Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Biomaterials ; 312: 122739, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39096840

RÉSUMÉ

The biofilm-induced "relatively immune-compromised zone" creates an immunosuppressive microenvironment that is a significant contributor to refractory infections in orthopedic endophytes. Consequently, the manipulation of immune cells to co-inhibit or co-activate signaling represents a crucial strategy for the management of biofilm. This study reports the incorporation of Mn2+ into mesoporous dopamine nanoparticles (Mnp) containing the stimulator of interferon genes (STING) pathway activator cGAMP (Mncp), and outer wrapping by M1-like macrophage cell membrane (m-Mncp). The cell membrane enhances the material's targeting ability for biofilm, allowing it to accumulate locally at the infectious focus. Furthermore, m-Mncp mechanically disrupts the biofilm through photothermal therapy and induces antigen exposure through photodynamic therapy-generated reactive oxygen species (ROS). Importantly, the modulation of immunosuppression and immune activation results in the augmentation of antigen-presenting cells (APCs) and the commencement of antigen presentation, thereby inducing biofilm-specific humoral immunity and memory responses. Additionally, this approach effectively suppresses the activation of myeloid-derived suppressor cells (MDSCs) while simultaneously boosting the activity of T cells. Our study showcases the efficacy of utilizing m-Mncp immunotherapy in conjunction with photothermal and photodynamic therapy to effectively mitigate residual and recurrent infections following the extraction of infected implants. As such, this research presents a viable alternative to traditional antibiotic treatments for biofilm that are challenging to manage.

2.
Int J Nanomedicine ; 19: 7509-7527, 2024.
Article de Anglais | MEDLINE | ID: mdl-39071503

RÉSUMÉ

Purpose: Skin injury, often caused by physical or medical mishaps, presents a significant challenge as wound healing is critical to restore skin integrity and tissue function. However, external factors such as infection and inflammation can hinder wound healing, highlighting the importance of developing biomaterials with antibiotic and wound healing properties to treat infections and inflammation. In this study, a novel photothermal nanomaterial (MMPI) was synthesized for infected wound healing by loading indocyanine green (ICG) on magnesium-incorporated mesoporous bioactive glass (Mg-MBG) and coating its surface with polydopamine (PDA). Results: In this study, Mg-MBG and MMPI was synthesized via the sol-gel method and characterized it using various techniques such as scanning electron microscopy (SEM), the energy dispersive X-ray spectrometry (EDS) system and X-ray diffraction (XRD). The cytocompatibility of MMPI was evaluated by confocal laser scanning microscopy (CLSM), CCK8 assay, live/dead staining and F-actin staining of the cytoskeleton. The antibacterial efficiency was assessed using bacterial dead-acting staining, spread plate method (SPM) and TEM. The impact of MMPI on macrophage polarization was initially evaluated through flow cytometry, qPCR and ELISA. Additionally, an in vivo experiment was performed on a mouse model with skin excision infected. Histological analysis and RNA-seq analysis were utilized to analyze the in vivo wound healing and immunomodulation effect. Conclusion: Collectively, the new photothermal and photodynamic nanomaterial (MMPI) can achieve low-temperature antibacterial activity while accelerating wound healing, holds broad application prospects.


Sujet(s)
Antibactériens , Vert indocyanine , Indoles , Photothérapie dynamique , Thérapie photothermique , Espèces réactives de l'oxygène , Cicatrisation de plaie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Animaux , Souris , Vert indocyanine/composition chimique , Vert indocyanine/pharmacologie , Photothérapie dynamique/méthodes , Antibactériens/pharmacologie , Antibactériens/composition chimique , Thérapie photothermique/méthodes , Indoles/composition chimique , Indoles/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Polymères/composition chimique , Polymères/pharmacologie , Staphylococcus aureus/effets des médicaments et des substances chimiques , Staphylococcus aureus/physiologie , Cellules RAW 264.7 , Peau/effets des médicaments et des substances chimiques , Nanostructures/composition chimique , Rayons infrarouges
3.
Colloids Surf B Biointerfaces ; 241: 114042, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38924850

