Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Ann Biomed Eng ; 52(6): 1744-1762, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38517621

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.


Subject(s)
Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells , Osseointegration , Rats, Sprague-Dawley , Wnt Proteins , Animals , Wnt Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Rats , Cell Proliferation/drug effects , Osseointegration/drug effects , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Male , Titanium , Disease Models, Animal , Cells, Cultured
2.
J Biomed Mater Res B Appl Biomater ; 112(1): e35322, 2024 01.
Article in English | MEDLINE | ID: mdl-37737450

ABSTRACT

Three-dimensional macroporous titanium-aluminum-vanadium (TiAl6V4) implants produced by additive manufacturing (AM) can be grit blasted (GB) to yield microtextured exterior surfaces, with additional micro/nano-scale surface features provided by subsequent acid etching (AE). However, the line-of-sight nature of GB causes the topography of exterior GB + AE-modified surfaces to differ from internal GB-inaccessible surfaces. Previous in vitro studies using dense TiAl6V4 substrates indicated that a nonline-of-sight, calciothermic-reaction (CaR)-based process provided homogeneous osteogenic nanotextures on GB + AE surfaces, suggesting it could be used to achieve a homogeneous nanotopography on external and internal surfaces of macroporous AM constructs. Macroporous TiAl6V4 (3D) constructs were produced by direct laser melting and modified by GB + AE, with the CaR process then applied to 50% of constructs (3DCaR). The CaR process yielded nanoporous/nanorough internal surfaces throughout the macroporous constructs. Skeletally mature, male Sprague-Dawley rats were implanted with these constructs using a cranial on-lay model. Prior to implantation, a Cu++-free click hydrogel was applied to half of the constructs (3D + H, 3DCaR + H) to act as a challenge to osseointegration. Osseointegration was compared between the four implant groups (3D, 3DCaR, 3D + H, 3DCaR + H) at 4w. 3D + H implants exhibited lower bone volume (BV) and percent bone ingrowth (%BI) than the 3D implants. In contrast, osseointegrated 3DCaR + H implants had similar BV and %BI as the 3DCaR implants. Implant pull-off forces correlated with these results. In vitro analyses indicated that human bone marrow stromal cells (MSCs) exhibited enhanced production of osteoblast differentiation markers and factors associated with osteogenesis when grown on CaR-modified 3D substrates relative to control (TCPS) substrates. This work confirms that the CaR process provides osteogenic nanotextures on internal surfaces of macroporous 3D implants, and suggests that CaR-modified surfaces can promote osseointegration in cases where osteogenesis is impaired.


Subject(s)
Osteogenesis , Titanium , Rats , Animals , Male , Humans , Rats, Sprague-Dawley , Titanium/pharmacology , Aluminum , Vanadium , Biomimetics , Osseointegration , Surface Properties
3.
Biomimetics (Basel) ; 8(1)2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36975323

ABSTRACT

Reduced skeletal loading associated with many conditions, such as neuromuscular injuries, can lead to bone fragility and may threaten the success of implant therapy. Our group has developed a botulinum toxin A (botox) injection model to imitate disease-reduced skeletal loading and reported that botox dramatically impaired the bone formation and osseointegration of titanium implants. Semaphorin 3A (sema3A) is an osteoprotective factor that increases bone formation and inhibits bone resorption, indicating its potential therapeutic role in improving osseointegration in vivo. We first evaluated the sema3A effect on whole bone morphology following botox injections by delivering sema3A via injection. We then evaluated the sema3A effect on the osseointegration of titanium implants with two different surface topographies by delivering sema3A to cortical bone defect sites prepared for implant insertion and above the implants after insertion using a copper-free click hydrogel that polymerizes rapidly in situ. Implants had hydrophobic smooth surfaces (PT) or multiscale biomimetic micro/nano topography (SLAnano). Sema3A rescued the botox-impaired bone formation. Furthermore, biomimetic Ti implants improved the bone-to-implant contact (BIC) and mechanical properties of the integrated bone in the botox-treated rats, which sema3A enhanced. This study demonstrated the value of biomimetic approaches combining multiscale topography and biologics in improving the clinical outcomes of implant therapy.

4.
Biomed Mater ; 18(3)2023 03 08.
Article in English | MEDLINE | ID: mdl-36827708

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.


