricate coatings on implantable devices. Herein, we investigated whether two polymer materials, D3 and D7, induced an immune response, measured by their effects on a dendritic cell (DC) line, JAWS II. Using a lactate dehydrogenase cytotoxicity assay and Annexin V/PI staining, we found that the PU materials did not induce cytotoxicity in DC cells. Using confocal microscopy, we also showed that the materials did not induce activation or maturation, as compared to positive controls. This was confirmed by looking at various markers, CD80, CD86, MHC class I, and MHC class II, via flow cytometry. Overall, the results indicated that the investigated PU films are biocompatible in terms of immunotoxicology and immunogenicity and show great promise for use in regenerative medicine.
Collapse
Affiliation(s)
- Ingrid Safina
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Karrer M Alghazali
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Luke Childress
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Christopher Griffin
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Ahmed Hashoosh
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Ganesh Kannarpady
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Fumiya Watanabe
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Shawn E Bourdo
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Ruud P M Dings
- Department of Radiation Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Alexandru S Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Kieng Bao Vang
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| |
Collapse
|