1
|
Cohen DJ, Van Duyn CM, Deng J, Lodi MK, Gallagher MB, Sugar JT, Rawlinson JJ, Ghosh P, Boyan BD, Schwartz Z. Bone Marrow Stromal Cells Generate a Pro-Healing Inflammasome When Cultured on Titanium-Aluminum-Vanadium Surfaces with Microscale/Nanoscale Structural Features. Biomimetics (Basel) 2025; 10:66. [PMID: 39851782 PMCID: PMC11759188 DOI: 10.3390/biomimetics10010066] [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: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The surface topography and chemistry of titanium-aluminum-vanadium (Ti6Al4V) implants play critical roles in the osteoblast differentiation of human bone marrow stromal cells (MSCs) and the creation of an osteogenic microenvironment. To assess the effects of a microscale/nanoscale (MN) topography, this study compared the effects of MN-modified, anodized, and smooth Ti6Al4V surfaces on MSC response, and for the first time, directly contrasted MN-induced osteoblast differentiation with culture on tissue culture polystyrene (TCPS) in osteogenic medium (OM). Surface characterization revealed distinct differences in microroughness, composition, and topography among the Ti6Al4V substrates. MSCs on MN surfaces exhibited enhanced osteoblastic differentiation, evidenced by increased expression of RUNX2, SP7, BGLAP, BMP2, and BMPR1A (fold increases: 3.2, 1.8, 1.4, 1.3, and 1.2). The MN surface also induced a pro-healing inflammasome with upregulation of anti-inflammatory mediators (170-200% increase) and downregulation of pro-inflammatory factors (40-82% reduction). Integrin expression shifted towards osteoblast-associated integrins on MN surfaces. RNA-seq analysis revealed distinct gene expression profiles between MSCs on MN surfaces and those in OM, with only 199 shared genes out of over 1000 differentially expressed genes. Pathway analysis showed that MN surfaces promoted bone formation, maturation, and remodeling through non-canonical Wnt signaling, while OM stimulated endochondral bone development and mineralization via canonical Wnt3a signaling. These findings highlight the importance of Ti6Al4V surface properties in directing MSC differentiation and indicate that MN-modified surfaces act via signaling pathways that differ from OM culture methods, more accurately mimicking peri-implant osteogenesis in vivo.
Collapse
Affiliation(s)
- David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Christine M. Van Duyn
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Jingyao Deng
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Musaddiq K. Lodi
- Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | | | - James T. Sugar
- Medtronic Spine, Memphis, TN 38132, USA; (M.B.G.); (J.T.S.); (J.J.R.)
| | | | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
- Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
2
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
3
|
Ramaraju H, Garcia-Gomez E, McAtee AM, Verga AS, Hollister SJ. Shape memory cycle conditions impact human bone marrow stromal cell binding to RGD- and YIGSR-conjugated poly (glycerol dodecanedioate). Acta Biomater 2024; 186:246-259. [PMID: 39111679 DOI: 10.1016/j.actbio.2024.07.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/21/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024]
Abstract
Bioresorbable shape memory polymers (SMP) are an emerging class of polymers that can help address several challenges associated with minimally invasive surgery by providing a solution for structural tissue repair. Like most synthetic polymer networks, SMPs require additional biorelevance and modification for biomedical applications. Methodologies used to incorporate bioactive ligands must preserve SMP thermomechanics and ensure biofunctionality following in vivo delivery. We have previously described the development of a novel thermoresponsive bioresorbable SMP, poly (glycerol dodecanedioate) (PGD). In this study, cell-adhesive peptide sequences RGD and YIGSR were conjugated with PGD. We investigated 1) the impact of conjugated peptides on the fixity (Rf), recovery (Rr), and recovery rate (dRr/dT), 2) the impact of conjugated peptides on cell binding, and 3) the impact of the shape memory cycle (Tprog) on conjugated peptide functionality towards binding human bone marrow stromal cells (BMSC). Peptide conjugation conditions impact fixity but not the recovery or recovery rate (p < 0.01). Peptide-conjugated substrates increased cell attachment and proliferation compared with controls (p < 0.001). Using complementary integrin binding cell-adhesive peptides increased proliferation compared with using single peptides (p < 0.05). Peptides bound to PGD substrates exhibited specificity to their respective integrin targets. Following the shape memory cycle, peptides maintained functionality and specificity depending on the shape memory cycle conditions (p < 0.001). The dissipation of strain energy during recovery can drive differential arrangement of conjugated sequences impacting functionality, an important design consideration for functionalized SMPs. STATEMENT OF SIGNIFICANCE: Shape memory elastomers are an emerging class of polymers that are well-suited for minimally invasive repair of soft tissues. Tissue engineering approaches commonly utilize biodegradable scaffolds to deliver instructive cues, including cells and bioactive signals. Delivering these instructive cues on biodegradable shape memory elastomers requires modification with bioactive ligands. Furthermore, it is necessary to ensure the specificity of the ligands to their biological targets when conjugated to the polymer. Moreover, the bioactive ligand functionality must be conserved after completing the shape memory cycle, for applications in tissue engineering.
Collapse
Affiliation(s)
- Harsha Ramaraju
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States.
| | - Elisa Garcia-Gomez
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Annabel M McAtee
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Adam S Verga
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Scott J Hollister
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
4
|
Wu S, Zhang Q, Lin D, Al-Shaaobi BA, Sun Y, Si W, Ding X, Ma P, Shen X, Liu J. Near-Infrared Responsive Biomimetic Titanate/TiO 2-X Heterostructure: A Therapeutic Strategy for Combating Implant-Associated Infection and Enhancing Osseointegration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43227-43243. [PMID: 39121390 DOI: 10.1021/acsami.4c06154] [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: 08/11/2024]
Abstract
Implant-associated infections and delayed osseointegration are major challenges for the clinical success of titanium implants. To enhance antibacterial effects and promote early osseointegration, we developed a synergistic photothermal (PTT)/photodynamic (PDT) therapy strategy based on near-infrared (NIR) responsive biomimetic micro/nano titanate/TiO2-X heterostructure coatings (KMNW and NaMNS) in situ constructed on the surface of titanium implants. Specifically, KMNW and NaMNS significantly enhanced photothermal conversion capabilities, achieving localized high temperatures of 48-51 °C and promoting substantial amounts of reactive oxygen species production under 808 nm irradiation. In vitro antibacterial experiments demonstrated that KMNW achieved the highest antibacterial rates against Staphylococcus aureus and Escherichia coli, at 98.78 and 98.33% respectively. Moreover, by mimicking the three-dimensional fibrous network of the extracellular matrix during bone healing, both KMNW and NaMNS markedly promoted the proliferation and osteogenic differentiation of osteoblasts. In vivo implantation studies further confirmed these findings, with KMNW and NaMNS exhibiting superior antibacterial performance under NIR irradiation─94.45% for KMNW and 92.66% for NaMNS. Moreover, KMNW and NaMNS also significantly promoted new bone formation and improved osseointegration in vivo. This study presents a promising PTT/PDT therapeutic strategy for dentistry and orthopedics by employing NIR-responsive biomimetic coatings to combat implant-associated infection and accelerate osseointegration.
Collapse
Affiliation(s)
- Shuyi Wu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Qihong Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Dini Lin
- Science and Education Division, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), Wenzhou 325200, People's Republic of China
| | - Bilal A Al-Shaaobi
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Yingyue Sun
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Wen Si
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Xi Ding
- Department of Stomatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Pingping Ma
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Xinkun Shen
- Science and Education Division, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), Wenzhou 325200, People's Republic of China
| | - Jinsong Liu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| |
Collapse
|
5
|
Matsuura T, Komatsu K, Cheng J, Park G, Ogawa T. Beyond microroughness: novel approaches to navigate osteoblast activity on implant surfaces. Int J Implant Dent 2024; 10:35. [PMID: 38967690 PMCID: PMC11226592 DOI: 10.1186/s40729-024-00554-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
Considering the biological activity of osteoblasts is crucial when devising new approaches to enhance the osseointegration of implant surfaces, as their behavior profoundly influences clinical outcomes. An established inverse correlation exists between osteoblast proliferation and their functional differentiation, which constrains the rapid generation of a significant amount of bone. Examining the surface morphology of implants reveals that roughened titanium surfaces facilitate rapid but thin bone formation, whereas smooth, machined surfaces promote greater volumes of bone formation albeit at a slower pace. Consequently, osteoblasts differentiate faster on roughened surfaces but at the expense of proliferation speed. Moreover, the attachment and initial spreading behavior of osteoblasts are notably compromised on microrough surfaces. This review delves into our current understanding and recent advances in nanonodular texturing, meso-scale texturing, and UV photofunctionalization as potential strategies to address the "biological dilemma" of osteoblast kinetics, aiming to improve the quality and quantity of osseointegration. We discuss how these topographical and physicochemical strategies effectively mitigate and even overcome the dichotomy of osteoblast behavior and the biological challenges posed by microrough surfaces. Indeed, surfaces modified with these strategies exhibit enhanced recruitment, attachment, spread, and proliferation of osteoblasts compared to smooth surfaces, while maintaining or amplifying the inherent advantage of cell differentiation. These technology platforms suggest promising avenues for the development of future implants.
Collapse
Affiliation(s)
- Takanori Matsuura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, 10833 Le Conte Avenue B3-087, Box951668, Los Angeles, CA, 90095-1668, USA
| | - Keiji Komatsu
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, 10833 Le Conte Avenue B3-087, Box951668, Los Angeles, CA, 90095-1668, USA
| | - James Cheng
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, 10833 Le Conte Avenue B3-087, Box951668, Los Angeles, CA, 90095-1668, USA
- Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, USA
| | - Gunwoo Park
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, 10833 Le Conte Avenue B3-087, Box951668, Los Angeles, CA, 90095-1668, USA
| | - Takahiro Ogawa
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, 10833 Le Conte Avenue B3-087, Box951668, Los Angeles, CA, 90095-1668, USA.
- Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, USA.
| |
Collapse
|
6
|
Park G, Matsuura T, Komatsu K, Ogawa T. Optimizing implant osseointegration, soft tissue responses, and bacterial inhibition: A comprehensive narrative review on the multifaceted approach of the UV photofunctionalization of titanium. J Prosthodont Res 2024:JPR_D_24_00086. [PMID: 38853001 DOI: 10.2186/jpr.jpr_d_24_00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Titanium implants have revolutionized restorative and reconstructive therapy, yet achieving optimal osseointegration and ensuring long-term implant success remain persistent challenges. In this review, we explore a cutting-edge approach to enhancing implant properties: ultraviolet (UV) photofunctionalization. By harnessing UV energy, photofunctionalization rejuvenates aging implants, leveraging and often surpassing the intrinsic potential of titanium materials. The primary aim of this narrative review is to offer an updated perspective on the advancements made in the field, providing a comprehensive overview of recent findings and exploring the relationship between UV-induced physicochemical alterations and cellular responses. There is now compelling evidence of significant transformations in titanium surface chemistry induced by photofunctionalization, transitioning from hydrocarbon-rich to carbon pellicle-free surfaces, generating superhydrophilic surfaces, and modulating the electrostatic properties. These changes are closely associated with improved cellular attachment, spreading, proliferation, differentiation, and, ultimately, osseointegration. Additionally, we discuss clinical studies demonstrating the efficacy of UV photofunctionalization in accelerating and enhancing the osseointegration of dental implants. Furthermore, we delve into recent advancements, including the development of one-minute vacuum UV (VUV) photofunctionalization, which addresses the limitations of conventional UV methods as well as the newly discovered functions of photofunctionalization in modulating soft tissue and bacterial interfaces. By elucidating the intricate relationship between surface science and biology, this body of research lays the groundwork for innovative strategies aimed at enhancing the clinical performance of titanium implants, marking a new era in implantology.