Subject(s)
Mesenchymal Stem Cells , Titanium , Animals , Mice , Titanium/chemistry , Aluminum/metabolism , Vanadium/metabolism , Interleukin-6/metabolism , X-Ray Microtomography , Biomimetics , Interleukin-10/metabolism , Interleukin-4/metabolism , Mice, Nude , Osteogenesis , Cell Differentiation , Polyethylene Glycols/chemistry , Cytokines/metabolism , DNA/metabolism , Surface Properties , Osseointegration , Osteoblasts , Cells, Cultured
5.
J Biomed Mater Res B Appl Biomater ; 111(4): 829-845, 2023 04.
Article in English | MEDLINE | ID: mdl-36372947

ABSTRACT

Current standards in bone-facing implant fabrication by metal 3D (M3D) printing require post-manufacturing modifications to create distinct surface properties and create implant microenvironments that promote osseointegration. However, the biological consequences of build parameters and surface modifications are not well understood. This study evaluated the relative contributions of build parameters and post-manufacturing modification techniques to cell responses that impact osseointegration in vivo. Biomimetic testing constructs were created by using a M3D printer with standard titanium-aluminum-vanadium (Ti6Al4V) print parameters. These constructs were treated by either grit-blasting and acid-etching (GB + AE) or GB + AE followed by hot isostatic pressure (HIP) (GB + AE, HIP). Next, nine constructs were created by using a M3D printer with three build parameters: (1) standard, (2) increased hatch spacing, and (3) no infill, and additional contour trace. Each build type was further processed by either GB + AE, or HIP, or a combination of HIP treatment followed by GB + AE (GB + AE, HIP). Resulting constructs were assessed by SEM, micro-CT, optical profilometry, XPS, and mechanical compression. Cellular response was determined by culturing human bone marrow stromal cells (MSCs) for 7 days. Surface topography differed depending on processing method; HIP created micro-/nano-ridge like structures and GB + AE created micro-pits and nano-scale texture. Micro-CT showed decreases in closed pore number and closed porosity after HIP treatment in the third build parameter constructs. Compressive moduli were similar for all constructs. All constructs exhibited ability to differentiate MSCs into osteoblasts. MSCs responded best to micro-/nano-structures created by final post-processing by GB + AE, increasing OCN, OPG, VEGFA, latent TGFß1, IL4, and IL10. Collectively these data demonstrate that M3D-printed constructs can be readily manufactured with distinct architectures based on the print parameters and post-build modifications. MSCs are sensitive to discrete surface topographical differences that may not show up in qualitative assessments of surface properties and respond by altering local factor production. These factors are vital for osseointegration after implant insertion, especially in patients with compromised bone qualities.


Subject(s)
Mesenchymal Stem Cells , Titanium , Humans , Titanium/pharmacology , Titanium/chemistry , Aluminum , Vanadium , Osseointegration , Surface Properties , Printing, Three-Dimensional
6.
Biomimetics (Basel) ; 7(3)2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36134921

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.

7.
Biomimetics (Basel) ; 7(2)2022 Apr 16.
Article in English | MEDLINE | ID: mdl-35466263

ABSTRACT

The use of metallic and polymeric materials for implants has been increasing over the past decade. This trend can be attributed to a variety of factors including a significant increase in basic science research focused on implant material characteristics and how various surface modifications may stimulate osseointegration and, ultimately, fusion. There are many interbody fusion devices and dental implants commercially available; however, detailed information about their surface properties, and the effects that various materials and surface modifications may have on osteogenesis, is lacking in the literature. While the concept of bone-implant osseointegration is a relatively recent addition to the spine fusion literature, there is a comparatively large body of literature related to dental implants. The purpose of this article is to summarize the science of surface modified bone-facing implants, focusing on biomimetic material chemistry and topography of titanium implants, to promote a better understanding of how these characteristics may impact bone formation and osseointegration. This manuscript has the following aspects: highlights the role of titanium and its alloys as potent osteoconductive bioactive materials; explores the importance of biomimetic surface topography at the macro-, micro- and nano-scale; summarizes how material surface design can influence osteogenesis and immune responses in vitro; focuses on the kinds of surface modifications that play a role in the process. Biomimetic surface modifications can be varied across many clinically available biomaterials, and the literature supports the hypothesis that those biomaterial surfaces that exhibit physical properties of bone resorption pits, such as roughness and complex hierarchical structures at the submicron and nanoscale, are more effective in supporting osteoblast differentiation in vitro and osteogenesis in vivo.