RÉSUMÉ

In the field of orthopedics, surgeons have long been facing the challenge of loosening of external fixation screws due to inherent material characteristics. Despite Polyetheretherketone (PEEK) being employed as an orthopedic implant material for many years, its bio-inert nature often hinders bone healing due to the limited bioactivity, which restricts its clinical applications. Herein, a new type of orthopedic implant (Sr-SPK) was developed by introducing strontium (Sr)-doped mesoporous bioactive glass (Sr-MBG) onto the surface of PEEK implants through a simple and feasible method. In vitro experiments revealed that Sr-SPK effectively promotes osteogenic differentiation while concurrently suppressing the formation of osteoclasts. The same results were validated in vivo with Sr-SPK significantly improving bone integration. Upon investigation, it was found that Sr-SPK promotes adhesion among bone marrow mesenchymal stem cells (BMSCs) thereby promoting osteogenesis by activating the regulation of actin cytoskeletal and focal adhesion pathways, as identified via transcriptome analysis. In essence, these findings suggest that the newly constructed Sr-doped biofunctionalized PEEK implant developed in this research can promote osteoblast differentiation and suppress osteoclast activity by enhancing cell adhesion processes. These results underline the immense potential of such an implant for wide-ranging clinical applications in orthopedics.


Sujet(s)
Benzophénones , Adhérence cellulaire , Verre , Cétones , Cellules souches mésenchymateuses , Ostéo-intégration , Ostéogenèse , Polyéthylène glycols , Polymères , Strontium , Strontium/pharmacologie , Strontium/composition chimique , Ostéo-intégration/effets des médicaments et des substances chimiques , Polymères/composition chimique , Polymères/pharmacologie , Adhérence cellulaire/effets des médicaments et des substances chimiques , Cétones/composition chimique , Cétones/pharmacologie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Animaux , Ostéogenèse/effets des médicaments et des substances chimiques , Verre/composition chimique , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Propriétés de surface , Matériaux biocompatibles/pharmacologie , Matériaux biocompatibles/composition chimique , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Ostéoclastes/cytologie , Souris , Cellules cultivées , Taille de particule
4.
ISA Trans ; 148: 191-200, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38548502

RÉSUMÉ

In this paper, the secure containment control issue for heterogeneous multi-agent systems subject to hybrid attacks is studied. In the network channels among agents, adversaries launch replay attacks such that the followers can only retrieve the previous information of their neighbors. To characterize the effects of replay attacks, a distributed auxiliary system with heterogeneous time-varying delay is constructed to estimate the convex hull of the leaders' state. The control input of each follower may be contaminated by the malicious false data injection (FDI) attacks with unknown bounded time-varying signals, which are intended to compromise the containment performance of the whole system. A local adaptive compensator is designed for each follower to eliminate the adverse effects of the FDI attacks. Based on the distributed auxiliary system and the local adaptive compensator, a secure distributed control protocol is proposed to ensure that the output trajectories of each follower can converge to the dynamic convex hull spanned by the leader outputs. Finally, two illustrative examples are provided to show the feasibility of the proposed secure control scheme.

5.
J Biomed Mater Res A ; 112(9): 1565-1577, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38514993

RÉSUMÉ

Polyether ether ketone (PEEK) is gaining recognition as a highly promising polymer for orthopedic implants, attributed to its exceptional biocompatibility, ease of processing, and radiation resistance. However, its long-term in vivo application faces challenges, primarily due to suboptimal osseointegration from postimplantation inflammation and immune reactions. Consequently, biofunctionalization of PEEK implant surfaces emerges as a strategic approach to enhance osseointegration and increase the overall success rates of these implants. In our research, we engineered a multifaceted PEEK implant through the in situ integration of chitosan-coated zinc-doped bioactive glass nanoparticles (Zn-BGNs). This novel fabrication imbues the implant with immunomodulatory capabilities while bolstering its osseointegration potential. The biofunctionalized PEEK composite elicited several advantageous responses; it facilitated M2 macrophage polarization, curtailed the production of inflammatory mediators, and augmented the osteogenic differentiation of bone marrow mesenchymal stem cells. The experimental findings underscore the vital and intricate role of biofunctionalized PEEK implants in preserving normal bone immunity and metabolism. This study posits that utilizing chitosan-BGNs represents a direct and effective method for creating multifunctional implants. These implants are designed to facilitate biomineralization and immunomodulation, making them especially apt for orthopedic applications.