Collapse
Affiliation(s)
- Gunwoo Park
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, USA
| | - Takanori Matsuura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, USA
| | - Keiji Komatsu
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, USA
| | - Takahiro Ogawa
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, USA
- Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, USA
| |
Collapse
|
7
|
Berger MB, Bosh K, Deng J, Jacobs TW, Cohen DJ, Boyan BD, Schwartz Z. Wnt16 Increases Bone-to-Implant Contact in an Osteopenic Rat Model by Increasing Proliferation and Regulating the Differentiation of Bone Marrow Stromal Cells. Ann Biomed Eng 2024; 52:1744-1762. [PMID: 38517621 PMCID: PMC11082046 DOI: 10.1007/s10439-024-03488-y] [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: 04/24/2023] [Accepted: 03/07/2024] [Indexed: 03/24/2024]
Abstract
Osseointegration is a complex biological cascade that regulates bone regeneration after implant placement. Implants possessing complex multiscale surface topographies augment this regenerative process through the regulation of bone marrow stromal cells (MSCs) that are in contact with the implant surface. One pathway regulating osteoblastic differentiation is Wnt signaling, and upregulation of non-canonical Wnts increases differentiation of MSCs on these titanium substrates. Wnt16 is a non-canonical Wnt shown to regulate bone morphology in mouse models. This study evaluated the role of Wnt16 during surface-mediated osteoblastic differentiation of MSCs in vitro and osseointegration in vivo. MSCs were cultured on Ti substrates with different surface properties and non-canonical Wnt expression was determined. Subsequently, MSCs were cultured on Ti substrates +/-Wnt16 (100 ng/mL) and anti-Wnt16 antibodies (2 μg/mL). Wnt16 expression was increased in cells grown on microrough surfaces that were processed to be hydrophilic and have nanoscale roughness. However, treatment MSCs on these surfaces with exogenous rhWnt16b increased total DNA content and osteoprotegerin production, but reduced osteoblastic differentiation and production of local factors necessary for osteogenesis. Addition of anti-Wnt16 antibodies blocked the inhibitor effects of Wnt16. The response to Wnt16 was likely independent of other osteogenic pathways like Wnt11-Wnt5a signaling and semaphorin 3a signaling. We used an established rat model of cortical and trabecular femoral bone impairment following botox injections (2 injections of 8 units/leg each, starting and maintenance doses) to assess Wnt16 effects on whole bone morphology and implant osseointegration. Wnt16 injections did not alter whole bone morphology significantly (BV/TV, cortical thickness, restoration of trabecular bone) but were effective at increasing cortical bone-to-implant contact during impaired osseointegration in the botox model. The mechanical quality of the increased bone was not sufficient to rescue the deleterious effects of botox. Clinically, these results are important to understand the interaction of cortical and trabecular bone during implant integration. They suggest a role for Wnt16 in modulating bone remodeling by reducing osteoclastic activity. Targeted strategies to temporally regulate Wnt16 after implant placement could be used to improve osseointegration by increasing the net pool of osteoprogenitor cells.
Collapse
Affiliation(s)
- Michael B Berger
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Kyla Bosh
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Jingyao Deng
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Thomas W Jacobs
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - D Joshua Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA.
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
- Department of Periodontology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| |
Collapse
|
8
|
Kaur G, Pippin JA, Chang S, Redmond J, Chesi A, Wells AD, Maerz T, Grant SFA, Coleman RM, Hankenson KD, Wagley Y. Osteoporosis GWAS-implicated DNM3 locus contextually regulates osteoblastic and chondrogenic fate of mesenchymal stem/progenitor cells through oscillating miR-199a-5p levels. JBMR Plus 2024; 8:ziae051. [PMID: 38686038 PMCID: PMC11056323 DOI: 10.1093/jbmrpl/ziae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Genome wide association study (GWAS)-implicated bone mineral density (BMD) signals have been shown to localize in cis-regulatory regions of distal effector genes using 3D genomic methods. Detailed characterization of such genes can reveal novel causal genes for BMD determination. Here, we elected to characterize the "DNM3" locus on chr1q24, where the long non-coding RNA DNM3OS and the embedded microRNA MIR199A2 (miR-199a-5p) are implicated as effector genes contacted by the region harboring variation in linkage disequilibrium with BMD-associated sentinel single nucleotide polymorphism, rs12041600. During osteoblast differentiation of human mesenchymal stem/progenitor cells (hMSC), miR-199a-5p expression was temporally decreased and correlated with the induction of osteoblastic transcription factors RUNX2 and Osterix. Functional relevance of miR-199a-5p downregulation in osteoblastogenesis was investigated by introducing miR-199a-5p mimic into hMSC. Cells overexpressing miR-199a-5p depicted a cobblestone-like morphological change and failed to produce BMP2-dependent extracellular matrix mineralization. Mechanistically, a miR-199a-5p mimic modified hMSC propagated normal SMAD1/5/9 signaling and expressed osteoblastic transcription factors RUNX2 and Osterix but depicted pronounced upregulation of SOX9 and enhanced expression of essential chondrogenic genes ACAN, COMP, and COL10A1. Mineralization defects, morphological changes, and enhanced chondrogenic gene expression associated with miR-199a-5p mimic over-expression were restored with miR-199a-5p inhibitor suggesting specificity of miR-199a-5p in chondrogenic fate specification. The expression of both the DNM3OS and miR-199a-5p temporally increased and correlated with hMSC chondrogenic differentiation. Although miR-199a-5p overexpression failed to further enhance chondrogenesis, blocking miR-199a-5p activity significantly reduced chondrogenic pellet size, extracellular matrix deposition, and chondrogenic gene expression. Taken together, our results indicate that oscillating miR-199a-5p levels dictate hMSC osteoblast or chondrocyte terminal fate. Our study highlights a functional role of miR-199a-5p as a BMD effector gene at the DNM3 BMD GWAS locus, where patients with cis-regulatory genetic variation which increases miR-199a-5p expression could lead to reduced osteoblast activity.
Collapse
Affiliation(s)
- Gurcharan Kaur
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - James A Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Solomon Chang
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Justin Redmond
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Division of Diabetes and Endocrinology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Kurt D Hankenson
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| |
Collapse
|
9
|
Wu Y, Liu J, Kang L, Tian J, Zhang X, Hu J, Huang Y, Liu F, Wang H, Wu Z. An overview of 3D printed metal implants in orthopedic applications: Present and future perspectives. Heliyon 2023; 9:e17718. [PMID: 37456029 PMCID: PMC10344715 DOI: 10.1016/j.heliyon.2023.e17718] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
With the ability to produce components with complex and precise structures, additive manufacturing or 3D printing techniques are now widely applied in both industry and consumer markets. The emergence of tissue engineering has facilitated the application of 3D printing in the field of biomedical implants. 3D printed implants with proper structural design can not only eliminate the stress shielding effect but also improve in vivo biocompatibility and functionality. By combining medical images derived from technologies such as X-ray scanning, CT, MRI, or ultrasonic scanning, 3D printing can be used to create patient-specific implants with almost the same anatomical structures as the injured tissues. Numerous clinical trials have already been conducted with customized implants. However, the limited availability of raw materials for printing and a lack of guidance from related regulations or laws may impede the development of 3D printing in medical implants. This review provides information on the current state of 3D printing techniques in orthopedic implant applications. The current challenges and future perspectives are also included.
Collapse
Affiliation(s)
- Yuanhao Wu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jieying Liu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lin Kang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jingjing Tian
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xueyi Zhang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jin Hu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yue Huang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Fuze Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hai Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhihong Wu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Bone and Joint Disease, Beijing, China
| |
Collapse
|
10
|
Berger MB, Cohen DJ, Bosh KB, Kapitanov M, Slosar PJ, Levit MM, Gallagher M, Rawlinson JJ, Schwartz Z, Boyan BD. Bone marrow stromal cells generate an osteoinductive microenvironment when cultured on titanium-aluminum-vanadium substrates with biomimetic multiscale surface roughness. Biomed Mater 2023; 18. [PMID: 36827708 PMCID: PMC9993812 DOI: 10.1088/1748-605x/acbf15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/24/2023] [Indexed: 02/26/2023]
Abstract
Osseointegration of titanium-based implants possessing complex macroscale/microscale/mesoscale/nanoscale (multiscale) topographies support a direct and functional connection with native bone tissue by promoting recruitment, attachment and osteoblastic differentiation of bone marrow stromal cells (MSCs). Recent studies show that the MSCs on these surfaces produce factors, including bone morphogenetic protein 2 (BMP2) that can cause MSCs not on the surface to undergo osteoblast differentiation, suggesting they may produce an osteogenic environmentin vivo. This study examined if soluble factors produced by MSCs in contact with titanium-aluminum-vanadium (Ti6Al4V) implants possessing a complex multiscale biomimetic topography are able to induce osteogenesis ectopically. Ti6Al4V disks were grit-blasted and acid-etched to create surfaces possessing macroscale and microscale roughness (MM), micro/meso/nanoscale topography (MN), and macro/micro/meso/nanoscale topography (MMNTM). Polyether-ether-ketone (PEEK) disks were also fabricated by machining to medical-grade specifications. Surface properties were assessed by scanning electron microscopy, contact angle, optical profilometry, and x-ray photoelectron spectroscopy. MSCs were cultured in growth media (GM). Proteins and local factors in their conditioned media (CM) were measured on days 4, 8, 10 and 14: osteocalcin, osteopontin, osteoprotegerin, BMP2, BMP4, and cytokines interleukins 6, 4 and 10 (IL6, IL4, and IL10). CM was collected from D14 MSCs on MMNTMand tissue culture polystyrene (TCPS) and lyophilized. Gel capsules containing active demineralized bone matrix (DBM), heat-inactivated DBM (iDBM), and iDBM + MMN-GM were implanted bilaterally in the gastrocnemius of athymic nude mice (N= 8 capsules/group). Controls included iDBM + GM; iDBM + TCPS-CM from D5 to D10 MSCs; iDBM + MMN-CM from D5 to D10; and iDBM + rhBMP2 (R&D Systems) at a concentration similar to D5-D10 production of MSCs on MMNTMsurfaces. Legs were harvested at 35D. Bone formation was assessed by micro computed tomography and histomorphometry (hematoxylin and eosin staining) with the histology scored according to ASTM 2529-13. DNA was greatest on PEEK at all time points; DNA was lowest on MN at early time points, but increased with time. Cells on PEEK exhibited small changes in differentiation with reduced production of BMP2. Osteoblast differentiation was greatest on the MN and MMNTM, reflecting increased production of BMP2 and BMP4. Pro-regenerative cytokines IL4 and IL10 were increased on Ti-based surfaces; IL6 was reduced compared to PEEK. None of the media from TCPS cultures was osteoinductive. However, MMN-CM exhibited increased bone formation compared to iDBM and iDBM + rhBMP2. Furthermore, exogenous rhBMP2 alone, at the concentration found in MMN-CM collected from D5 to D10 cultures, failed to induce new bone, indicating that other factors in the CM play a critical role in that osteoinductive microenvironment. MSCs cultured on MMNTMTi6Al4V surfaces differentiate and produce an increase in local factors, including BMP2, and the CM from these cultures can induce ectopic bone formation compared to control groups, indicating that the increased bone formation arises from the local response by MSCs to a biomimetic, multiscale surface topography.
Collapse
Affiliation(s)
- Michael B Berger
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America
| | - D Joshua Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America
| | - Kyla B Bosh
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America
| | - Marina Kapitanov
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America
| | - Paul J Slosar
- SpineCare Medical Group, 455 Hickey Blvd., Suite 310, Daly City, CA 94015, United States of America
| | - Michael M Levit
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America
| | - Michelle Gallagher
- Medtronic, Applied Research-Spine, Minneapolis, MN, United States of America
| | - Jeremy J Rawlinson
- Medtronic, Applied Research-Spine, Minneapolis, MN, United States of America
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America.,Department of Periodontology, University of Texas Health Science Center at San Antonio, 7703, Floyd Curl Drive, San Antonio, TX 78229, United States of America
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA 23284, United States of America.,Wallace H. Coulter Department of Biomedical Engineering at the Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, United States of America
| |
Collapse
|
11
|
Nellinger S, Kluger PJ. How Mechanical and Physicochemical Material Characteristics Influence Adipose-Derived Stem Cell Fate. Int J Mol Sci 2023; 24:ijms24043551. [PMID: 36834966 PMCID: PMC9961531 DOI: 10.3390/ijms24043551] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are a subpopulation of mesenchymal stem cells. Compared to bone marrow-derived stem cells, they can be harvested with minimal invasiveness. ASCs can be easily expanded and were shown to be able to differentiate into several clinically relevant cell types. Therefore, this cell type represents a promising component in various tissue engineering and medical approaches (e.g., cell therapy). In vivo cells are surrounded by the extracellular matrix (ECM) that provides a wide range of tissue-specific physical and chemical cues, such as stiffness, topography, and chemical composition. Cells can sense the characteristics of their ECM and respond to them in a specific cellular behavior (e.g., proliferation or differentiation). Thus, in vitro biomaterial properties represent an important tool to control ASCs behavior. In this review, we give an overview of the current research in the mechanosensing of ASCs and current studies investigating the impact of material stiffens, topography, and chemical modification on ASC behavior. Additionally, we outline the use of natural ECM as a biomaterial and its interaction with ASCs regarding cellular behavior.
Collapse
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra Juliane Kluger
- School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-07121-271-2061
| |
Collapse
|
12
|
Levy HA, Karamian BA, Yalla GR, Canseco JA, Vaccaro AR, Kepler CK. Impact of surface roughness and bulk porosity on spinal interbody implants. J Biomed Mater Res B Appl Biomater 2023; 111:478-489. [PMID: 36075112 DOI: 10.1002/jbm.b.35161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 07/19/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022]
Abstract
Spinal fusion surgeries are performed to treat a multitude of cervical and lumbar diseases that lead to pain and disability. Spinal interbody fusion involves inserting a cage between the spinal vertebrae, and is often utilized for indirect neurologic decompression, correction of spinal alignment, anterior column stability, and increased fusion rate. The long-term success of interbody fusion relies on complete osseointegration between the implant surface and vertebral end plates. Titanium (Ti)-based alloys and polyetheretherketone (PEEK) interbody cages represent the most commonly utilized materials and provide sufficient mechanics and biocompatibility to assist in fusion. However, modification to the surface and bulk characteristics of these materials has been shown to maximize osseointegration and long-term stability. Specifically, the introduction of intrinsic porosity and surface roughness has been shown to affect spinal interbody mechanics, vascularization, osteoblast attachment, and ingrowth potential. This narrative review synthesizes the mechanical, in vitro, in vivo, and clinical effects on fusion efficacy associated with introduction of porosity in Ti (neat and alloy) and PEEK intervertebral implants.