8.
J Am Acad Orthop Surg ; 30(13): e894-e898, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35383608

ABSTRACT

Bone marrow stromal cells are regulated by the chemical and physical features of a biomaterial surface. When grown on titanium (Ti) and Ti alloy surfaces, such as titanium-aluminum-vanadium, with specific topographies that mimic the microscale, mesoscale, and nanoscale features of an osteoclast resorption pit, they undergo a rapid change in cell shape to assume a columnar morphology typical of a secretory osteoblast. These cells exhibit markers associated with an osteoblast phenotype, including osteocalcin and osteopontin, and they secrete factors associated with osteogenesis, including bone morphogenetic protein 2, vascular endothelial growth factor, and neurotrophic semaphorins. The pathway involves a shift in integrin expression from α5ß1 to α2ß1 and signaling by Wnt5a rather than Wnt3a. Conditioned media from these cultures can stimulate vasculogenesis by human endothelial cells and osteoblastic differentiation of marrow stromal cells not grown on the biomimetic substrate, suggesting that the surface could promote osteogenesis in vivo through similar mechanisms. In vivo studies using a variety of animal models confirm that implants with biomimetic surfaces result in improved osseointegration compared with Ti implants with smooth surfaces, as do meta-analyses comparing clinical performance of implant surface topographies.


Subject(s)
Osseointegration , Osteogenesis , Animals , Cell Differentiation/physiology , Endothelial Cells , Humans , Osseointegration/physiology , Osteogenesis/physiology , Surface Properties , Titanium , Vascular Endothelial Growth Factor A
9.
Dent Mater ; 38(4): 632-645, 2022 04.
Article in English | MEDLINE | ID: mdl-35184898

ABSTRACT

OBJECTIVES: Increased wettability of titanium and titanium alloy surfaces due to processing and storage methods increases osteoprogenitor cell differentiation and osseointegration compared to microroughness alone. Implants that are exposed to air have a hydrophobic surface due to adsorption of atmospheric hydrocarbons, which can limit overall implant success. Dielectric barrier discharge plasma (DBD) is one method to increase surface hydrophilicity. Although current DBD methods yield a hydrophilic surface, adsorbed hydrocarbons rapidly restore hydrophobicity. We demonstrated that application of DBD to implants previously packaged in a vacuum, generates a hydrophilic surface that supports osteoblastic differentiation in vitro and this can be done immediately prior to use. In the present study, we tested the hypothesis that DBD treatment to alter surface wettability at the time of implant placement will improve osseointegration in vivo. MATERIALS AND METHODS: Twenty male and sixteen female rabbits were used in a preclinical trans-axial femur model of osseointegration. Control and DBD treatment implants were inserted randomized per hind limb in each rabbit (1 implant/hind-limb). At 6 weeks post-surgery, bone-to-implant contact, adjacent bone volume, and torque to failure were assessed by micro-CT, calcified histology, and mechanical testing. RESULTS: DBD plasma treatment of vacuum-sealed implants increased surface wettability and did not change surface chemistry or roughness. Peak torque and torsional energy, and bone-to-implant contact increased with DBD treatment in males. In contrast, female rabbits showed increased osseointegration equal to DBD treated male implants regardless of DBD plasma treatment. CONCLUSION: DBD treatment is an effective method to enhance osseointegration by increasing surface wettability; however, this response is sex dependent. In healthy female patients, DBD treatment may not be necessary but in older patients or patients with compromised bone, this treatment could be an effective measure to ensure implant success.


Subject(s)
Dental Implants , Osseointegration , Animals , Female , Hydrophobic and Hydrophilic Interactions , Male , Osseointegration/physiology , Rabbits , Surface Properties , Titanium/chemistry
10.
Dent Mater ; 37(4): 690-700, 2021 04.
Article in English | MEDLINE | ID: mdl-33589272

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.


Subject(s)
Dental Implants , Titanium , Cell Differentiation , Osteoblasts , Plasma , Surface Properties
11.
J Orthop Res ; 39(9): 1908-1920, 2021 09.
Article in English | MEDLINE | ID: mdl-33002223

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.


Subject(s)
Mesenchymal Stem Cells , Titanium , Aluminum/metabolism , Bone Marrow Cells , Cell Differentiation , Cells, Cultured , Osteoblasts/metabolism , Osteogenesis , Surface Properties , Titanium/chemistry , Vanadium
12.
Bone ; 134: 115260, 2020 05.
Article in English | MEDLINE | ID: mdl-32028017

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 α2ß1. 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 ß-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.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Osteoblasts , Semaphorin-3A , Titanium , Cells, Cultured , Humans , Osteogenesis , Semaphorin-3A/pharmacology , Surface Properties
13.
J Biomed Mater Res B Appl Biomater ; 108(4): 1262-1273, 2020 05.
Article in English | MEDLINE | ID: mdl-31469519

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.