Sujet(s)
Benzophénones , Régénération osseuse , Cétones , Cellules souches mésenchymateuses , Polyéthylène glycols , Polymères , Zinc , Polymères/composition chimique , Polyéthylène glycols/composition chimique , Régénération osseuse/effets des médicaments et des substances chimiques , Animaux , Cétones/composition chimique , Cétones/pharmacologie , Zinc/composition chimique , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Souris , Chitosane/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Verre/composition chimique , Cellules RAW 264.7 , Différenciation cellulaire/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique
6.
J Nanobiotechnology ; 22(1): 80, 2024 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-38418972

RÉSUMÉ

The advancement of biomaterials with antimicrobial and wound healing properties continues to present challenges. Macrophages are recognized for their significant role in the repair of infection-related wounds. However, the interaction between biomaterials and macrophages remains complex and requires further investigation. In this research, we propose a new sequential immunomodulation method to enhance and expedite wound healing by leveraging the immune properties of bacteria-related wounds, utilizing a novel mixed hydrogel dressing. The hydrogel matrix is derived from porcine acellular dermal matrix (PADM) and is loaded with a new type of bioactive glass nanoparticles (MBG) doped with magnesium (Mg-MBG) and loaded with Curcumin (Cur). This hybrid hydrogel demonstrates controlled release of Cur, effectively eradicating bacterial infection in the early stage of wound infection, and the subsequent release of Mg ions (Mg2+) synergistically inhibits the activation of inflammation-related pathways (such as MAPK pathway, NF-κB pathway, TNF-α pathway, etc.), suppressing the inflammatory response caused by infection. Therefore, this innovative hydrogel can safely and effectively expedite wound healing during infection. Our design strategy explores novel immunomodulatory biomaterials, offering a fresh approach to tackle current clinical challenges associated with wound infection treatment.


Sujet(s)
Anti-infectieux , Curcumine , Infection de plaie , Animaux , Suidae , Hydrogels/pharmacologie , Cicatrisation de plaie , Biomimétique , Bandages , Antibactériens/usage thérapeutique , Matériaux biocompatibles , Immunothérapie , Infection de plaie/traitement médicamenteux
7.
Adv Sci (Weinh) ; 11(5): e2302674, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38037309

RÉSUMÉ

Developing biomaterials with antimicrobial and wound-healing activities for the treatment of wound infections remains challenging. Macrophages play non-negligible roles in healing infection-related wounds. In this study, a new sequential immunomodulatory approach is proposed to promote effective and rapid wound healing using a novel hybrid hydrogel dressing based on the immune characteristics of bacteria-associated wounds. The hydrogel dressing substrate is derived from a porcine dermal extracellular matrix (PADM) and loaded with a new class of bioactive glass nanoparticles (BGns) doped with copper (Cu) and zinc (Zn) ions (Cu-Zn BGns). This hybrid hydrogel demonstrates a controlled release of Cu2+ and Zn2+ and sequentially regulates the phenotypic transition of macrophages from M1 to M2 by alternately activating nucleotide-binding oligomerization domain (NOD) and inhibiting mitogen-activated protein kinases (MAPK) signaling pathways. Additionally, its dual-temporal bidirectional immunomodulatory function facilitates enhanced antibacterial activity and wound healing. Hence, this novel hydrogel is capable of safely and efficiently accelerating wound healing during infections. As such, the design strategy provides a new direction for exploring novel immunomodulatory biomaterials to address current clinical challenges related to the treatment of wound infections.