Collapse
Affiliation(s)
- Hannah A Levy
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Brian A Karamian
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, USA
| | - Goutham R Yalla
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jose A Canseco
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alexander R Vaccaro
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christopher K Kepler
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Analysis of disordered abrasive scratches on titanium surfaces and their impact on nuclear translocation of yes-associated protein. Sci Rep 2022; 12:21705. [PMID: 36522392 PMCID: PMC9755118 DOI: 10.1038/s41598-022-26203-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The morphology of the metallic surface of an implant is important for its contact with bone tissue as it directly affects osteoblast functions, such as cell adhesion, proliferation, and differentiation. Firm contact between the implant and cells creates a barrier that prevents inflammation and bacterial infections. Therefore, optimizing surface morphology, such as surface roughness adjustments, is essential to improving the adhesion between the implant and cells for successful tissue regeneration. However, the manner in which the cells sense the surface roughness and morphology remains unclear. Previously, we analyzed cell adhesion behavior and observed that inhibited cell spreading can delay osteoblast functions. Therefore, assuming that the surface morphology can be sensed through cell spreading, we investigated the cell spreading area and yes-associated protein (YAP) localization in mouse osteoblasts (MC3T3-E1) on a titanium surface with disordered abrasive scratches. Surface roughness of 100-150 nm was obtained by polishing, which inhibited the cell spreading, indicating that YAP localization in the nucleus was lower than that on other surfaces. The obtained results indicate that the cells sense the surface environment based on their spreading area, which regulates cellular functions via the Hippo pathway.
Collapse
|
14
|
Shirazi S, Ravindran S, Cooper LF. Topography-mediated immunomodulation in osseointegration; Ally or Enemy. Biomaterials 2022; 291:121903. [PMID: 36410109 PMCID: PMC10148651 DOI: 10.1016/j.biomaterials.2022.121903] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Osteoimmunology is at full display during endosseous implant osseointegration. Bone formation, maintenance and resorption at the implant surface is a result of bidirectional and dynamic reciprocal communication between the bone and immune cells that extends beyond the well-defined osteoblast-osteoclast signaling. Implant surface topography informs adherent progenitor and immune cell function and their cross-talk to modulate the process of bone accrual. Integrating titanium surface engineering with the principles of immunology is utilized to harness the power of immune system to improve osseointegration in healthy and diseased microenvironments. This review summarizes current information regarding immune cell-titanium implant surface interactions and places these events in the context of surface-mediated immunomodulation and bone regeneration. A mechanistic approach is directed in demonstrating the central role of osteoimmunology in the process of osseointegration and exploring how regulation of immune cell function at the implant-bone interface may be used in future control of clinical therapies. The process of peri-implant bone loss is also informed by immunomodulation at the implant surface. How surface topography is exploited to prevent osteoclastogenesis is considered herein with respect to peri-implant inflammation, osteoclastic precursor-surface interactions, and the upstream/downstream effects of surface topography on immune and progenitor cell function.
Collapse
Affiliation(s)
- Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA.
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - Lyndon F Cooper
- School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
15
|
Avery D, Morandini L, Sheakley LS, Shah AH, Bui L, Abaricia JO, Olivares-Navarrete R. Canonical Wnt signaling enhances pro-inflammatory response to titanium by macrophages. Biomaterials 2022; 289:121797. [PMID: 36156410 PMCID: PMC10262842 DOI: 10.1016/j.biomaterials.2022.121797] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
Biomaterial characteristics like surface roughness and wettability can determine the phenotype of macrophages following implantation. We have demonstrated that inhibiting Wnt ligand secretion abolishes macrophage polarization in vitro and in vivo; however, the role of canonical Wnt signaling in macrophage activation in response to physical and chemical biomaterial cues is unknown. The aim of this study was to understand whether canonical Wnt signaling affects the response of macrophages to titanium (Ti) surface roughness or wettability in vitro and in vivo. Activating canonical Wnt signaling increased expression of toll-like receptors and interleukin receptors and secreted pro-inflammatory cytokines and reduced anti-inflammatory cytokines on Ti, regardless of surface properties. Inhibiting canonical Wnt signaling reduced pro-inflammatory cytokines on all Ti surfaces and increased anti-inflammatory cytokines on rough or rough-hydrophilic Ti. In vivo, activating canonical Wnt signaling increased total macrophages, pro-inflammatory macrophages, and T cells and decreased anti-inflammatory macrophages on both smooth and rough-hydrophilic implants. Functionally, canonical Wnt activation increases pro-inflammatory macrophage response to cell and cell-extracellular matrix lysates. These results demonstrate that activating canonical Wnt signaling primes macrophages to a pro-inflammatory phenotype that affects their response to Ti implants in vitro and in vivo.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Luke S Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Arth H Shah
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Loc Bui
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Jefferson O Abaricia
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
16
|
Ogura N, Berger MB, Srivas P, Hwang S, Li J, Cohen DJ, Schwartz Z, Boyan BD, Sandhage KH. Tailoring of TiAl6V4 Surface Nanostructure for Enhanced In Vitro Osteoblast Response via Gas/Solid (Non-Line-of-Sight) Oxidation/Reduction Reactions. Biomimetics (Basel) 2022; 7:biomimetics7030117. [PMID: 36134921 PMCID: PMC9496476 DOI: 10.3390/biomimetics7030117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022] Open
Abstract
An aging global population is accelerating the need for better, longer-lasting orthopaedic and dental implants. Additive manufacturing can provide patient-specific, titanium-alloy-based implants with tailored, three-dimensional, bone-like architecture. Studies using two-dimensional substrates have demonstrated that osteoblastic differentiation of bone marrow stromal cells (MSCs) is enhanced on surfaces possessing hierarchical macro/micro/nano-scale roughness that mimics the topography of osteoclast resorption pits on the bone surface. Conventional machined implants with these surfaces exhibit successful osseointegration, but the complex architectures produced by 3D printing make consistent nanoscale surface texturing difficult to achieve, and current line-of-sight methods used to roughen titanium alloy surfaces cannot reach all internal surfaces. Here, we demonstrate a new, non-line-of-sight, gas/solid-reaction-based process capable of generating well-controlled nanotopographies on all open (gas-exposed) surfaces of titanium alloy implants. Dense 3D-printed titanium-aluminum-vanadium (TiAl6V4) substrates were used to evaluate the evolution of surface nanostructure for development of this process. Substrates were either polished to be smooth (for easier evaluation of surface nanostructure evolution) or grit-blasted and acid-etched to present a microrough biomimetic topography. An ultrathin (90 ± 16 nm) conformal, titania-based surface layer was first formed by thermal oxidation (600 °C, 6 h, air). A calciothermic reduction (CaR) reaction (700 °C, 1 h) was then used to convert the surface titania (TiO2) into thin layers of calcia (CaO, 77 ± 16 nm) and titanium (Ti, 51 ± 20 nm). Selective dissolution of the CaO layer (3 M acetic acid, 40 min) then yielded a thin nanoporous/nanorough Ti-based surface layer. The changes in surface nanostructure/chemistry after each step were confirmed by scanning and transmission electron microscopies with energy-dispersive X-ray analysis, X-ray diffraction, selected area electron diffraction, atomic force microscopy, and mass change analyses. In vitro studies indicated that human MSCs on CaR-modified microrough surfaces exhibited increased protein expression associated with osteoblast differentiation and promoted osteogenesis compared to unmodified microrough surfaces (increases of 387% in osteopontin, 210% in osteocalcin, 282% in bone morphogenic protein 2, 150% in bone morphogenic protein 4, 265% in osteoprotegerin, and 191% in vascular endothelial growth factor). This work suggests that this CaR-based technique can provide biomimetic topography on all biologically facing surfaces of complex, porous, additively manufactured TiAl6V4 implants.
Collapse
Affiliation(s)
- Naotaka Ogura
- School of Materials Engineering, Purdue University, W. Lafayette, IN 47907, USA
| | - Michael B. Berger
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Pavan Srivas
- School of Materials Engineering, Purdue University, W. Lafayette, IN 47907, USA
| | - Sunghwan Hwang
- School of Materials Engineering, Purdue University, W. Lafayette, IN 47907, USA
| | - Jiaqi Li
- School of Materials Engineering, Purdue University, W. Lafayette, IN 47907, USA
| | - David Joshua Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
- Correspondence: (Z.S.); (B.D.B.); (K.H.S.)
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
- Correspondence: (Z.S.); (B.D.B.); (K.H.S.)
| | - Kenneth H. Sandhage
- School of Materials Engineering, Purdue University, W. Lafayette, IN 47907, USA
- Correspondence: (Z.S.); (B.D.B.); (K.H.S.)
| |
Collapse
|
17
|
Chan JL, Bae HW, Harrison Farber S, Uribe JS, Eastlack RK, Walker CT. Evolution of Bioactive Implants in Lateral Interbody Fusion. Int J Spine Surg 2022; 16:S61-S68. [PMID: 35387890 PMCID: PMC9983556 DOI: 10.14444/8237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lateral lumbar interbody fusion (LLIF) is an advantageous approach for spinal arthrodesis for a wide range of spinal disorders including degenerative, genetic, and traumatic conditions. LLIF techniques have evolved over the past 15 years regarding surgical approach, with concomitant improvements in implant material design. Bioactive materials have been a focus in the development of novel methods, which reduce the risk of subsidence and pseudarthrosis. Historically, polyetheretherketone and titanium cages have been selected for their advantageous biomechanical properties; however, both have their limitations, regarding optimal modulus or osseointegrative properties. Recent modifications to these 2 materials have focused on devising bioactive implants, which may enhance the rate of bony fusion in spinal arthrodesis by addressing the shortcomings of each. Specific emphasis has been placed on developing improvements in surface coating, porosity, microroughness, and nanotopography of interbody cages. This has been coupled with advances in additive manufacturing to generate cages with ideal biomechanical properties. Three-dimensional-printed titanium cages may be particularly beneficial in spinal arthrodesis during LLIF and reduce the historical rates of subsidence and pseudarthrosis by combining a number of these putatively beneficial biomaterial properties.
Collapse
Affiliation(s)
- Julie L. Chan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hyun W. Bae
- Department of Orthopedic Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - S. Harrison Farber
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Juan S. Uribe
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | - Corey T. Walker
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
18
|
Kaniowska D, Wenk K, Rademacher P, Weiss R, Fabian C, Schulz I, Guthardt M, Lange F, Greiser S, Schmidt M, Braumann UD, Emmrich F, Koehl U, Jaimes Y. Extracellular Vesicles of Mesenchymal Stromal Cells Can be Taken Up by Microglial Cells and Partially Prevent the Stimulation Induced by β-amyloid. Stem Cell Rev Rep 2022; 18:1113-1126. [PMID: 35080744 PMCID: PMC8942956 DOI: 10.1007/s12015-021-10261-4] [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] [Accepted: 09/07/2021] [Indexed: 01/22/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) have great capacity for immune regulation. MSCs provide protective paracrine effects, which are partially exerted by extracellular vesicles (EVs). It has been reported that MSCs-derived EVs (MSC-EVs) contain soluble factors, such as cytokines, chemokines, growth factors and even microRNAs, which confer them similar anti-inflammatory and regenerative effects to MSCs. Moreover, MSCs modulate microglia activation through a dual mechanism of action that relies both on cell contact and secreted factors. Microglia cells are the central nervous system immune cells and the main mediators of the inflammation leading to neurodegenerative disorders. Here, we investigated whether MSC-EVs affect the activation of microglia cells by β-amyloid aggregates. We show that the presence of MSC-EVs can prevent the upregulation of pro-inflammatory mediators such as tumor necrosis factor (TNF)-α and nitric oxide (NO). Both are up-regulated in neurodegenerative diseases representing chronic inflammation, as in Alzheimer’s disease. We demonstrate that MSC-EVs are internalized by the microglia cells. Further, our study supports the use of MSC-EVs as a promising therapeutic tool to treat neuroinflammatory diseases. Significance Statement It has been reported that mesenchymal stromal/stem cells and MSC-derived small extracellular vesicles have therapeutic effects in the treatment of various degenerative and inflammatory diseases. Extracellular vesicles are loaded with proteins, lipids and RNA and act as intercellular communication mediators. Here we show that extracellular vesicles can be taken up by murine microglial cells. In addition, they partially reduce the activation of microglial cells against β-amyloid aggregates. This inhibition of microglia activation may present an effective strategy for the control/therapy of neurodegenerative diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Dorota Kaniowska
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany. .,Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany.
| | - Kerstin Wenk
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Phil Rademacher
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Ronald Weiss
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Claire Fabian
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany
| | - Isabell Schulz
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany
| | - Max Guthardt
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany
| | - Franziska Lange
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany
| | - Sebastian Greiser
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany
| | - Matthias Schmidt
- Department of Isotope Biogeochemistry, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Ulf-Dietrich Braumann
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany.,Faculty of Engineering, Leipzig University of Applied Sciences (HTWK), Leipzig, Germany.,Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Frank Emmrich
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany.,Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany.,Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany.,Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Yarúa Jaimes
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstrasse 1, 04103, Leipzig, Germany.,Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany.,Fraunhofer Cluster of Excellence for Immune-mediated Diseases CIMD, Frankfurt, Germany
| |
Collapse
|
19
|
Stich T, Alagboso F, Křenek T, Kovářík T, Alt V, Docheva D. Implant-bone-interface: Reviewing the impact of titanium surface modifications on osteogenic processes in vitro and in vivo. Bioeng Transl Med 2022; 7:e10239. [PMID: 35079626 PMCID: PMC8780039 DOI: 10.1002/btm2.10239] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/08/2021] [Accepted: 06/13/2021] [Indexed: 12/12/2022] Open
Abstract
Titanium is commonly and successfully used in dental and orthopedic implants. However, patients still have to face the risk of implant failure due to various reasons, such as implant loosening or infection. The risk of implant loosening can be countered by optimizing the osteointegration capacity of implant materials. Implant surface modifications for structuring, roughening and biological activation in favor for osteogenic differentiation have been vastly studied. A key factor for a successful stable long-term integration is the initial cellular response to the implant material. Hence, cell-material interactions, which are dependent on the surface parameters, need to be considered in the implant design. Therefore, this review starts with an introduction to the basics of cell-material interactions as well as common surface modification techniques. Afterwards, recent research on the impact of osteogenic processes in vitro and vivo provoked by various surface modifications is reviewed and discussed, in order to give an update on currently applied and developing implant modification techniques for enhancing osteointegration.