Subject(s)
Alloys , Hot Temperature , Materials Testing , Mesenchymal Stem Cells/metabolism , Printing, Three-Dimensional , Titanium , Alloys/chemistry , Alloys/pharmacology , Humans , Pressure , Surface Properties , Titanium/chemistry , Titanium/pharmacology
14.
Acta Biomater ; 97: 578-586, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31349056

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 α5ß1 and collagen-binding α1ß1 and α2ß1. Knockdown of α2 or ß1 integrin subunits inhibits the production of factors that promote an osteogenic microenvironment, including osteocalcin, osteoprotegerin, and TGFß1. 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 ß1 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 ß1 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 ß1. In integrin α1, α2, and ß1-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.


Subject(s)
Angiopoietin-1/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Integrins/metabolism , Osteoblasts/metabolism , Titanium/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Cell Differentiation/drug effects , Cell Line , Humans , Osteoblasts/cytology , Surface Properties , Titanium/chemistry
15.
J Biomed Mater Res A ; 107(2): 423-433, 2019 02.
Article in English | MEDLINE | ID: mdl-30461195

ABSTRACT

Establishment of a patent vasculature at the bone-implant interface plays a significant role in determining overall success of orthopedic and dental implants. Osteoblasts produce vascular endothelial growth factor-A (VEGF-A), an important regulator of angiogenesis during bone formation and healing, and the amount secreted is sensitive to titanium (Ti) surface microtopography and surface energy. The purpose of this study was to determine if surface properties modulate cellular response to VEGF-A. MG63 osteoblast-like cells were transfected with shRNA targeting VEGF-A at >80% knockdown. Cells stably silenced for VEGF-A secreted reduced levels of osteocalcin, osteoprotegerin, FGF-2, and angiopoietin-1 when cultured on grit-blasted/acid-etched (SLA) and hydrophilic SLA (modSLA) Ti surfaces and conditioned media from these cultures caused reduced angiogenesis in an endothelial tubule formation assay. Treatment of MG63 cells with 20 ng/mL rhVEGF-A165 rescued production in silenced cells and increased production of osteocalcin, osteoprotegerin, FGF-2, and angiopoietin-1, with greatest effects on control cells cultured on modSLA. Addition of a neutralization antibody against VEGF receptor 2 (VEGFR2; Flk-1) resulted in a significant increase in VEGF-A production. Overall, this study indicates that VEGF-A has two roles in osseointegration: enhanced angiogenesis and an autocrine/paracrine role in maturation of osteoblast-like cells in response to Ti surface properties. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 423-433, 2019.


Subject(s)
Bone Substitutes/chemistry , Neovascularization, Physiologic , Osteoblasts/cytology , Titanium/chemistry , Vascular Endothelial Growth Factor A/metabolism , Autocrine Communication , Biocompatible Materials/chemistry , Cell Line , Humans , Osseointegration , Osteoblasts/metabolism , Osteogenesis , Paracrine Communication , Surface Properties
16.
Biol Sex Differ ; 9(1): 30, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29970177

ABSTRACT

BACKGROUND: Osseointegration is dependent on the implant surface, surrounding bone quality, and the systemic host environment, which can differ in male and female patients. Titanium (Ti) implants with microstructured surfaces exhibit greater pullout strength when compared to smooth-surfaced implants and exhibit enhanced osteogenic cellular responses in vitro. Previous studies showed that 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3] has a greater effect on rat osteoblast differentiation on microstructured Ti compared to smooth Ti surfaces and tissue culture polystyrene (TCPS). The stimulatory effect of 17ß-estradiol (E2) on differentiation is observed in female osteoblasts on micro-rough Ti, but it is not known if male osteoblasts behave similarly in response to E2 and microtopography. This study assessed whether human male and female osteoblasts exhibit sex-specific differences in response to E2 and 1α,25(OH)2D3 when cultured on microstructured Ti surfaces. METHODS: Osteoblasts from three male and three female human donors were cultured on Ti discs with varying surface profiles: a smooth pretreatment (PT), a coarse grit-blasted/acid-etched (SLA), and an SLA surface having undergone modification in a nitrogen environment and stored in saline to maintain hydrophilicity (modSLA). Cells cultured on these surfaces were treated with E2 or 1α,25(OH)2D3. RESULTS: Male and female human osteoblasts responded similarly to microstructure although there were donor-specific differences; cell number decreased, and osteocalcin (OCN), osteoprotegerin (OPG), and latent and active transforming growth factor 1 increased on SLA and modSLA compared to TCPS. Female osteoblasts had higher alkaline phosphatase activity and OCN production than male counterparts but produced less OPG. Both sexes responded similarly to 1α,25(OH)2D3. E2 treatment reduced cell number and increased osteoblast differentiation and factor production only in female cells. CONCLUSIONS: Male and female human osteoblasts respond similarly to microstructure and 1α,25(OH)2D3 but exhibit sexual dimorphism in substrate-dependent responses to E2. E2 affected female osteoblasts, suggesting that signaling is sex-specific and surface-dependent. Donor osteoblasts varied in response, demonstrating the need to test multiple donors when examining human samples. Understanding how male and female cells respond to orthopedic biomaterials will enable greater predictability post-implantation as well as therapies that are more patient-specific.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Osteoblasts/drug effects , Sex Characteristics , Titanium , Biocompatible Materials , Cells, Cultured , Humans , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteoprotegerin/metabolism , Surface Properties , Vitamin D/analogs & derivatives , Vitamin D/pharmacology
17.
Sci Rep ; 8(1): 8588, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29872092