Sujet(s)
Staphylococcus aureus résistant à la méticilline , Infection de plaie , Animaux , Suidae , Cuivre , Hydrogels/composition chimique , Cicatrisation de plaie , Matériaux biocompatibles/composition chimique , Infection de plaie/traitement médicamenteux , Zinc
8.
Adv Healthc Mater ; 12(32): e2301772, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37723927

RÉSUMÉ

Key factors contributing to implantation failures include implant-associated infections (IAIs) and insufficient osseointegration of the implants. Polyetheretherketone (PEEK) is widely used in orthopedics, yet its clinical applications are restricted due to its poor osteogenic and antibacterial properties as well as inadequate immune responses. To overcome these drawbacks, a novel spatiotemporal immunomodulation approach is proposed, chelating Cu-Sr bilayer bioactive glass nanoparticles (CS-BGNs) onto the PEEK surface via polydopamine (PDA). The CS-BGNs possess a bilayer core-shell structure where copper is distributed in the outer layer and strontium is clustered in the inner layer. The results show that CS-BGNs/PDA functionalized PEEK demonstrates a controlled and sequential release of Cu2+ and Sr2+ . In the early stage, Cu2+ from the outer layer releases rapidly, while Sr2+ from the inner layer releases in the late stage. This well-ordered release pattern modulates the phenotypic transition of macrophages, which induces M1 polarization in the early stage and M2 polarization in the late stage. Combined with the direct effects of Cu2+ and Sr2+ , the spatiotemporal immunomodulation not only benefits the early antibacterial and tissue-healing process, but also promotes the long-term process of osseointegration, providing new perspectives on the design of novel immunomodulatory biomaterials.


Sujet(s)
Cuivre , Nanoparticules , Cuivre/pharmacologie , Cuivre/composition chimique , Ostéogenèse , Polyéthylène glycols/composition chimique , Cétones/pharmacologie , Cétones/composition chimique , Nanoparticules/composition chimique , Antibactériens/pharmacologie , Antibactériens/composition chimique , Ostéo-intégration , Propriétés de surface
9.
Colloids Surf B Biointerfaces ; 230: 113523, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37634286

RÉSUMÉ

Polyetheretherketone (PEEK) is being increasingly recognized as a highly promising polymer implant in orthopaedics due to its advantageous biocompatibility, favorable processability, and radiation resistance. Nonetheless, the long-term application of PEEK implants in vivo faces challenges due to unfavorable post-implantation inflammatory and immune reactions, which result in suboptimal osseointegration rates. Hence, biofunctionalizing the surface of PEEK implants emerges as a viable strategy to enhance osseointegration and increase the success rate. In this study, we developed a multifunctional PEEK implant through the in-situ incorporation of chitosan-coated bioactive glass nanoparticles (BGNs). This approach can impart immunomodulatory properties and enhance the potential for osseointegration. The resulting biofunctionalized PEEK material exhibited multiple beneficial effects. For instance, it facilitated M2 phenotypic polarization of macrophages, diminished the expression of inflammatory factors, and enhanced the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro. Moreover, it exhibited an improved capacity for osseointegration when tested in vivo. The findings of the experiment highlighted the pivotal and complex role of the biofunctionalized PEEK implant in maintaining typical bone immunity and metabolism. The study proposes that the application of chitosan-BGNs presents a straightforward approach to developing multifunctional implants with the ability to promote biomineralization and immunomodulation, specifically tailored for orthopaedic applications.


Sujet(s)
Chitosane , Magnésium , Ostéogenèse , Régénération osseuse , Polyéthylène glycols/pharmacologie , Différenciation cellulaire
10.
Oxid Med Cell Longev ; 2022: 3458560, 2022.
Article de Anglais | MEDLINE | ID: mdl-36164394