Collapse
Affiliation(s)
- Theresia Stich
- Experimental Trauma Surgery, Department of Trauma SurgeryUniversity Regensburg Medical CentreRegensburgGermany
| | - Francisca Alagboso
- Experimental Trauma Surgery, Department of Trauma SurgeryUniversity Regensburg Medical CentreRegensburgGermany
| | - Tomáš Křenek
- New Technologies Research CentreUniversity of West BohemiaPilsenCzech Republic
| | - Tomáš Kovářík
- New Technologies Research CentreUniversity of West BohemiaPilsenCzech Republic
| | - Volker Alt
- Experimental Trauma Surgery, Department of Trauma SurgeryUniversity Regensburg Medical CentreRegensburgGermany
- Clinic and Polyclinic for Trauma Surgery, University Regensburg Medical CentreRegensburgGermany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma SurgeryUniversity Regensburg Medical CentreRegensburgGermany
| |
Collapse
|
20
|
Toop N, Gifford C, Motiei-Langroudi R, Farzadi A, Boulter D, Forghani R, Farhadi HF. Can activated titanium interbody cages accelerate or enhance spinal fusion? a review of the literature and a design for clinical trials. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 33:1. [PMID: 34921610 PMCID: PMC8684547 DOI: 10.1007/s10856-021-06628-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 06/14/2023]
Abstract
While spinal interbody cage options have proliferated in the past decade, relatively little work has been done to explore the comparative potential of biomaterial technologies in promoting stable fusion. Innovations such as micro-etching and nano-architectural designs have shown purported benefits in in vitro studies, but lack clinical data describing their optimal implementation. Here, we critically assess the pre-clinical data supportive of various commercially available interbody cage biomaterial, topographical, and structural designs. We describe in detail the osteointegrative and osteoconductive benefits conferred by these modifications with a focus on polyetheretherketone (PEEK) and titanium (Ti) interbody implants. Further, we describe the rationale and design for two randomized controlled trials, which aim to address the paucity of clinical data available by comparing interbody fusion outcomes between either PEEK or activated Ti lumbar interbody cages. Utilizing dual-energy computed tomography (DECT), these studies will evaluate the relative implant-bone integration and fusion rates achieved by either micro-etched Ti or standard PEEK interbody devices. Taken together, greater understanding of the relative osseointegration profile at the implant-bone interface of cages with distinct topographies will be crucial in guiding the rational design of further studies and innovations.
Collapse
Affiliation(s)
- Nathaniel Toop
- Departments of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Connor Gifford
- Departments of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Arghavan Farzadi
- Departments of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel Boulter
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Reza Forghani
- Department of Radiology, McGill University, Montreal, QC, Canada
| | - H Francis Farhadi
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
21
|
Abaricia JO, Whitehead AJ, Kandalam S, Shah AH, Hotchkiss KM, Morandini L, Olivares-Navarrete R. E-cigarette Aerosol Mixtures Inhibit Biomaterial-Induced Osseointegrative Cell Phenotypes. MATERIALIA 2021; 20:101241. [PMID: 34778733 PMCID: PMC8589285 DOI: 10.1016/j.mtla.2021.101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
OBJECTIVES Smoking is a known contributor to the failure of dental implants. Despite a decline in cigarette use, the popularity of e-cigarettes has exploded. However, little is known about how e-cigarettes affect the biologic response to implants. This study examines the effect of e-cigarette aerosol mixtures (ecig-AM) on macrophage activation and osteoblastogenesis of mesenchymal stem cells (MSCs) in response to titanium (Ti) implant surfaces. METHODS Ecig-AMs were prepared by bubbling aerosol through PBS. Human-derived MSCs or murine-derived macrophages were plated on smooth, rough-hydrophobic, or rough-hydrophilic Ti surfaces in media supplemented with ecig-AM. In macrophages, expression of inflammatory markers was measured by qPCR and macrophage immunophenotype characterized by flow cytometry after 24 hours of exposure. In MSCs, expression of osteogenic markers and inflammatory cytokines was measured by qPCR and ELISA, while alkaline phosphatase activity (ALP) was determined by colorimetric assay. RESULTS Ecig-AM polarized primary macrophages into a pro-inflammatory state with higher effect on ecig-AM with flavorants and nicotine. Metabolic activity of MSCs decreased in a concentration dependent fashion and was stronger in ecig-AM containing nicotine. MSCs reduced expression of osteogenic markers in response to ecig-AM, but increased RANKL secretion, particularly at the highest ecig-AM concentrations. The effect of ecig-AM exposure was lessened when macrophages or MSCs were cultured on rough-hydrophilic substrates. SIGNIFICANCE Ecig-AM activated macrophages into a pro-inflammatory phenotype and impaired MSC-to-osteoblast differentiation in response to Ti implant surfaces. These effects were potentiated by flavorants and nicotine, suggesting that e-cigarette use may compromise the osseointegration of dental implants.
Collapse
Affiliation(s)
| | | | - Suraj Kandalam
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Arth H. Shah
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Kelly M Hotchkiss
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Lais Morandini
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
22
|
Abar B, Kelly C, Pham A, Allen N, Barber H, Kelly A, Mirando AJ, Hilton MJ, Gall K, Adams SB. Effect of surface topography on in vitro osteoblast function and mechanical performance of 3D printed titanium. J Biomed Mater Res A 2021; 109:1792-1802. [PMID: 33754494 PMCID: PMC8373644 DOI: 10.1002/jbm.a.37172] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 02/13/2021] [Accepted: 03/09/2021] [Indexed: 12/18/2022]
Abstract
Critical-sized defects remain a significant challenge in orthopaedics. 3D printed scaffolds are a promising treatment but are still limited due to inconsistent osseous integration. The goal of the study is to understand how changing the surface roughness of 3D printed titanium either by surface treatment or artificially printing rough topography impacts the mechanical and biological properties of 3D printed titanium. Titanium tensile samples and discs were printed via laser powder bed fusion. Roughness was manipulated by post-processing printed samples or by directly printing rough features. Experimental groups in order of increasing surface roughness were Polished, Blasted, As Built, Sprouts, and Rough Sprouts. Tensile behavior of samples showed reduced strength with increasing surface roughness. MC3T3 pre-osteoblasts were seeded on discs and analyzed for cellular proliferation, differentiation, and matrix deposition at 0, 2, and 4 weeks. Printing roughness diminished mechanical properties such as tensile strength and ductility without clear benefit to cell growth. Roughness features were printed on mesoscale, unlike samples in literature in which roughness on microscale demonstrated an increase in cell activity. The data suggest that printing artificial roughness on titanium scaffold is not an effective strategy to promote osseous integration.
Collapse
Affiliation(s)
- Bijan Abar
- Duke University Department of Mechanical Engineering and Material Sciences
| | - Cambre Kelly
- Duke University Department of Mechanical Engineering and Material Sciences
| | - Anh Pham
- Duke University Department of Mechanical Engineering and Material Sciences
| | | | | | - Alexander Kelly
- Duke University Department of Mechanical Engineering and Material Sciences
| | | | | | - Ken Gall
- Duke University Department of Mechanical Engineering and Material Sciences
| | | |
Collapse
|
23
|
Wang L, Zheng F, Song R, Zhuang L, Yang M, Suo J, Li L. Integrins in the Regulation of Mesenchymal Stem Cell Differentiation by Mechanical Signals. Stem Cell Rev Rep 2021; 18:126-141. [PMID: 34536203 DOI: 10.1007/s12015-021-10260-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Mesenchymal stem cells (MSCs) can sense and convert mechanical stimuli signals into a chemical response. Integrins are involved in the mechanotransduction from inside to outside and from outside to inside, and ultimately affect the fate of MSCs responding to different mechanical signals. Different integrins participate in different signaling pathways to regulate MSCs multi-differentiation. In this review, we summarize the latest advances in the effects of mechanical signals on the differentiation of MSCs, the importance of integrins in mechanotransduction, the relationship between integrin heterodimers and different mechanical signals, and the interaction among mechanical signals. We put forward our views on the prospect and challenges of developing mechanical biology in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastrointestinal Surgery, Jilin University First Hospital, Jilin University, 130021, Changchun, People's Republic of China
| | - Fuwen Zheng
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Ruixue Song
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Lequan Zhuang
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Ming Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, 130021, Changchun, People's Republic of China.
| | - Jian Suo
- Department of Gastrointestinal Surgery, Jilin University First Hospital, Jilin University, 130021, Changchun, People's Republic of China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 130021, Changchun, People's Republic of China.
| |
Collapse
|
24
|
Berger MB, Bosh KB, Jacobs TW, Cohen DJ, Schwartz Z, Boyan BD. Growth factors produced by bone marrow stromal cells on nanoroughened titanium-aluminum-vanadium surfaces program distal MSCs into osteoblasts via BMP2 signaling. J Orthop Res 2021; 39:1908-1920. [PMID: 33002223 PMCID: PMC8012402 DOI: 10.1002/jor.24869] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 02/04/2023]
Abstract
Statement of Clinical Significance: There remains the need to develop materials and surfaces that can increase the rate of implant osseointegration. Though osteoanabolic agents, like bone morphogenetic protein (BMP), can provide signaling for osteogenesis, the appropriate design of implants can also produce an innate cellular response that may reduce or eliminate the need to use additional agents to stimulate bone formation. Studies show that titanium implant surfaces that mimic the physical properties of osteoclast resorption pits regulate cellular responses of bone marrow stromal cells (MSCs) by altering cell morphology, transcriptomes, and local factor production to increase their differentiation into osteoblasts without osteogenic media supplements required for differentiation of MSCs on tissue culture polystyrene (TCPS). The goal of this study was to determine how cells in contact with biomimetic implant surfaces regulate the microenvironment around these surfaces in vitro. Two different approaches were used. First, unidirectional signaling was assessed by treating human MSCs grown on TCPS with conditioned media from MSC cultures grown on Ti6Al4V biomimetic surfaces. In the second set of studies, bidirectional signaling was assessed by coculturing MSCs grown on mesh inserts that were placed into culture wells in which MSCs were grown on the biomimetic Ti6Al4V substrates. The results show that biomimetic Ti6Al4V surface properties induce MSCs to produce factors within 7 days of culture that stimulate MSCs not in contact with the surface to exhibit an osteoblast phenotype via endogenous BMP2 acting in a paracrine signaling manner.
Collapse
Affiliation(s)
- Michael B. Berger
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Kyla B. Bosh
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Thomas W. Jacobs
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - D. Joshua Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA;,Department of Periodontology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA;,Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
25
|
Ge J, Wang F, Xu Z, Shen X, Gao C, Wang D, Hu G, Gu J, Tang T, Wei J. Influences of niobium pentoxide on roughness, hydrophilicity, surface energy and protein absorption, and cellular responses to PEEK based composites for orthopedic applications. J Mater Chem B 2021; 8:2618-2626. [PMID: 32129420 DOI: 10.1039/c9tb02456e] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To improve the bio-performances of polyetheretherketone (PEEK) for orthopedic applications, submicro-particles of niobium pentoxide (Nb2O5) were synthesized using a sol-gel method, and PEEK/Nb2O5 composites (PNC) with a Nb2O5 content of 25v% (PNC25) and 50v% (PNC50) were fabricated by utilizing a process of pressing-sintering. The results showed that the Nb2O5 particles were not only dispersed in the composites but also exposed on the surface of the composites, which formed submicro-structural surfaces. In addition, the hydrophilicity, surface energy, surface roughness and absorption of proteins of the composites were improved with increasing Nb2O5 content. Moreover, the release of Nb ions with the highest concentration of 5.01 × 10-6 mol L-1 from the composite into the medium displayed no adverse effects on cell proliferation and morphology, indicating no cytotoxicity. Furthermore, compared with PEEK, the composites, especially PNC50, obviously stimulated adhesion and proliferation as well as osteogenic differentiation of bone mesenchymal stem cells of rats. The results suggested that the incorporation of Nb2O5 submicro-particles into PEEK produced novel bioactive composites with improved surface properties, which played important roles in regulating cell behaviors. In conclusion, the composites, especially PNC50 with good cytocompatibility and promotion of cellular responses, exhibited great potential as implantable materials for bone repair.