ABSTRACT

Successful osseointegration of an endosseous implant involves migration and differentiation of mesenchymal stem cells (MSCs) on the implant surface. Micro-structured, hydrophilic titanium surfaces direct MSCs to undergo osteoblastic differentiation in vitro, in the absence of media additives commonly used in cultures grown on tissue culture polystyrene (TCPS). This process involves non-canonical Wnt5a, in contrast to canonical Wnt3a typically credited with osteoblastic differentiation on TCPS. Wnt proteins have been implicated in morphological development and tissue patterning, suggesting that additional Wnts may participate. Here, we demonstrate that Wnt11 is a mediator of osteoblast commitment of MSCs, and increases in a surface-roughness dependent manner. Experiments using cells silenced for Wnt11 indicate that cross-talk between Wnt5a and Wnt11 occurs. Wnt11 potentially acts upstream to Wnt5a, increasing Wnt5a expression and factors associated with osteogenesis. Thus, Wnt11 contributes to peri-implant bone formation distal to the implant surface through a heavily regulated signaling cascade of autocrine/paracrine proteins.


Subject(s)
Cell Differentiation/genetics , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Titanium/chemistry , Wnt Proteins/genetics , Cells, Cultured , Gene Expression , Humans , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , RNA Interference , Surface Properties , Wnt Proteins/metabolism , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
18.
Acta Biomater ; 68: 296-307, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29292169

ABSTRACT

A critical stage during osseointegration of a titanium (Ti) implant is primary bone remodeling, which involves cross talk among osteoclast precursors, osteoclasts, mesenchymal stem cells (MSCs), and osteoblasts. This phase couples the processes of bone formation and resorption. During remodeling, osteoclasts produce factors capable of regulating MSC migration and osteogenesis. Furthermore, they degrade primary bone, creating a foundation with a specific chemistry, stiffness, and morphology for osteoblasts to synthesize and calcify their matrix. MSCs and osteoblasts receiving cues from the implant surface produce factors capable of regulating osteoclasts in order to promote net new bone formation. The purpose of this study was to determine the effects Ti implant surfaces have on bone remodeling. Human MSCs and normal human osteoblasts (NHOsts) were cultured separately on 15 mm grade 2 smooth PT, hydrophobic-microrough SLA, hydrophilic-microrough Ti (mSLA) (Institut Straumann AG, Basel, Switzerland), or tissue culture polystyrene (TCPS). After 7d, conditioned media from surface cultures were used to treat human osteoclasts for 2d. Activity was measured by fluorescence of released collagen followed by mRNA quantification. This study demonstrates that MSC and NHOst cultures are able to suppress osteoclast activity in a surface dependent manner and osteoclast mRNA levels are selectively regulated by surface treatments. The substrate-dependent regulatory effect was mitigated when MSCs were silenced for integrin subunits and when conditioned media were denatured. These results indicate that MSCs and NHOsts regulate at least two aspects of remodeling: reduced fusion of new osteoclasts and reduced activity of existing osteoclasts. STATEMENT OF SIGNIFICANCE: In this study, we developed a novel in vitro model to study how microstructured and hydrophilic titanium implants impact bone remodeling for dental and orthopaedic applications. Our approach intersects biomaterials and systems physiology, revealing for the first time that implant surface properties are capable of regulating the communication among the cells involved in remodeling of primary bone during osseointegration. We believe that the basic research presented in our manuscript will provide important knowledge in our understanding of factors that impact implant success. Furthermore, it provides a solid foundation for the development of materials that enable rapid osseointegration and earlier loading times for implants in bone that has been compromised by trauma or disease.


Subject(s)
Cell Lineage , Osteoblasts/cytology , Osteoclasts/cytology , Prostheses and Implants , Titanium/pharmacology , Cell Lineage/drug effects , Cells, Cultured , Humans , Integrins/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/pharmacology , Phenotype , Surface Properties , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...