RÉSUMÉ

Osteoporosis (OP) is one of the most common bone disorders among the elderly, characterized by abnormally elevated bone resorption caused by formation and activation of osteoblast (OC). Excessive reactive oxygen species (ROS) accumulation might contribute to the formation process of OC as an essential role. Although accumulated advanced treatment target on OP have been proposed in recent years, clinical outcomes remain unexcellence attributed to severe side effects. The purpose of present study was to explore the underlying mechanisms of GSK 650394 (GSK) on inhibiting formation and activation of OC and bone resorption in vitro and in vivo. GSK could inhibit receptor activator of nuclear-κB ligand (RANKL-)-mediated Oc formation via suppressing the activation of NF-κB and MAPK signaling pathways, regulating intracellular redox status, and downregulate the expression of nuclear factor of activated T cells c1 (NFATc1). In addition, quantitative RT-PCR results show that GSK could suppress the expression of OC marker gene and antioxidant enzyme genes. Consistent with in vitro cellular results, GSK treatment improved bone density in the mouse with ovariectomized-induced bone loss according to the results of CT parameters, HE staining, and Trap staining. Furthermore, GSK treatment could enhance the capacity of antioxidant enzymes in vivo. In conclusion, this study suggested that GSK could suppress the activation of osteoclasts and therefore maybe a potential therapeutic reagent for osteoclast activation-related osteoporosis.


Sujet(s)
Benzoates , Résorption osseuse , Composés hétérocycliques bicycliques , Ostéoclastes , Ostéoporose , Animaux , Benzoates/pharmacologie , Benzoates/usage thérapeutique , Résorption osseuse/métabolisme , Composés hétérocycliques bicycliques/pharmacologie , Composés hétérocycliques bicycliques/usage thérapeutique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Ligands , Souris , Souris de lignée C57BL , Facteur de transcription NF-kappa B/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéoporose/traitement médicamenteux , Ostéoporose/prévention et contrôle , Ligand de RANK/métabolisme , Espèces réactives de l'oxygène/métabolisme
11.
Bioact Mater ; 16: 134-148, 2022 Oct.
Article de Anglais | MEDLINE | ID: mdl-35386313

RÉSUMÉ

Orthopedic implants account for 99% of orthopedic surgeries, however, orthopedic implant-related infection is one of the most serious complications owing to the potential for limb-threatening sequelae and mortality. Current antibiotic treatments still lack the capacity to target bone infection sites, thereby resulting in unsatisfactory therapeutic effects. Here, the bone infection site targeting efficacy of D6 and UBI29-41 peptides was investigated, and bone-and-bacteria dual-targeted nanoparticles (NPs) with D6 and UBI29-41 peptides were first fabricated to target bone infection site and control the release of vancomycin in bone infection site. The results of this study demonstrated that the bone-and-bacteria dual-targeted mesoporous silica NPs exhibit excellent bone and bacteria targeting efficacy, excellent biocompatibility and effective antibacterial properties in vitro. Furthermore, in a rat model of orthopedic implant-related infection with methicillin-resistant Staphylococcus aureus, the growth of bacteria was evidently inhibited without cytotoxicity, thus realizing the early treatment of implant-related infection. Hence, the bone-and-bacteria dual-targeted molecule-modified NPs may target bacteria-infected bone sites and act as ideal candidates for the therapy of orthopedic implant-related infections.

12.
J Control Release ; 343: 314-325, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35085700

RÉSUMÉ

Rheumatoid arthritis (RA) is a common inflammatory disease and its treatment is largely limited by drug ineffectiveness or severe side effects. In RA progression, multiple signalling pathways, such as hypoxia-inducible factor (HIF)-1α, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) pathways, act synergistically to maintain the inflammatory response. To downregulate HIF-1α, NF-κB, and MAPK expression, we proposed HIF-1α siRNA-loaded calcium phosphate nanoparticles encapsulated in apolipoprotein E3-reconstituted high-density lipoprotein (HIF-CaP-rHDL) for RA therapy. Here, we evaluated the potential of CaP-rHDL nanoparticles in RA therapy using a murine macrophage line (RAW 264.7) and a collagen-induced arthritis (CIA) mouse model. The CaP-rHDL nanoparticles showed significant anti-inflammatory effects along with HIF-1α knockdown and NF-κB and MAPK signalling pathway inhibition in lipopolysaccharide-activated macrophages. Moreover, they inhibited receptor activator of NF-κB ligand (RANKL)-induced osteoclast formation. In CIA mice, their intravenous administration resulted in high accumulation at the arthritic joint sites, and HIF-CaP-rHDL effectively suppressed inflammatory cytokine secretion and relieved bone erosion, cartilage damage, and osteoclastogenesis. Thus, HIF-CaP-rHDL demonstrated great potential in RA precision therapy by inhibiting multiple inflammatory signalling pathways.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Nanoparticules , Animaux , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Souris , Facteur de transcription NF-kappa B , Petit ARN interférent/génétique , Petit ARN interférent/usage thérapeutique
13.
Front Bioeng Biotechnol ; 9: 694635, 2021.
Article de Anglais | MEDLINE | ID: mdl-34589470