Collapse
Affiliation(s)
- Junpeng Ge
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | - Fan Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | - Zhiyan Xu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | - Xuening Shen
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | - Chao Gao
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | - Dongliang Wang
- Department of Orthopedic Surgery, Xin-Hua Hospital, Shanghai Jiao-Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Gangfeng Hu
- The First People's Hospital of Xiaoshan District, 199 Shixinnan Road, Hangzhou 311200, Zhejiang, China
| | - Jinlou Gu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200011, China
| | - Jie Wei
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
26
|
Berger MB, Bosh KB, Cohen DJ, Boyan BD, Schwartz Z. Benchtop plasma treatment of titanium surfaces enhances cell response. Dent Mater 2021; 37:690-700. [PMID: 33589272 PMCID: PMC7981249 DOI: 10.1016/j.dental.2021.01.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/20/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Modifications to implant surface properties, including topography, chemistry, and wettability, alter immune response, osteoblast differentiation of bone marrow stromal cells (MSCs), and implant integration in vivo. Dielectric barrier discharge (DBD) plasma treatment has been used to sterilize surfaces and remove adsorbed carbon, improving wettability. However, unless it is used immediately prior to placement, ambient atmospheric hydrocarbons rapidly adhere to the surface, thereby reducing its hydrophilicity. Moreover, this method is not practical in many clinical settings. The aim of this study was to evaluate the effectiveness of an on-site benchtop modification technique for implants at time of placement, consisting of a DBD plasma that is used to sterilize implants that are pre-packaged in a vacuum. Effects of the plasma-treatment on implant surface properties and cellular response of MSCs and osteoblasts were assessed in vitro. METHODS Titanium-aluminum-vanadium implant surfaces were grit-blasted (GB) or grit-blasted and acid-etched (AE), and packaged under vacuum. AE surfaces were also plasma-treated using the benchtop device (GB + AE) and then removed from the vacuum. GB surface morphology was altered with AE but AE microroughness was not changed with the plasma-treatment. Plasma-treatment increased the surface wettability, but did not alter surface atomic concentrations of titanium, oxygen, or carbon. RESULTS MSCs and osteoblast-like cells (MG63 s) produced increased concentrations of osteocalcin, osteopontin, and osteoprotegerin after plasma-treatment of AE surfaces compared to non-plasma-treated AE surfaces; production of IL6 was reduced and IL10 was. Aging GB + AE surfaces for 7 days after plasma-treatment but still in the vacuum environment reduced the effectiveness of plasma on cellular response. SIGNIFICANCE Overall, these data suggest that application of benchtop plasma at the time of implant placement can alter the surface free energy of an implant surface without modifying surface chemical composition and enhance the differentiation and activity of MSCs and osteoblasts that are in contact with these implant surfaces.
Collapse
Affiliation(s)
- Michael B Berger
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| | - Kyla B Bosh
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| | - D Joshua Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| | - Barbara D Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA; Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA; Department of Periodontology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
27
|
Rousseau N, Msolli I, Chabrand P, Destainville A, Richart O, Milan JL. Local tissue effects and peri-implant bone healing induced by implant surface treatment: an in vivo study in the sheep. J Periodontal Res 2021; 56:789-803. [PMID: 33788298 DOI: 10.1111/jre.12878] [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: 09/02/2020] [Revised: 02/16/2021] [Accepted: 03/03/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The aim of this study was to assess, through biological analysis, the local effects and osseointegration of dental implants incorporating surface micro/nanofeatures compared with implants of identical design without surface treatment. BACKGROUND Known to impact bone cell behavior, surface chemical and topography modifications target improved osseointegration and long-term success of dental implants. Very few studies assess the performance of implants presenting both micro- and nanofeatures in vivo on the animal models used in preclinical studies for medical device certification. METHODS Implant surfaces were characterized in terms of topography and surface chemical composition. After 4 weeks and 13 weeks of implantation in sheep femoral condyles, forty implants were evaluated through micro-computed tomography, histopathologic, and histomorphometric analyses. RESULTS No local adverse effects were observed around implants. Histomorphometric analyses showed significantly higher bone-to-implant contact in the coronal region of the surface-treated implant at week 4 and week 13, respectively, was 79.3 ± 11.2% and 86.4 ± 6.7%, compared with the untreated implants (68.3 ± 8.8% and 74.8 ± 13%). Micro-computed tomography analyses revealed that healing patterns differed between coronal and apical regions, with higher coronal bone-to-implant contact at week 13. Histopathologic results showed, at week 13, bone healing around the surface-treated implant with undistinguishable defect margins, while the untreated implant still presented bone condensation and traces of the initial drill defect. CONCLUSION Our results suggest that the surface-treated implant not only shows no deleterious effects on local tissues but also promotes faster bone healing around the implant.
Collapse
Affiliation(s)
- Nicolas Rousseau
- CNRS, ISM, Aix Marseille Univ, Marseille, France.,Selenium Medical, La Rochelle, France
| | | | | | | | | | | |
Collapse
|
28
|
Wang Q, Wu M, Xu X, Ding C, Luo J, Li J. Direct Current Stimulation for Improved Osteogenesis of MC3T3 Cells Using Mineralized Conductive Polyaniline. ACS Biomater Sci Eng 2021; 7:852-861. [PMID: 33715374 DOI: 10.1021/acsbiomaterials.9b01821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hydroxyapatites (HAPs) are usually coated on the surface of an implant to improve the osseointegration with defect bone tissue. Besides, conducting polymers have the advantages of good conductivity, reasonable biocompatibility, and easy of modification, which endow them applicable to electrical stimulation therapy. However, it still remains a great challenge to fabricate hybrid coating combing HAP with conducting polymer on implant surface efficiently. In this work, phytic acid-doped polyaniline (PANI) were successfully synthesized on medical titanium (Ti) sheets. By virtue of the abundant anodic phosphoric groups of phytic acid, HAP nanocrystals were biomineralized on PANI. The PANI-HAP hybrid layer exhibits good cell compatibility with MC3T3 cells. More importantly, HAP nanocrystals and PANI operate synergistically on cell proliferation and osteogenesis through electrical stimulation. Alkaline phosphatase activity and extracellular calcium contents of cells on PANI-HAP display 3-fold and 2.6-fold increases, compared with bare Ti sheets, respectively. The valid integration of mineralization and electrical stimulation in this work renders an efficient strategy for implant coating, which might have potential applications in bone-related defects.
Collapse
Affiliation(s)
- Quanxin Wang
- College of Polymer Science and Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China.,College of Chemistry and Environmental Protection Engineering, Southwest University for Nationalities, No. 16 South Section 4, Yihuan Road, Chengdu 610041, China
| | - Mingzhen Wu
- College of Polymer Science and Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Xiaoyang Xu
- College of Polymer Science and Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Chunmei Ding
- College of Polymer Science and Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Jianbin Luo
- College of Chemistry and Environmental Protection Engineering, Southwest University for Nationalities, No. 16 South Section 4, Yihuan Road, Chengdu 610041, China
| | - Jianshu Li
- College of Polymer Science and Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| |
Collapse
|
29
|
Molecular Mechanisms of Topography Sensing by Osteoblasts: An Update. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11041791] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Bone is a specialized tissue formed by different cell types and a multiscale, complex mineralized matrix. The architecture and the surface chemistry of this microenvironment can be factors of considerable influence on cell biology, and can affect cell proliferation, commitment to differentiation, gene expression, matrix production and/or composition. It has been shown that osteoblasts encounter natural motifs in vivo, with various topographies (shapes, sizes, organization), and that cell cultures on flat surfaces do not reflect the total potential of the tissue. Therefore, studies investigating the role of topographies on cell behavior are important in order to better understand the interaction between cells and surfaces, to improve osseointegration processes in vivo between tissues and biomaterials, and to find a better topographic surface to enhance bone repair. In this review, we evaluate the main available data about surface topographies, techniques for topographies’ production, mechanical signal transduction from surfaces to cells and the impact of cell–surface interactions on osteoblasts or preosteoblasts’ behavior.
Collapse
|
30
|
Luo Y, Pan H, Jiang J, Zhao C, Zhang J, Chen P, Lin X, Fan S. Desktop-Stereolithography 3D Printing of a Polyporous Extracellular Matrix Bioink for Bone Defect Regeneration. Front Bioeng Biotechnol 2020; 8:589094. [PMID: 33240866 PMCID: PMC7677189 DOI: 10.3389/fbioe.2020.589094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/19/2020] [Indexed: 12/28/2022] Open
Abstract
Introduction Decellularized tendon extracellular matrix (tECM) perfectly provides the natural environment and holds great potential for bone regeneration in Bone tissue engineering (BTE) area. However, its densifying fiber structure leads to reduced cell permeability. Our study aimed to combine tECM with polyethylene glycol diacrylate (PEGDA) to form a biological scaffold with appropriate porosity and strength using stereolithography (SLA) technology for bone defect repair. Methods The tECM was produced and evaluated. Mesenchymal stem cell (MSC) was used to evaluate the biocompatibility of PEGDA/tECM bioink in vitro. Mineralization ability of the bioink was also evaluated in vitro. After preparing 3D printed polyporous PEGDA/tECM scaffolds (3D-pPES) via SLA, the calvarial defect generation capacity of 3D-pPES was assessed. Results The tECM was obtained and the decellularized effect was confirmed. The tECM increased the swelling ratio and porosity of PEGDA bioink, both cellular proliferation and biomineralization in vitro of the bioink were significantly optimized. The 3D-pPES was fabricated. Compared to the control group, increased cell migration efficiency, up-regulation of osteogenic differentiation RNA level, and better calvarial defect repair in rat of the 3D-pPES group were observed. Conclusion This study demonstrates that the 3D-pPES may be a promising strategy for bone defect treatment.
Collapse
Affiliation(s)
- Yunxiang Luo
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hao Pan
- Department of Orthopaedic, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiuzhou Jiang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chenchen Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jianfeng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
31
|
A multifaceted biomimetic interface to improve the longevity of orthopedic implants. Acta Biomater 2020; 110:266-279. [PMID: 32344174 DOI: 10.1016/j.actbio.2020.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/23/2020] [Accepted: 04/09/2020] [Indexed: 01/22/2023]
Abstract
The rise of additive manufacturing has provided a paradigm shift in the fabrication of precise, patient-specific implants that replicate the physical properties of native bone. However, eliciting an optimal biological response from such materials for rapid bone integration remains a challenge. Here we propose for the first time a one-step ion-assisted plasma polymerization process to create bio-functional 3D printed titanium (Ti) implants that offer rapid bone integration. Using selective laser melting, porous Ti implants with enhanced bone-mimicking mechanical properties were fabricated. The implants were functionalized uniformly with a highly reactive, radical-rich polymeric coating generated using a unique combination of plasma polymerization and plasma immersion ion implantation. We demonstrated the performance of such activated Ti implants with a focus on the coating's homogeneity, stability, and biological functionality. It was shown that the optimized coating was highly robust and possessed superb physico-chemical stability in a corrosive physiological solution. The plasma activated coating was cytocompatible and non-immunogenic; and through its high reactivity, it allowed for easy, one-step covalent immobilization of functional biomolecules in the absence of solvents or chemicals. The activated Ti implants bio-functionalized with bone morphogenetic protein 2 (BMP-2) showed a reduced protein desorption and a more sustained osteoblast response both in vitro and in vivo compared to implants modified through conventional physisorption of BMP-2. The versatile new approach presented here will enable the development of bio-functionalized additively manufactured implants that are patient-specific and offer improved integration with host tissue. STATEMENT OF SIGNIFICANCE: Additive manufacturing has revolutionized the fabrication of patient-specific orthopedic implants. Although such 3D printed implants can show desirable mechanical and mass transport properties, they often require surface bio-functionalities to enable control over the biological response. Surface covalent immobilization of bioactive molecules is a viable approach to achieve this. Here we report the development of additively manufactured titanium implants that precisely replicate the physical properties of native bone and are bio-functionalized in a simple, reagent-free step. Our results show that covalent attachment of bone-related growth factors through ion-assisted plasma polymerized interlayers circumvents their desorption in physiological solution and significantly improves the bone induction by the implants both in vitro and in vivo.