RÉSUMÉ

Biofilms refer to complex bacterial communities that are attached to the surface of animate or inanimate objects, which highly resist the antibiotics or the host immune defense mechanisms. Pathogenic biofilms in medicine are general, chronic, and even costly, especially on medical devices and orthopedic implants. Bacteria within biofilms are the cause of many persistent infections, which are almost impossible to eradicate. Though some progress has been made in comprehending the mechanisms of biofilm formation and persistence, novel alternative compounds or strategies and effective anti-biofilm antibiotics are still lacking. Smart materials of nano size which are able to respond to an external stimulus or internal environment have a great range of applications in clinic. Recently, smart nanomaterials with or without carriage of antibiotics, targeting specific bacteria and biofilm under some stimuli, have shown great potential for pathogenic biofilm and resident bacteria eradication. First, this review briefly summarizes and describes the significance of biofilms and the process of biofilm formation. Then, we focus on some of the latest research studies involving biofilm elimination, which probably could be applied in orthopedic implants. Finally, some outstanding challenges and limitations that need to be settled urgently in order to make smart nanomaterials effectively target and treat implant biofilms are also discussed. It is hoped that there will be more novel anti-biofilm strategies for biofilm infection in the prospective future.

15.
Int Immunopharmacol ; 94: 107458, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-33626422

RÉSUMÉ

Osteoporosis is one of the most common metabolic bone diseases among pre- and post-menopausal women. Despite numerous advances in the treatment of osteoporosis in recent years, the outcomes remain poor due to severe side effects. In this study, we investigated whether A-485, a highly selective catalytic p300/CBP inhibitor, could attenuate RANKL-induced osteoclast differentiation and explored the underlying molecular mechanisms. The protective role of A-485 in osteoporosis was verified using a mouse model of ovariectomy (OVX)-induced bone loss and micro-CT scanning. A-485 inhibited RANKL-induced osteoclast differentiation in vitro by reducing the number of tartrate-resistant acid phosphatase-positive osteoclasts without inducing significant cytotoxicity. In particular, A-485 dose-dependently disrupted F-actin ring formation and downregulated the expression of genes associated with osteoclast differentiation, such as CTSK, c-Fos, TRAF6, VATPs-d2, DC-STAMP, and NFATc1, in a time- and dose-dependent manner. Moreover, A-485 inhibited the RANKL-induced phosphorylation of MAPK pathways and attenuated OVX-induced bone loss in the mouse model while rescuing the loss of bone mineral density. Our in vitro and in vivo findings suggest for the first time that A-485 has the potential to prevent postmenopausal osteoporosis and could therefore be considered as a therapeutic molecule against osteoporosis.


Sujet(s)
Agents de maintien de la densité osseuse/usage thérapeutique , Résorption osseuse/traitement médicamenteux , Protéine p300-E1A/antagonistes et inhibiteurs , Composés hétérocycliques avec 4 noyaux ou plus/usage thérapeutique , Protéines membranaires/antagonistes et inhibiteurs , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoporose/traitement médicamenteux , Phosphoprotéines/antagonistes et inhibiteurs , Animaux , Agents de maintien de la densité osseuse/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Femelle , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Souris de lignée C57BL , Ostéoclastes/physiologie , Ovariectomie , Rats
16.
J Biomed Mater Res A ; 109(6): 879-892, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-32780520