Collapse
|
32
|
Wang M, Ge X, Zheng Y, Wang C, Zhang Y, Lin Y. Microarray analysis reveals that lncRNA PWRN1-209 promotes human bone marrow mesenchymal stem cell osteogenic differentiation on microtopography titanium surface in vitro. J Biomed Mater Res B Appl Biomater 2020; 108:2889-2902. [PMID: 32447825 DOI: 10.1002/jbm.b.34620] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 03/23/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022]
Abstract
Sandblasted, large-grit, and acid-etched (SLA) titanium (Ti) with microtopography is currently one of the most widely used implant materials to accelerate osseointegration. Numerous long noncoding RNAs (lncRNAs) have been involved in bone remodeling, with their role in osseointegration, and the underlying mechanisms remain largely unclear. Here, microarrays of human bone marrow mesenchymal stem cells (hBMSCs) were used to identify differentially expressed lncRNAs during early cell differentiation stages (0-7 days) on SLA Ti and polished Ti surfaces. The function of lncRNAs in the osteogenic differentiation of hBMSCs was identified by RNA silencing and overexpression assays. RT-PCR and Western blot were used to detect RNA and protein expression. Alkaline phosphatase (ALP) protein activity was tested by ALP staining. Altogether, 4112 differentially expressed lncRNAs were identified from day 0 to day 7 on SLA Ti with a novel lncRNA, Prader-willi region non-coding RNA 1-209 (PWRN1-209) upregulated. We then proved that PWRN1-209 promoted osteogenic differentiation in hBMSCs by genetic tools. The upregulation of PWRN1-209 was further confirmed to be related to the surface topography of Ti by comparing SLA Ti and polished Ti. Interestingly, this trend seems to have a certain correlation with the mRNA expression level of integrins (α2, αV, β1, β2) and the phosphorylation of focal adhesion kinase (FAK). Taken together, the lncRNA PWRN1-209 was upregulated by the SLA microtopography Ti surface, which may regulate osteogenic differentiation of hBMSCs through integrin-FAK-ALP signaling. Our results provide new insights into the relationship between surface topography and osseointergration.
Collapse
Affiliation(s)
- Mingyue Wang
- Department of Implantology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, People's Republic of China
| | - Xiyuan Ge
- Central Laboratory, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, People's Republic of China
| | - Yan Zheng
- Department of Implantology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, People's Republic of China
| | - Chenxi Wang
- Department of Implantology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, People's Republic of China
| | - Yu Zhang
- Department of Implantology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, People's Republic of China
| | - Ye Lin
- Department of Implantology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, People's Republic of China
| |
Collapse
|
33
|
Lotz EM, Berger MB, Boyan BD, Schwartz Z. Regulation of mesenchymal stem cell differentiation on microstructured titanium surfaces by semaphorin 3A. Bone 2020; 134:115260. [PMID: 32028017 PMCID: PMC7749709 DOI: 10.1016/j.bone.2020.115260] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 10/25/2022]
Abstract
Peri-implant bone formation depends on the ability of mesenchymal stem cells (MSCs) to colonize implant surfaces and differentiate into osteoblasts, but the precise mechanisms controlling this process remain unclear. In vitro, MSCs undergo osteoblastic differentiation on microstructured titanium (Ti) surfaces in the absence of exogenous media supplements and produce factors that promote osteogenesis while regulating osteoclast activity, including semaphorins. The goal of this study was to evaluate the role of semaphorin 3A (Sema3A) on surface-mediated osteoblastic differentiation and determine the hierarchy of this signaling cascade. Human MSCs were cultured on 15 mm grade 2 smooth (pretreatment, PT), hydrophobic-microrough (sand blasted/acid etched, SLA), hydrophilic-microrough Ti (mSLA) (Institut Straumann AG, Basel, Switzerland), or tissue culture polystyrene (TCPS). Expression of SEMA3A family proteins increased after 7 days of culture, and the increased expression in response to microstructured Ti was dependent on recognition of the surface by integrin α2β1. Exogenous Sema3A increased differentiation whereas differentiation was decreased in cells treated with a Sema3A antibody. Furthermore, Sema3A influenced the production of osteoprotegerin and osteopontin suggesting it as an important local regulator of bone remodeling. Inhibition of Wnt3A and Wnt5A revealed that activation of Sema3A occurs downstream of Wnt5A and may facilitate the translocation of β-catenin bypassing the canonical Wnt3A initiating signal associated with osteoblastic differentiation. Furthermore, chemical inhibition of calmodulin (CaM), Ca2+/calmodulin-dependent protein kinase (CaMKII), phospholipase A2 (PLA2), protein kinase C (PKC), and BMP receptors suggest that Sema3A could serve as a feedback mechanism for both Wnt5A and BMP2. Here, we show novel roles for Sema3A family proteins in the surface-dependent modulation of MSCs as well as important interactions with pathways known to be associated with osteoblastic differentiation. Moreover, their effects on bone remodeling markers have significant implications for peri-implant bone remodeling and downstream modulation of osteoclastic activity. These results suggest that Sema3A aids in peri-implant bone formation through regulation on multiple stages of osseointegration, making it a potential target to promote osseointegration in patients with compromised bone remodeling.
Collapse
Affiliation(s)
- Ethan M Lotz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Michael B Berger
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
34
|
Berger MB, Jacobs TW, Boyan BD, Schwartz Z. Hot isostatic pressure treatment of 3D printed Ti6Al4V alters surface modifications and cellular response. J Biomed Mater Res B Appl Biomater 2020; 108:1262-1273. [PMID: 31469519 PMCID: PMC7048629 DOI: 10.1002/jbm.b.34474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/26/2019] [Accepted: 08/06/2019] [Indexed: 11/05/2022]
Abstract
Additive manufacturing can be used to create personalized orthopedic and dental implants with varying geometries and porosities meant to mimic morphological properties of bone. These qualities can alleviate stress shielding and increase osseointegration through bone ingrowth, but at the expense of reduced fatigue properties compared to machined implants, and potential for loose build particle erosion. Hot isostatic pressure (HIP) treatment is used to increase fatigue resistance; implant surface treatments like grit-blasting and acid-etching create microroughness and reduce the presence of loose particles. However, it is not known how HIP treatment affects surface treatments and osseointegration of the implant to bone. We manufactured two titanium-aluminum-vanadium constructs, one with simple through-and-through porosity and one possessing complex trabecular bone-like porosity. We observed HIP treatment varied in effect and was dependent on architecture. Micro/meso/nano surface properties generated by grit-blasting and acid-etching were altered on biomimetic HIP-treated constructs. Human mesenchymal stem cells (MSCs) were cultured on constructs fabricated +/- HIP and subsequently surface-treated. MSCs were sensitive to 3D-architecture, exhibiting greater osteogenic differentiation on constructs with complex trabecular bone-like porosity. HIP-treatment did not alter the osteogenic response of MSCs to these constructs. Thus, HIP may provide mechanical and biological advantages during implant osseointegration and function.
Collapse
Affiliation(s)
- Michael B. Berger
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond VA 23284, USA
| | - Thomas W. Jacobs
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond VA 23284, USA
| | - Barbara D. Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond VA 23284, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond VA 23284, USA
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| |
Collapse
|
35
|
Andrukhov O, Behm C, Blufstein A, Wehner C, Gahn J, Pippenger B, Wagner R, Rausch-Fan X. Effect of implant surface material and roughness to the susceptibility of primary gingival fibroblasts to inflammatory stimuli. Dent Mater 2020; 36:e194-e205. [PMID: 32360041 DOI: 10.1016/j.dental.2020.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/04/2020] [Accepted: 04/13/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVES The impact of the implant surface material and roughness on inflammatory processes in peri-implantitis is not entirely clear. Hence, we investigated how titanium and zirconia surfaces with different roughness influence the susceptibility of primary human gingival fibroblasts to different inflammatory stimuli. METHODS Primary human gingival fibroblasts were isolated from 8 healthy individuals and cultured on following surfaces: smooth titanium machined surface (TiM), smooth zirconia machined surface (ZrM), moderately rough titanium surface (SLA), or moderately rough zirconia surface (ZLA). Subsequently, stimulation with one of the following stimuli was performed: Porphyromonas gingivalis lipopolysaccharide (LPS), tumor necrosis factor (TNF)-α, interleukin (IL)-1β. The resulting production of IL-6, IL-8, and monocyte chemoattractant protein (MCP)-1 was measured by qPCR and ELISA. RESULTS P. gingivalis LPS induced IL-6 and MCP-1 production was slightly higher on titanium surfaces compared to zirconia surfaces. IL-1β induced IL-6 production was not affected by any surface characteristic. The production of MCP-1 in response to IL-1β was higher on smooth compared to rough surfaces and was not affected by the material. The production of IL-6 and MCP-1 in response to TNF-α was most strongly affected by surface characteristics. Higher production of these cytokine was observed on smooth compared to rough surfaces and on titanium compared to zirconia surfaces. Surface characteristics had only minor effects on IL-8 production. SIGNIFICANCE The susceptibility of primary gingival fibroblasts to inflammation depends on various factors, such as surface material, surface roughness and the nature of inflammatory stimuli. All these factors might determine susceptibility to peri-implantitis.
Collapse
Affiliation(s)
- Oleh Andrukhov
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.
| | - Christian Behm
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Alice Blufstein
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Christian Wehner
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Johannes Gahn
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | | | | | - Xiaohui Rausch-Fan
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Zhu W, Guo M, Yang W, Tang M, Chen T, Gan D, Zhang D, Ding X, Zhao A, Zhao P, Yan W, Zhang J. CD41-deficient exosomes from non-traumatic femoral head necrosis tissues impair osteogenic differentiation and migration of mesenchymal stem cells. Cell Death Dis 2020; 11:293. [PMID: 32341357 PMCID: PMC7184624 DOI: 10.1038/s41419-020-2496-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
Non-traumatic osteonecrosis of the femoral head (ONFH) is clinically a devastating and progressive disease without an effective treatment. Mesenchymal stem cells (MSCs) transplantation has been used to treat ONFH in early stage, but the failure rate of this therapy is high due to the reduced osteogenic differentiation and migration of the transplanted MSCs related with pathological bone tissues. However, the mechanism responsible for this decrease is still unclear. Therefore, we assume that the implanted MSCs might be influenced by signals delivered from pathological bone tissue, where the exosomes might play a critical role in this delivery. This study showed that exosomes from ONFH bone tissues (ONFH-exos) were able to induce GC-induced ONFH-like damage, in vivo and impair osteogenic differentiation and migration of MSCs, in vitro. Then, we analyzed the differentially expressed proteins (DEPs) in ONFH-exos using proteomic technology and identified 842 differentially expressed proteins (DEPs). On the basis of gene ontology (GO) enrichment analysis of DEPs, fold-changes and previous report, cell adhesion-related CD41 (integrin α2b) was selected for further investigation. Our study showed that the CD41 (integrin α2b) was distinctly decreased in ONFH-exos, compared to NOR-exos, and downregulation of CD41 could impair osteogenic differentiation and migration of the MSCs, where CD41-integrin β3-FAK-Akt-Runx2 pathway was involved. Finally, our study further suggested that CD41-affluent NOR-exos could restore the glucocorticoid-induced decline of osteogenic differentiation and migration in MSCs, and prevent GC-induced ONFH-like damage in rat models. Taken together, our study results revealed that in the progress of ONFH, exosomes from the pathological bone brought about the failure of MSCs repairing the necrotic bone for lack of some critical proteins, like integrin CD41, and prompted the progression of experimentally induced ONFH-like status in the rat. CD41 could be considered as the target of early diagnosis and therapy in ONFH.
Collapse
Affiliation(s)
- Weiwen Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - MinKang Guo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wu Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Min Tang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Tingmei Chen
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Delu Gan
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Dian Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaojuan Ding
- Department of Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Anping Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pei Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenlong Yan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jian Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
37
|
Lotz EM, Lohmann CH, Boyan BD, Schwartz Z. Bisphosphonates inhibit surface-mediated osteogenesis. J Biomed Mater Res A 2020; 108:1774-1786. [PMID: 32276287 DOI: 10.1002/jbm.a.36944] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Bisphosphonates (BPs) target osteoclasts, slowing bone resorption thus providing rationale to support osseointegration. However, BPs may negatively affect osteoblasts, impairing peri-implant bone formation. The goal of this study was to assess the effects BPs have on surface-mediated osteogenesis of osteoblasts. MG63 cells were cultured on 15-mm grade 2 titanium disks: smooth, hydrophobic-microrough, or hydrophilic-microrough (Institut Straumann AG, Basel, Switzerland). Tissue culture polystyrene (TCPS) was used as a control. At confluence, cells were treated with 0, 10-8 , 10-7 , and 10-6 M of alendronate, zoledronate, or ibandronate for 24 hr. Sprague Dawley rats were also treated with 1 μg/kg/day ibandronate or phosphate-buffered saline control for 5 weeks. Calvarial osteoblasts (rat osteoblasts [rOBs]) were isolated, characterized, and cultured on surfaces. Osteogenic markers in the media were quantified using ELISAs. BP treatment reduced osteocalcin, osteoprotegerin, osteopontin, bone morphogenetic protein-2, prostaglandin E2 , transforming growth factor β1, interleukin 10, and vascular endothelial growth factor in MG63 cells. The effect was more robust on rough surfaces, and higher concentrations of BPs stunted production to TCPS/PT levels. Ibandronate conditioned rOBs produced less osteogenic markers similar to direct BP treatment. These results suggest that BP exposure jeopardizes the pro-osteogenic response osteoblasts have to microstructured surfaces. Their effects persist in vivo and negatively condition osteoblast response in vitro. Clinically, BPs could compromise osseointegration.