RÉSUMÉ

Due to its excellent mechanical and low-friction properties, polyetheretherketone (PEEK) has been widely investigated for use in orthopedic applications over the past decade. However, the bioinertness and poor osteogenic properties of PEEK have hampered its clinical application. In this study, the surface of PEEK was modified by co-treatment with hydrofluoric acid and nitric acid (AFN). The microstructures of the modified PEEK surfaces were investigated using scanning electron microscopy. The water contact angles of the surfaces were also measured. To evaluate their cytocompatibility, PEEK samples were used as substrates to culture rat bone mesenchymal stem cells, and cell adhesion, viability, and expression of specific marker genes were measured. Treatment of PEEK with AFN (PEEK-AFN) was found to enable better osteoblast adhesion, spreading, and proliferation; the activity of alkaline phosphatase (an early osteogenic differentiation marker) was also found to be enhanced post-treatment. Furthermore, PEEK-AFN was able to modulate macrophage polarization and down regulated the expression of proinflammatory factors via inhibiting the NF-κB pathway. Thus, treatment of PEEK with AFN could promote M2 polarization of the macrophages and stimulate the differentiation of osteoblasts. These results provide valuable information that could facilitate the use of PEEK-based composites as bone implant materials.


Sujet(s)
Benzophénones/composition chimique , Matériaux biocompatibles/composition chimique , Acide fluorhydrique/composition chimique , Macrophages/effets des médicaments et des substances chimiques , Acide nitrique/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Polymères/composition chimique , Agents angiogéniques/pharmacologie , Animaux , Marqueurs biologiques , Calcification physiologique/effets des médicaments et des substances chimiques , Adhérence cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire , Cellules souches mésenchymateuses , Microscopie électronique à balayage , Rats , Propriétés de surface
17.
ACS Infect Dis ; 6(12): 3147-3162, 2020 12 11.
Article de Anglais | MEDLINE | ID: mdl-33236626

RÉSUMÉ

Staphylococcus aureus is a primary pathogen responsible for causing postoperative infections as it survives and persists in host cells, including osteoblasts and macrophages. These cells then serve as reservoirs resulting in chronic infections. Most traditional antibiotics have poor effects on intracellular S. aureus because they cannot enter the cell. Herein, a cell-penetrating peptide TAT-KR-12 was derived from the trans-activating transcription (TAT) peptide and KR-12 (residues 18-29 of human cathelicidin LL-37). The TAT acts as a "trojan horse" to deliver KR-12 peptide into the cells to kill S. aureus. Moreover, effective antibacterial properties and biocompatibility were observed in vitro, demonstrating that TAT-KR-12 is effective not only in eliminating planktonic S. aureus, but also in eliminating intracellular S. aureus cells in vitro. TAT-KR-12, as with LL-37, also elicits strong anti-inflammatory activities in LPS-stimulated macrophages, as demonstrated by significant inhibition of NO, TNF-α, and IL-1ß expression and secretion from LPS-stimulated RAW264.7 cells. In the subcutaneous infection mouse model of planktonic and intracellular infections, the growth of S. aureusin vivo is evidently inhibited without cytotoxicity. These results suggest that the novel antimicrobial TAT-KR-12 may prove to be an effective treatment option to overcome antibiotic resistance caused by intracellular bacterial infections.


Sujet(s)
Peptides de pénétration cellulaire , Infections à staphylocoques , Staphylococcus aureus , Plancton , Perforines , Infections à staphylocoques/traitement médicamenteux , Staphylococcus aureus/pathogénicité
18.
World J Stem Cells ; 12(7): 545-561, 2020 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-32843913

RÉSUMÉ

Bone marrow-derived mesenchymal stem cells (BMSCs) play a critical role in the osseointegration of bone and orthopedic implant. However, osseointegration between the Ti-based implants and the surrounding bone tissue must be improved due to titanium's inherent defects. Surface modification stands out as a versatile technique to create instructive biomaterials that can actively direct stem cell fate. Here, we summarize the current approaches to promoting BMSC osteogenesis on the surface of titanium and its alloys. We will highlight the utilization of the unique properties of titanium and its alloys in promoting tissue regeneration, and discuss recent advances in understanding their role in regenerative medicine. We aim to provide a systematic and comprehensive review of approaches to promoting BMSC osteogenesis on the orthopedic implant surface.