Collapse
Affiliation(s)
- Ethan M Lotz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Christoph H Lohmann
- Department of Orthopaedics, Otto-von-Guericke-University, Magdeburg, Germany
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
38
|
Martelli G, Bloise N, Merlettini A, Bruni G, Visai L, Focarete ML, Giacomini D. Combining Biologically Active β-Lactams Integrin Agonists with Poly(l-lactic acid) Nanofibers: Enhancement of Human Mesenchymal Stem Cell Adhesion. Biomacromolecules 2020; 21:1157-1170. [PMID: 32011862 PMCID: PMC7997109 DOI: 10.1021/acs.biomac.9b01550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Regulating
stem cell adhesion and growth onto functionalized biomaterial
scaffolds is an important issue in the field of tissue engineering
and regenerative medicine. In this study, new electrospun scaffolds
of poly(l-lactic acid) (PLLA), as bioresorbable polymer,
and β-lactam compounds agonists of selected integrins, as functional
components with cell adhesive properties, are designed. The new β-lactam-PLLA
scaffolds contribute significantly in guiding protein translation
involved in human bone marrow mesenchymal stem cells (hBM-MSC) adhesion
and integrin gene expression. Scanning electron microscopy, confocal
laser scanning microscopy, and Western Blot analyses reveal that GM18-PLLA
shows the best results, promoting cell adhesion by significantly driving
changes in focal adhesion proteins distribution (β1 integrin and vinculin) and activation (pFAK), with a notable increase
of GM18-targets subunits integrin gene expression, α4 and β1. These novel functionalized submicrometric
fibrous scaffolds demonstrate, for the first time, the powerful combination
of selective β-lactams agonists of integrins with biomimetic
scaffolds, suggesting a designed rule that could be suitably applied
to tissue repair and regeneration.
Collapse
Affiliation(s)
- Giulia Martelli
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Nora Bloise
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Via S. Boezio 28, 27100 Pavia, Italy
| | - Andrea Merlettini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giovanna Bruni
- Department of Chemistry, Section of Physical Chemistry, University of Pavia, Viale Taramelli 16, 27100 Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Via S. Boezio 28, 27100 Pavia, Italy
| | - Maria Letizia Focarete
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Daria Giacomini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
39
|
Jarolimova P, Voltrova B, Blahnova V, Sovkova V, Pruchova E, Hybasek V, Fojt J, Filova E. Mesenchymal stem cell interaction with Ti 6Al 4V alloy pre-exposed to simulated body fluid. RSC Adv 2020; 10:6858-6872. [PMID: 35493900 PMCID: PMC9049760 DOI: 10.1039/c9ra08912h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/13/2020] [Indexed: 11/21/2022] Open
Abstract
Titanium and its alloys are widely used for substitution of hard tissues, especially in orthopaedic and dental surgery. Despite the benefit of the use of titanium for such applications, there are still questions which must be sorted out. Surface properties are crucial for cell adhesion, proliferation and differentiation. Mainly, micro/nanostructured surfaces positively influence osteogenic differentiation of human mesenchymal stem cells. Ti6Al4V is a biocompatible α + β alloy which is widely used in orthopaedics. The aim of this study was to investigate the interaction of the nanostructured and ground Ti6Al4V titanium alloys with simulated body fluid complemented by the defined precipitation of hydroxyapatite-like coating and to study the cytotoxicity and differentiation capacity of cells with such a modified titanium alloy. Nanostructures were fabricated using electrochemical oxidation. Human mesenchymal stem cells (hMSC) were used to evaluate cell adhesion, metabolic activity and proliferation on the specimens. The differentiation potential of the samples was investigated using PCR and specific staining of osteogenic markers collagen type I and osteocalcin. Our results demonstrate that both pure Ti6Al4V, nanostructured samples, and hydroxyapatite-like coating supported hMSC growth and metabolic activity. Nanostructured samples improved collagen type I synthesis after 14 days, while both nanostructured and hydroxyapatite-like coated samples enhanced collagen synthesis on day 21. Osteocalcin synthesis was the most enhanced by hydroxyapatite-like coating on the nanostructured surfaces. Our results indicate that hydroxyapatite-like coating is a useful tool guiding hMSC osteogenic differentiation.
Collapse
Affiliation(s)
- Petra Jarolimova
- Department of Metals and Corrosion Engineering, Faculty of Chemical Technology, University of Chemistry and Technology Technická 5 166 28 Prague Czech Republic
| | - Barbora Voltrova
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 Prague 4 142 20 Czech Republic
- Faculty of Science, Charles University in Prague Albertov 2038/6 128 00 Prague Czech Republic
| | - Veronika Blahnova
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 Prague 4 142 20 Czech Republic
- Second Faculty of Medicine, Charles University in Prague V Úvalu 84 150 06 Prague Czech Republic
- University Centre for Energy Efficient Buildings, Czech Technical University in Prague Třinecká 1024 273 43 Buštěhrad Czech Republic
| | - Vera Sovkova
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 Prague 4 142 20 Czech Republic
- University Centre for Energy Efficient Buildings, Czech Technical University in Prague Třinecká 1024 273 43 Buštěhrad Czech Republic
| | - Eva Pruchova
- Department of Metals and Corrosion Engineering, Faculty of Chemical Technology, University of Chemistry and Technology Technická 5 166 28 Prague Czech Republic
| | - Vojtech Hybasek
- Department of Metals and Corrosion Engineering, Faculty of Chemical Technology, University of Chemistry and Technology Technická 5 166 28 Prague Czech Republic
| | - Jaroslav Fojt
- Department of Metals and Corrosion Engineering, Faculty of Chemical Technology, University of Chemistry and Technology Technická 5 166 28 Prague Czech Republic
| | - Eva Filova
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 Prague 4 142 20 Czech Republic
- Second Faculty of Medicine, Charles University in Prague V Úvalu 84 150 06 Prague Czech Republic
| |
Collapse
|
40
|
Lopes HB, Souza ATP, Freitas GP, Elias CN, Rosa AL, Beloti MM. Effect of focal adhesion kinase inhibition on osteoblastic cells grown on titanium with different topographies. J Appl Oral Sci 2020; 28:e20190156. [PMID: 32049134 PMCID: PMC6999121 DOI: 10.1590/1678-7757-2019-0156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 09/23/2019] [Indexed: 12/30/2022] Open
Abstract
Objective The present study aimed to investigate the participation of focal adhesion kinases (FAK) in interactions between osteoblastic cells and titanium (Ti) surfaces with three different topographies, namely, untreated (US), microstructured (MS), and nanostructured (NS). Methodology Osteoblasts harvested from the calvarial bones of 3-day-old rats were cultured on US, MS and NS discs in the presence of PF-573228 (FAK inhibitor) to evaluate osteoblastic differentiation. After 24 h, we evaluated osteoblast morphology and vinculin expression, and on day 10, the following parameters: gene expression of osteoblastic markers and integrin signaling components, FAK protein expression and alkaline phosphatase (ALP) activity. A smooth surface, porosities at the microscale level, and nanocavities were observed in US, MS, and NS, respectively. Results FAK inhibition decreased the number of filopodia in cells grown on US and MS compared with that in NS. FAK inhibition decreased the gene expression of Alp, bone sialoprotein, osteocalcin, and ALP activity in cells grown on all evaluated surfaces. FAK inhibition did not affect the gene expression of Fak, integrin alpha 1 ( Itga1 ) and integrin beta 1 ( Itgb1 ) in cells grown on MS, increased the gene expression of Fak in cells grown on NS, and increased the gene expression of Itga1 and Itgb1 in cells grown on US and NS. Moreover, FAK protein expression decreased in cells cultured on US but increased in cells cultured on MS and NS after FAK inhibition; no difference in the expression of vinculin was observed among cells grown on all surfaces. Conclusions Our data demonstrate the relevance of FAK in the interactions between osteoblastic cells and Ti surfaces regardless of surface topography. Nanotopography positively regulated FAK expression and integrin signaling pathway components during osteoblast differentiation. In this context, the development of Ti surfaces with the ability to upregulate FAK activity could positively impact the process of implant osseointegration.
Collapse
Affiliation(s)
- Helena Bacha Lopes
- Universidade de São Paulo, Faculdade de Odontologia de Ribeirão Preto, Bone Research Laboratory, Ribeirão Preto, São Paulo, Brasil
| | - Alann Thaffarell Portilho Souza
- Universidade de São Paulo, Faculdade de Odontologia de Ribeirão Preto, Bone Research Laboratory, Ribeirão Preto, São Paulo, Brasil
| | - Gileade Pereira Freitas
- Universidade de São Paulo, Faculdade de Odontologia de Ribeirão Preto, Bone Research Laboratory, Ribeirão Preto, São Paulo, Brasil
| | - Carlos Nelson Elias
- Instituto Militar de Engenharia, Laboratório de Biomateriais, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Adalberto Luiz Rosa
- Universidade de São Paulo, Faculdade de Odontologia de Ribeirão Preto, Bone Research Laboratory, Ribeirão Preto, São Paulo, Brasil
| | - Marcio Mateus Beloti
- Universidade de São Paulo, Faculdade de Odontologia de Ribeirão Preto, Bone Research Laboratory, Ribeirão Preto, São Paulo, Brasil
| |
Collapse
|
41
|
Osteoblasts grown on microroughened titanium surfaces regulate angiogenic growth factor production through specific integrin receptors. Acta Biomater 2019; 97:578-586. [PMID: 31349056 DOI: 10.1016/j.actbio.2019.07.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/27/2019] [Accepted: 07/22/2019] [Indexed: 01/08/2023]
Abstract
Cellular attachment and response to biomaterials are mediated by integrin receptor binding to extracellular matrix proteins adsorbed onto the material surface. Osteoblasts interact with their substrates via several integrin complexes including fibronectin-binding α5β1 and collagen-binding α1β1 and α2β1. Knockdown of α2 or β1 integrin subunits inhibits the production of factors that promote an osteogenic microenvironment, including osteocalcin, osteoprotegerin, and TGFβ1. Osteoblasts also secrete several angiogenic growth factors such as VEGF-A (VEGF165), FGF-2, and angiopoietin 1, which are regulated by titanium surface topography and surface energy. Here, we examined whether signaling through integrin receptor complexes regulates production and secretion of angiogenic factors during osteoblast differentiation on microtextured Ti surfaces. To do this, integrin subunits α1, α2, α5, and β1 were stably silenced in MG63 osteoblast-like cells cultured on grit-blasted/acid-etched hydrophobic Ti (SLA) or on hydrophilic SLA (modSLA). VEGF-A production increased in response to Ti surface topography and energy in integrin α2, α5, and β1 silenced cells but decreased in α1-silenced cells. FGF-2 decreased on modSLA substrates in both α1 and α2-silenced cells but was unchanged in response to silencing of either α5 or β1. In integrin α1, α2, and β1-silenced cells, Ang-1 increased on modSLA but α5-silencing did not affect Ang-1 production during surface mediated differentiation. These results suggest that signaling through specific integrin receptor complexes during osteoblast differentiation on microstructured Ti substrates, regulates the production of angiogenic factors by those cells, and this is differentially regulated by surface hydrophilicity. STATEMENT OF SIGNIFICANCE: Successful implantation of synthetic biomaterials into bone depends on the biological process known as osseointegration. Osseointegration is a highly regulated communication of cells that orchestrates the migration of progenitor cells towards the implant site and promotes the deposition and mineralization of extracellular matrix proteins within the implant microenvironment, to tightly join the implant to native bone. In this process, angiogenesis functions as the initiation site of progenitor cell migration and is necessary for matrix deposition by providing the necessary nutrients for bone formation. In the present study, we show a novel regulation of specific angiogenic growth factors by integrin receptor complexes. This research is important to develop biomaterials that promote and maintain osseointegration through proper vascularization and prevent implant failure in patients lacking sufficient angiogenesis.
Collapse
|
42
|
Effect of Mother’s Age and Pathology on Functional Behavior of Amniotic Mesenchymal Stromal Cells—Hints for Bone Regeneration. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9173471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human amnion-derived mesenchymal stromal cells (hAMSCs) are used increasingly in regenerative medicine applications, including dentistry. The aim of this study was to evaluate if hAMSCs from aged and pathological mothers could be affected in their phenotype and functional behavior. hAMSCs were isolated from placentas of women aged younger than 40 years (Group 1, n = 7), older than 40 years (Group 2, n = 6), and with pre-eclampsia (Group 3, n = 5). Cell yield and viability were assessed at isolation (p0). Cell proliferation was evaluated from p0 to p5. Passage 2 was used to determine the phenotype, the differentiation capacity, and the adhesion to machined and sandblasted titanium disks. hAMSCs recovered from Group 3 were fewer than in Group 1. Viability and doubling time were not different among the three groups. Percentages of CD29+ cells were significantly lower in Group 3, while percentages of CD73+ cells were significantly lower in Groups 2 and 3 as compared with Group 1. hAMSCs from Group 2 showed a significant lower differentiation capacity towards chondrogenic and osteogenic lineages. hAMSCs from Group 3 adhered less to titanium surfaces. In conclusion, pathology can affect hAMSCs in phenotype and functional behavior and may alter bone regeneration capacities.
Collapse
|
43
|
Titania nanotubes promote osteogenesis via mediating crosstalk between macrophages and MSCs under oxidative stress. Colloids Surf B Biointerfaces 2019; 180:39-48. [DOI: 10.1016/j.colsurfb.2019.04.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022]
|
44
|
Yin L, Chang Y, You Y, Liu C, Li J, Lai HC. Biological responses of human bone mesenchymal stem cells to Ti and TiZr implant materials. Clin Implant Dent Relat Res 2019; 21:550-564. [PMID: 31009155 DOI: 10.1111/cid.12756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/06/2019] [Accepted: 01/29/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Titanium-zirconium alloy (TiZr1317) is a new material used for biological implants. There are several studies on the effects of TiZr implants on the biological characteristics of human bone mesenchymal stem cells (hBMSCs). PURPOSE The purpose of this study was to investigate the biological responses of hBMSCs to implant holes affected by the physicochemical properties of oral implants (TiSLA , TiSLActive , TiZrSLA , and TiZrSLActive ). MATERIALS AND METHODS Grade 4 Ti and TiZr (13-17% Zr) substrates were modified by sand-blasted large-grit acid-etched (SLA) or hydrophilic sand-blasted large-grit acid-etched (SLActive), resulting in four types of surface with complex microstructures corresponding to the commercially-available implants SLA, RoxolidSLA, SLActive, and RoxolidSLActive (Institute Straumann AG, Basel, Switzerland). Physicochemical properties were detected and the biological responses of hBMSCs were observed. RESULTS Surface morphology characterization by scanning electron microscopy and atomic force microscopy revealed differences between the four groups. SLActive had higher surface energy/wettability than SLA, indicating that increased surface energy/wettability can promote the absorption of osteogenic proteins and enhance osseointegration. hBMSCs seeded on SLActive substrates exhibited better performance in terms of cell attachment, proliferation and osteoblastic differentiation than cells seeded on SLA. CONCLUSION Because of their more suitable physicochemical properties, TiSLActive and TiZrSLActive materials demonstrated more pronounced effects on the biological responses of hBMSCs compared with TiSLA and TiZrSLA .
Collapse
Affiliation(s)
- Lihua Yin
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yaoren Chang
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Yuanhe You
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Chun Liu
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Jie Li
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Hong-Chang Lai
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
45
|
Moura CEB, Queiroz Neto MF, Braz JKFS, de Medeiros Aires M, Silva Farias NB, Barboza CAG, Cavalcanti Júnior GB, Rocha HAO, Alves Junior C. Effect of plasma-nitrided titanium surfaces on the differentiation of pre-osteoblastic cells. Artif Organs 2019; 43:764-772. [PMID: 30779451 DOI: 10.1111/aor.13438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/07/2019] [Accepted: 02/16/2019] [Indexed: 12/15/2022]
Abstract
A titanium surface nitrided by plasma contains nitrogen ions that guarantee resistance to corrosion and biocompatibility. Despite this, no descriptions concerning the influence of the expression of cell adhesion proteins and their influence on osteogenic cell differentiation are available. Thus, the present study aimed to assess the response of murine pre-osteoblastic cells (MC3T3-E1) cultured on nitrided titanium surfaces. Pre-osteoblastic cells were grown on polished titanium discs, used as controls, and on previously characterized plasma-nitrided titanium discs. Cells from both groups were submitted to the MTT cell viability test. The expressions of α5, α2, and β1 integrin were assessed by flow cytometry and immunofluorescence, while osteocalcin expression was assessed by flow cytometry. The nitrided surface presented higher α2 and β1 integrin expressions, as well as osteocalcin expression, when compared to the polished surface, with no alterations in cell viability. These findings seem to suggest that the plasma nitriding treatment produces a titanium surface with the potential for effective in vitro osseointegration.
Collapse
Affiliation(s)
- Carlos Eduardo B Moura
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Moacir F Queiroz Neto
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Janine Karla F S Braz
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | | | - Naisandra B Silva Farias
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Carlos Augusto G Barboza
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | | | - Hugo Alexandre O Rocha
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Clodomiro Alves Junior
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| |
Collapse
|
46
|
Lopes HB, Freitas GP, Elias CN, Tye C, Stein JL, Stein GS, Lian JB, Rosa AL, Beloti MM. Participation of integrin β3 in osteoblast differentiation induced by titanium with nano or microtopography. J Biomed Mater Res A 2019; 107:1303-1313. [PMID: 30707485 DOI: 10.1002/jbm.a.36643] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/03/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022]
Abstract
The major role of integrins is to mediate cell adhesion but some of them are involved in the osteoblasts-titanium (Ti) interactions. In this study, we investigated the participation of integrins in osteoblast differentiation induced by Ti with nanotopography (Ti-Nano) and with microtopography (Ti-Micro). By using a PCR array, we observed that, compared with Ti-Micro, Ti-Nano upregulated the expression of five integrins in mesenchymal stem cells, including integrin β3, which increases osteoblast differentiation. Silencing integrin β3, using CRISPR-Cas9, in MC3T3-E1 cells significantly reduced the osteoblast differentiation induced by Ti-Nano in contrast to the effect on T-Micro. Concomitantly, integrin β3 silencing downregulated the expression of integrin αv, the parent chain that combines with other integrins and several components of the Wnt/β-catenin and BMP/Smad signaling pathways, all involved in osteoblast differentiation, only in cells cultured on Ti-Nano. Taken together, our results showed the key role of integrin β3 in the osteogenic potential of Ti-Nano but not of Ti-Micro. Additionally, we propose a novel mechanism to explain the higher osteoblast differentiation induced by Ti-Nano that involves an intricate regulatory network triggered by integrin β3 upregulation, which activates the Wnt and BMP signal transductions. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1303-1313, 2019.
Collapse
Affiliation(s)
- Helena B Lopes
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Gileade P Freitas
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Carlos N Elias
- Biomaterials Laboratory, Instituto Militar de Engenharia, Rio de Janeiro, RJ, Brazil
| | - Coralee Tye
- Department of Biochemistry, University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Janet L Stein
- Department of Biochemistry, University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Gary S Stein
- Department of Biochemistry, University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Jane B Lian
- Department of Biochemistry, University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Adalberto L Rosa
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Marcio M Beloti
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
47
|
Greiner JF, Gottschalk M, Fokin N, Büker B, Kaltschmidt BP, Dreyer A, Vordemvenne T, Kaltschmidt C, Hütten A, Kaltschmidt B. Natural and synthetic nanopores directing osteogenic differentiation of human stem cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:319-328. [PMID: 30771503 DOI: 10.1016/j.nano.2019.01.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 10/27/2022]
Abstract
Bone regeneration is a highly orchestrated process crucial for endogenous healing procedures after accidents, infections or tumor therapy. Changes in surface nanotopography are known to directly affect the formation of osteogenic cell types, although no direct linkage to the endogenous nanotopography of bone was described so far. Here we show the presence of pores of 31.93 ± 0.97 nm diameter on the surface of collagen type I fibers, the organic component of bone, and demonstrate these pores to be sufficient to induce osteogenic differentiation of adult human stem cells. We further applied SiO2 nanoparticles thermally cross-linked to a nanocomposite to artificially biomimic 31.93 ± 0.97 nm pores, which likewise led to in vitro production of bone mineral by adult human stem cells. Our findings show an endogenous mechanism of directing osteogenic differentiation of adult stem cells by nanotopological cues and provide a direct application using SiO2 nanocomposites with surface nanotopography biomimicking native bone architecture.
Collapse
Affiliation(s)
| | - Martin Gottschalk
- Thin Films & Physics of Nanostructures, Bielefeld University, Bielefeld, Germany
| | - Nadine Fokin
- Thin Films & Physics of Nanostructures, Bielefeld University, Bielefeld, Germany
| | - Björn Büker
- Thin Films & Physics of Nanostructures, Bielefeld University, Bielefeld, Germany
| | | | - Axel Dreyer
- Thin Films & Physics of Nanostructures, Bielefeld University, Bielefeld, Germany
| | - Thomas Vordemvenne
- Department of Trauma and Orthopedic Surgery, Evangelical Hospital Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, Bielefeld, Germany; Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Bielefeld, Germany
| | - Andreas Hütten
- Thin Films & Physics of Nanostructures, Bielefeld University, Bielefeld, Germany; Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, Bielefeld, Germany; Molecular Neurobiology, Bielefeld University, Bielefeld, Germany; Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
48
|
Cheng A, Schwartz Z, Kahn A, Li X, Shao Z, Sun M, Ao Y, Boyan BD, Chen H. Advances in Porous Scaffold Design for Bone and Cartilage Tissue Engineering and Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2019; 25:14-29. [PMID: 30079807 PMCID: PMC6388715 DOI: 10.1089/ten.teb.2018.0119] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022]
Abstract
IMPACT STATEMENT Challenges in musculoskeletal tissue regeneration affect millions of patients globally. Scaffolds for tissue engineering bone and cartilage provide promising solutions that increase healing and decrease need for complicated surgical procedures. Porous scaffolds have emerged as an attractive alternative to traditional scaffolds. However, the success of advanced materials, use of biological factors, and manufacturing techniques can vary depending on use case. This review provides perspective on porous scaffold manufacturing, characterization and application, and can be used to inform future scaffold design.
Collapse
Affiliation(s)
- Alice Cheng
- Department of Biomedical Engineering, Peking University, Beijing, China
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Department of Periodontology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Adrian Kahn
- Department of Oral and Maxillofacial Surgery, University of Tel Aviv, Tel Aviv, Israel
| | - Xiyu Li
- Department of Biomedical Engineering, Peking University, Beijing, China
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Zhenxing Shao
- Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Muyang Sun
- Department of Biomedical Engineering, Peking University, Beijing, China
| | - Yingfang Ao
- Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Barbara D. Boyan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Haifeng Chen
- Department of Biomedical Engineering, Peking University, Beijing, China
| |
Collapse
|
49
|
Lauria I, Kutz TN, Böke F, Rütten S, Zander D, Fischer H. Influence of nanoporous titanium niobium alloy surfaces produced via hydrogen peroxide oxidative etching on the osteogenic differentiation of human mesenchymal stromal cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:635-648. [PMID: 30813067 DOI: 10.1016/j.msec.2019.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 12/20/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
Titanium niobium alloys exhibit a lower stiffness compared to Ti6Al4V, the 'gold standard' for load-bearing bone implants. Thus, the critical mismatch in stiffness between the implant and adjacent bone tissue could be addressed with TiNb alloys and thereby reduce stress shielding, which can result in bone resorption and subsequent implant loosening; however, the cellular response on the specific material is crucial for sufficient osseointegration. We therefore hypothesize that the response of human mesenchymal stromal cells (hMSC) and osteoblast-like cells on Ti45Nb surfaces can be improved by a novel nanoporous surface structure. For this purpose, an etching technique using hydrogen peroxide electrolyte solution was applied to Ti45Nb. The treated surfaces were characterized using SEM, LSM, AFM, nanoindentation, and contact angle measurements. Cell culture experiments using hMCS and MG-63 were conducted. The H2O2 treatment resulted in surface nanopores, an increase in surface wettability and a reduction in surface hardness. The proliferation of MG-63 was enhanced on TiNb45 compared to Ti6Al4V. MG-63 focal adhesion complexes were detected on all Ti45Nb surfaces, whereas the nanostructures notably increased the cell area and decreased cell solidity, indicating stimulated cell spreading and pseudopodia formation. Alizarin red stainings indicated that the nanoporous surfaces stimulated the osteogenic differentiation of hMSC. It can be concluded that the proposed surface treatment could potentially help to stimulate the osseointegration behaviour of the advantageous low stiff Ti45Nb alloy.
Collapse
Affiliation(s)
- Ines Lauria
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Tatiana Nicole Kutz
- Chair of Corrosion and Corrosion Protection, Foundry Institute, RWTH Aachen University, Intzestrasse 5, 52072 Aachen, Germany.
| | - Frederik Böke
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Stephan Rütten
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Daniela Zander
- Chair of Corrosion and Corrosion Protection, Foundry Institute, RWTH Aachen University, Intzestrasse 5, 52072 Aachen, Germany.
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| |
Collapse
|
50
|
Rottmar M, Müller E, Guimond-Lischer S, Stephan M, Berner S, Maniura-Weber K. Assessing the osteogenic potential of zirconia and titanium surfaces with an advanced in vitro model. Dent Mater 2019; 35:74-86. [DOI: 10.1016/j.dental.2018.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/12/2018] [Accepted: 10/12/2018] [Indexed: 10/27/2022]
|