19.
J Orthop Translat ; 21: 66-72, 2020 Mar.
Article de Anglais | MEDLINE | ID: mdl-32099806

RÉSUMÉ

BACKGROUND/OBJECTIVE: Orthopaedic implants are important devices aimed at relieving pain and improving mobility. Staphylococcal infection and aseptic loosening are two common events associated with inflammatory osteolysis that lead to implant failures. Bone mass and biomechanical properties are important indicators that could influence patient outcomes after revision surgery. However, the dynamics of bacterial infections and their influence on bone mass and biomechanical properties remain unclear. Hence, in this study, we developed rabbit aseptic inflammation and staphylococcal infection models to determine the effects of coagulase-positive and coagulase-negative bacterial infection, as well as aseptic inflammation, on the mass and biomechanical properties of the bone. METHODS: Sixty New Zealand white rabbits were randomly assigned to 6 groups, and each group had 10 rabbits. The medullary cavities in rabbits of each group were injected with phosphate-buffered saline (100 â€‹µL), titanium (Ti)-wear particles (300 µg/100 â€‹µL), a low concentration of Staphylococcus epidermidis (105/100 â€‹µL), a high concentration of S. epidermidis (108/100 â€‹µL), a low concentration of Staphylococcus aureus (105/100 â€‹µL), and a high concentration of S. aureus (108/100 â€‹µL), respectively. At four and eight weeks after surgery, the rabbits were sacrificed, and the tibias on the surgical side were analysed via histopathology, microcomputed tomography, and nanoindentation testing. RESULTS: Histopathological analysis demonstrated that inflammatory responses and bacterial loads caused by high concentrations of staphylococcal infections, particularly coagulase-positive staphylococci, are more detrimental than low concentrations of bacterial infection and Ti-wear particles. Meanwhile, microcomputed tomography and nanoindentation testing showed that high concentrations of S. aureus caused the highest loss in bone mass and most biomechanical function impairment in rabbits experiencing aseptic inflammation and staphylococcal infections. CONCLUSIONS: Inflammatory osteolysis caused by a high concentration of coagulase-positive staphylococci is significantly associated with low bone mass and impaired biomechanical properties. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: It is necessary to obtain an overall assessment of the bone mass and biomechanical properties before revision surgery, especially when S. aureus infection is involved. In addition, a better understanding of these two parameters might help develop a reasonable treatment regimen and reduce the risk of adverse events after revision surgery.

20.
Exp Cell Res ; 387(2): 111800, 2020 02 15.
Article de Anglais | MEDLINE | ID: mdl-31877305

RÉSUMÉ

Postmenopausal osteoporosis is one of the most common types of osteoporosis resulting from estrogen deficiency in elderly women. In addition, hypertension is another common disease in the elderly, and it has become an independent risk factor for osteoporosis and osteoporotic fractures. Here, we report for the first time that felodipine, a first-line antihypertensive agent, significantly prevents postmenopausal osteoporosis in addition to its vasodilation properties. Quantitative RT-PCR analysis revealed that treatment with felodipine significantly downregulated the genes associated with osteoclast differentiation. RNA-sequencing and western blotting suggested that felodipine could inhibit bone resorption by suppressing MAPK pathway phosphorylation. Moreover, micro-CT scanning and histological analysis in an ovariectomy (OVX)-induced bone-loss mouse model indicated that felodipine might be a potent drug for preventing osteoporotic fractures. Therefore, this study proposes an attractive and promising agent with vasodilation properties to treat postmenopausal osteoporosis.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Oestrogènes/métabolisme , Félodipine/pharmacologie , Mitogen-Activated Protein Kinases/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoporose post-ménopausique/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Maladies osseuses métaboliques/traitement médicamenteux , Maladies osseuses métaboliques/métabolisme , Résorption osseuse/traitement médicamenteux , Résorption osseuse/métabolisme , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Souris de lignée C57BL , Ostéoclastes/métabolisme , Ostéoporose post-ménopausique/métabolisme , Ovariectomie/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE