Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36.521
Filtrar
1.
Int J Implant Dent ; 10(1): 35, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38967690

RESUMO

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.


Assuntos
Implantes Dentários , Osseointegração , Osteoblastos , Propriedades de Superfície , Osteoblastos/fisiologia , Osteoblastos/citologia , Humanos , Diferenciação Celular , Proliferação de Células , Titânio/química , Osteogênese/fisiologia
2.
Int J Mol Sci ; 25(13)2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-39000355

RESUMO

Postmenopausal osteoporosis, characterized by an imbalance between osteoclast-mediated bone resorption and osteoblast-driven bone formation, presents substantial health implications. In this study, we investigated the role of black goat extract (BGE), derived from a domesticated native Korean goat, estrogen-like activity, and osteoprotective effects in vitro. BGE's mineral and fatty acid compositions were analyzed via the ICP-AES method and gas chromatography-mass spectrometry, respectively. In vitro experiments were conducted using MCF-7 breast cancer cells, MC3T3-E1 osteoblasts, and RAW264.7 osteoclasts. BGE exhibits a favorable amount of mineral and fatty acid content. It displayed antimenopausal activity by stimulating MCF-7 cell proliferation and augmenting estrogen-related gene expression (ERα, ERß, and pS2). Moreover, BGE positively impacted osteogenesis and mineralization in MC3T3-E1 cells through Wnt/ß-catenin pathway modulation, leading to heightened expression of Runt-related transcription factor 2, osteoprotegerin, and collagen type 1. Significantly, BGE effectively suppressed osteoclastogenesis by curtailing osteoclast formation and activity in RAW264.7 cells, concurrently downregulating pivotal signaling molecules, including receptor activator of nuclear factor κ B and tumor necrosis factor receptor-associated factor 6. This study offers a shred of preliminary evidence for the prospective use of BGE as an effective postmenopausal osteoporosis treatment.


Assuntos
Diferenciação Celular , Cabras , Osteoblastos , Osteoclastos , Osteogênese , Animais , Camundongos , Células RAW 264.7 , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/citologia , Humanos , Estrogênios/farmacologia , Proliferação de Células/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Células MCF-7 , Extratos de Tecidos/farmacologia
3.
Bull Math Biol ; 86(9): 108, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39007985

RESUMO

Fibrous dysplasia (FD) is a mosaic non-inheritable genetic disorder of the skeleton in which normal bone is replaced by structurally unsound fibro-osseous tissue. There is no curative treatment for FD, partly because its pathophysiology is not yet fully known. We present a simple mathematical model of the disease incorporating its basic known biology, to gain insight on the dynamics of the involved bone-cell populations, and shed light on its pathophysiology. We develop an analytical study of the model and study its basic properties. The existence and stability of steady states are studied, an analysis of sensitivity on the model parameters is done, and different numerical simulations provide findings in agreement with the analytical results. We discuss the model dynamics match with known facts on the disease, and how some open questions could be addressed using the model.


Assuntos
Simulação por Computador , Displasia Fibrosa Óssea , Conceitos Matemáticos , Modelos Biológicos , Mutação , Humanos , Displasia Fibrosa Óssea/genética , Displasia Fibrosa Óssea/patologia , Osteoblastos/patologia
4.
Biomed Mater ; 19(5)2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39016135

RESUMO

The performance and long-term durability of dental implants hinge on the quality of bone integration and their resistance to bacteria. This research aims to introduce a surface modification strategy for zirconia implants utilizing femtosecond laser ablation techniques, exploring their impact on osteoblast cell behavior and bacterial performance, as well as the integral factors influencing the soft tissue quality surrounding dental implants. Ultrafast lasers were employed to craft nanoscale groove geometries on zirconia surfaces, with thorough analyses conducted using x-ray diffraction, scanning electron microscopy, atomic force microscopy, and water contact angle measurements. The study evaluated the response of human fetal osteoblastic cell lines to textured zirconia ceramics by assessing alkaline phosphatase activity, collagen I, and interleukin 1ßsecretion over a 7 day period. Additionally, the antibacterial behavior of the textured surfaces was investigated usingFusobacterium nucleatum, a common culprit in infections associated with dental implants. Ciprofloxacin (CIP), a widely used antibacterial antibiotic, was loaded onto zirconia ceramic surfaces. The results of this study unveiled a substantial reduction in bacterial adhesion on textured zirconia surfaces. The fine biocompatibility of these surfaces was confirmed through the MTT assay and observations of cell morphology. Moreover, the human fetal osteoblastic cell line exhibited extensive spreading and secreted elevated levels of collagen I and interleukin 1ßin the modified samples. Drug release evaluations demonstrated sustained CIP release through a diffusion mechanism, showcasing excellent antibacterial activity against pathogenic bacteria, includingStreptococcus mutans, Pseudomonas aeruginosa, andEscherichia coli.


Assuntos
Antibacterianos , Cerâmica , Lasers , Osteoblastos , Propriedades de Superfície , Zircônio , Zircônio/química , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Cerâmica/química , Cerâmica/farmacologia , Linhagem Celular , Implantes Dentários/microbiologia , Fusobacterium nucleatum/efeitos dos fármacos , Teste de Materiais , Ciprofloxacina/farmacologia , Ciprofloxacina/química , Interleucina-1beta/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Difração de Raios X , Microscopia Eletrônica de Varredura , Fosfatase Alcalina/metabolismo , Microscopia de Força Atômica , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
5.
Arch Microbiol ; 206(8): 354, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39017726

RESUMO

Titanium implants are subject to bacterial adhesion and peri-implantitis induction, and biosurfactants bring a new alternative to the fight against infections. This work aimed to produce and characterize the biosurfactant from Bacillus subtilis ATCC 19,659, its anti-adhesion and antimicrobial activity, and cell viability. Anti-adhesion studies were carried out against Streptococcus sanguinis, Staphylococcus aureus, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, and Proteus mirabilis as the minimum inhibitory concentration and the minimum bactericidal concentration. Cell viability was measured against osteoblast and fibroblast cells. The biosurfactant was classified as lipopeptide, with critical micelle concentration at 40 µg mL- 1, and made the titanium surface less hydrophobic. The anti-adhesion effect was observed for Staphylococcus aureus and Streptococcus sanguinis with 54% growth inhibition and presented a minimum inhibitory concentration of 15.7 µg mL- 1 for Streptococcus sanguinis and Aggregatibacter actinomycetemcomitans. The lipopeptide had no cytotoxic effect and demonstrated high potential application against bacterial biofilms.


Assuntos
Aderência Bacteriana , Biofilmes , Implantes Dentários , Lipopeptídeos , Testes de Sensibilidade Microbiana , Titânio , Titânio/farmacologia , Titânio/química , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Aderência Bacteriana/efeitos dos fármacos , Implantes Dentários/microbiologia , Lipopeptídeos/farmacologia , Humanos , Antibacterianos/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Bacillus subtilis/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/fisiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Propriedades de Superfície , Fibroblastos/efeitos dos fármacos , Fusobacterium nucleatum/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Tensoativos/farmacologia
6.
FASEB J ; 38(14): e23824, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39012304

RESUMO

The regenerative ability of limb bones after injury decreases during aging, but whether a similar phenomenon occurs in jawbones and whether autophagy plays a role in this process remain unclear. Through retrospective analysis of clinical data and studies on a mouse model of jawbone defects, we confirmed the presence of delayed or impaired bone regeneration in the jawbones of old individuals and mice. Subsequently, osteoblasts (OBs) derived from mouse jawbones were isolated, showing reduced osteogenesis in senescent osteoblasts (S-OBs). We observed a reduction in autophagy within both aged jawbones and S-OBs. Additionally, pharmacological inhibition of autophagy in normal OBs (N-OBs) led to cell aging and decreased osteogenesis, while autophagic activation reversed the aging phenotype of S-OBs. The activator rapamycin (RAPA) increased the autophagy level and bone regeneration in aged jawbones. Finally, we found that fatty acid-binding protein 3 (FABP3) was degraded by autolysosomes through its interaction with sequestosome 1 (P62/SQSTM1). Autophagy inhibition within senescent jawbones and S-OBs led to the excessive accumulation of FABP3, and FABP3 knockdown partially rescued the decreased osteogenesis in S-OBs and alleviated age-related compromised jawbone regeneration. In summary, we confirmed that autophagy inhibition plays an important role in delaying bone regeneration in aging jawbones. Autophagic activation or FABP3 knockdown can partially rescue the osteogenesis of S-OBs and the regeneration of aging jawbones, providing insight into jawbone aging.


Assuntos
Envelhecimento , Autofagia , Regeneração Óssea , Proteínas de Ligação a Ácido Graxo , Osteoblastos , Osteogênese , Animais , Autofagia/fisiologia , Osteoblastos/metabolismo , Camundongos , Osteogênese/fisiologia , Envelhecimento/fisiologia , Envelhecimento/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Masculino , Humanos , Camundongos Endogâmicos C57BL , Arcada Osseodentária , Feminino , Senescência Celular/fisiologia
7.
Int J Implant Dent ; 10(1): 34, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38963524

RESUMO

Dental implant therapy, established as standard-of-care nearly three decades ago with the advent of microrough titanium surfaces, revolutionized clinical outcomes through enhanced osseointegration. However, despite this pivotal advancement, challenges persist, including prolonged healing times, restricted clinical indications, plateauing success rates, and a notable incidence of peri-implantitis. This review explores the biological merits and constraints of microrough surfaces and evaluates the current landscape of nanofeatured dental implant surfaces, aiming to illuminate strategies for addressing existing impediments in implant therapy. Currently available nanofeatured dental implants incorporated nano-structures onto their predecessor microrough surfaces. While nanofeature integration into microrough surfaces demonstrates potential for enhancing early-stage osseointegration, it falls short of surpassing its predecessors in terms of osseointegration capacity. This discrepancy may be attributed, in part, to the inherent "dichotomy kinetics" of osteoblasts, wherein increased surface roughness by nanofeatures enhances osteoblast differentiation but concomitantly impedes cell attachment and proliferation. We also showcase a controllable, hybrid micro-nano titanium model surface and contrast it with commercially-available nanofeatured surfaces. Unlike the commercial nanofeatured surfaces, the controllable micro-nano hybrid surface exhibits superior potential for enhancing both cell differentiation and proliferation. Hence, present nanofeatured dental implants represent an evolutionary step from conventional microrough implants, yet they presently lack transformative capacity to surmount existing limitations. Further research and development endeavors are imperative to devise optimized surfaces rooted in fundamental science, thereby propelling technological progress in the field.


Assuntos
Implantes Dentários , Osseointegração , Propriedades de Superfície , Titânio , Humanos , Titânio/química , Nanoestruturas/química , Osteoblastos , Planejamento de Prótese Dentária
8.
ACS Appl Mater Interfaces ; 16(28): 36983-37006, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38953207

RESUMO

Repairing multiphasic defects is cumbersome. This study presents new soft and hard scaffold designs aimed at facilitating the regeneration of multiphasic defects by enhancing angiogenesis and improving cell attachment. Here, the nonimmunogenic, nontoxic, and cost-effective human serum albumin (HSA) fibril (HSA-F) was used to fabricate thermostable (up to 90 °C) and hard printable polymers. Additionally, using a 10.0 mg/mL HSA-F, an innovative hydrogel was synthesized in a mixture with 2.0% chitosan-conjugated arginine, which can gel in a cell-friendly and pH physiological environment (pH 7.4). The presence of HSA-F in both hard and soft scaffolds led to an increase in significant attachment of the scaffolds to the human periodontal ligament fibroblast (PDLF), human umbilical vein endothelial cell (HUVEC), and human osteoblast. Further studies showed that migration (up to 157%), proliferation (up to 400%), and metabolism (up to 210%) of these cells have also improved in the direction of tissue repair. By examining different in vitro and ex ovo experiments, we observed that the final multiphasic scaffold can increase blood vessel density in the process of per-vascularization as well as angiogenesis. By providing a coculture environment including PDLF and HUVEC, important cross-talk between these two cells prevails in the presence of roxadustat drug, a proangiogenic in this study. In vitro and ex ovo results demonstrated significant enhancements in the angiogenic response and cell attachment, indicating the effectiveness of the proposed design. This approach holds promise for the regeneration of complex tissue defects by providing a conducive environment for vascularization and cellular integration, thus promoting tissue healing.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Neovascularização Fisiológica , Alicerces Teciduais , Humanos , Alicerces Teciduais/química , Neovascularização Fisiológica/efeitos dos fármacos , Albumina Sérica Humana/química , Glicina/química , Glicina/farmacologia , Glicina/análogos & derivados , Fibroblastos/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/metabolismo , Proliferação de Células/efeitos dos fármacos , Amiloide/química , Amiloide/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/metabolismo , Ligamento Periodontal/citologia , Ligamento Periodontal/efeitos dos fármacos , Engenharia Tecidual , Hidrogéis/química , Hidrogéis/farmacologia , Temperatura , Isoquinolinas
9.
ACS Appl Mater Interfaces ; 16(28): 35964-35984, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38968558

RESUMO

Developing a neurovascular bone repair scaffold with an appropriate mechanical strength remains a challenge. Calcium phosphate (CaP) is similar to human bone, but its scaffolds are inherently brittle and inactive, which require recombination with active ions and polymers for bioactivity and suitable strength. This work discussed the synthesis of amorphous magnesium-calcium pyrophosphate (AMCP) and the subsequent development of a humidity-responsive AMCP/cassava starch (CS) scaffold. The scaffold demonstrated enhanced mechanical properties by strengthening the intermolecular hydrogen bonds and ionic bonds between AMCP and CS during the gelatinization and freeze-thawing processes. The release of active ions was rapid initially and stabilized into a long-term stable release after 3 days, which is well-matched with new bone growth. The release of pyrophosphate ions endowed the scaffold with antibacterial properties. At the cellular level, the released active ions simultaneously promoted the proliferation and mineralization of osteoblasts, the proliferation and migration of endothelial cells, and the proliferation of Schwann cells. At the animal level, the scaffold was demonstrated to promote vascular growth and peripheral nerve regeneration in a rat skull defect experiment, ultimately resulting in the significant and rapid repair of bone defects. The construction of the AMCP/CS scaffold offers practical suggestions and references for neurovascular bone repair.


Assuntos
Regeneração Óssea , Amido , Alicerces Teciduais , Animais , Regeneração Óssea/efeitos dos fármacos , Alicerces Teciduais/química , Ratos , Amido/química , Umidade , Humanos , Proliferação de Células/efeitos dos fármacos , Ratos Sprague-Dawley , Difosfatos/química , Difosfatos/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/citologia , Pirofosfato de Cálcio/química , Pirofosfato de Cálcio/farmacologia , Células de Schwann/efeitos dos fármacos , Células de Schwann/citologia , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Crânio/efeitos dos fármacos
10.
Theranostics ; 14(10): 3945-3962, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38994035

RESUMO

Rationale: NLRP3 inflammasome is critical in the development and progression of many metabolic diseases driven by chronic inflammation, but its effect on the pathology of postmenopausal osteoporosis (PMOP) remains poorly understood. Methods: We here firstly examined the levels of NLRP3 inflammasome in PMOP patients by ELISA. Then we investigated the possible mechanisms underlying the effect of NLRP3 inflammasome on PMOP by RNA sequencing of osteoblasts treated with NLRP3 siRNA and qPCR. Lastly, we accessed the effect of decreased NLRP3 levels on ovariectomized (OVX) rats. To specifically deliver NLRP3 siRNA to osteoblasts, we constructed NLRP3 siRNA wrapping osteoblast-specific aptamer (CH6)-functionalized lipid nanoparticles (termed as CH6-LNPs-siNLRP3). Results: We found that the levels of NLRP3 inflammasome were significantly increased in patients with PMOP, and were negatively correlated with estradiol levels. NLRP3 knock-down influenced signal pathways including immune system process, interferon signal pathway. Notably, of the top ten up-regulated genes in NLRP3-reduced osteoblasts, nine genes (except Mx2) were enriched in immune system process, and five genes were related to interferon signal pathway. The in vitro results showed that CH6-LNPs-siNLRP3 was relatively uniform with a dimeter of 96.64 ± 16.83 nm and zeta potential of 38.37 ± 1.86 mV. CH6-LNPs-siNLRP3 did not show obvious cytotoxicity and selectively delivered siRNA to bone tissue. Moreover, CH6-LNPs-siNLRP3 stimulated osteoblast differentiation by activating ALP and enhancing osteoblast matrix mineralization. When administrated to OVX rats, CH6-LNPs-siNLRP3 promoted bone formation and bone mass, improved bone microarchitecture and mechanical properties by decreasing the levels of NLRP3, IL-1ß and IL-18 and increasing the levels of OCN and Runx2. Conclusion: NLRP3 inflammasome may be a new biomarker for PMOP diagnosis and plays a key role in the pathology of PMOP. CH6-LNPs-siNLRP3 has potential application for the treatment of PMOP.


Assuntos
Inflamassomos , Lipossomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nanopartículas , Osteoblastos , Osteoporose Pós-Menopausa , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Feminino , Humanos , Ratos , Inflamassomos/metabolismo , Nanopartículas/química , Osteoporose Pós-Menopausa/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ratos Sprague-Dawley , RNA Interferente Pequeno/administração & dosagem , Aptâmeros de Nucleotídeos/farmacologia , Aptâmeros de Nucleotídeos/administração & dosagem , Modelos Animais de Doenças , Pessoa de Meia-Idade , Ovariectomia
11.
Front Endocrinol (Lausanne) ; 15: 1346094, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39022341

RESUMO

Background: The revolution of orthopedic implant manufacturing is being driven by 3D printing of titanium implants for large bony defects such as those caused by diabetic Charcot arthropathy. Unlike traditional subtractive manufacturing of orthopedic implants, 3D printing fuses titanium powder layer-by-layer, creating a unique surface roughness that could potentially enhance osseointegration. However, the metabolic impairments caused by diabetes, including negative alterations of bone metabolism, can lead to nonunion and decreased osseointegration with traditionally manufactured orthopedic implants. This study aimed to characterize the response of both healthy and diabetic primary human osteoblasts cultured on a medical-grade 3D-printed titanium surface under high and low glucose conditions. Methods: Bone samples were obtained from six patients, three with Type 2 Diabetes Mellitus and three without. Primary osteoblasts were isolated and cultured on 3D-printed titanium discs in high (4.5 g/L D-glucose) and low glucose (1 g/L D-Glucose) media. Cellular morphology, matrix deposition, and mineralization were assessed using scanning electron microscopy and alizarin red staining. Alkaline phosphatase activity and L-lactate concentration was measured in vitro to assess functional osteoblastic activity and cellular metabolism. Osteogenic gene expression of BGLAP, COL1A1, and BMP7 was analyzed using reverse-transcription quantitative polymerase chain reaction. Results: Diabetic osteoblasts were nonresponsive to variations in glucose levels compared to their healthy counterparts. Alkaline phosphatase activity, L-lactate production, mineral deposition, and osteogenic gene expression remained unchanged in diabetic osteoblasts under both glucose conditions. In contrast, healthy osteoblasts exhibited enhanced functional responsiveness in a high glucose environment and showed a significant increase in osteogenic gene expression of BGLAP, COL1A1, and BMP7 (p<.05). Conclusion: Our findings suggest that diabetic osteoblasts exhibit impaired responsiveness to variations in glucose concentrations, emphasizing potential osteoblast dysfunction in diabetes. This could have implications for post-surgery glucose management strategies in patients with diabetes. Despite the potential benefits of 3D printing for orthopedic implants, particularly for diabetic Charcot collapse, our results call for further research to optimize these interventions for improved patient outcomes.


Assuntos
Diabetes Mellitus Tipo 2 , Glucose , Osteoblastos , Impressão Tridimensional , Titânio , Humanos , Titânio/farmacologia , Osteoblastos/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células Cultivadas , Masculino , Fenótipo , Propriedades de Superfície , Feminino , Pessoa de Meia-Idade , Proteína Morfogenética Óssea 7/metabolismo , Osteogênese/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I/genética , Idoso
12.
Chem Biol Drug Des ; 104(1): e14574, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38958121

RESUMO

To develop novel bovine lactoferrin (bLF) peptides targeting bLF-tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) binding sites, we identified two peptides that could target bLF-TRAF6 binding sites using structural analysis. Moreover, another peptide that could bind to the TRAF6 dimerization area was selected from the bLF sequence. The effects of each peptide on cytokine expression in lipopolysaccharide (LPS)-stimulated osteoblasts (ST2) and on osteoclastogenesis were examined using an LPS-treated co-culture of primary bone marrow cells (BMCs) with ST2 cells and a single culture of osteoclast precursor cells (RAW-D) treated with soluble receptor activator of NF-κB ligand. Finally, the effectiveness of these peptides against LPS-induced alveolar bone destruction was assessed. Two of the three peptides significantly suppressed LPS-induced TNF-α and interleukin-1ß expression in ST2 cells. Additionally, these peptides inhibited and reversed LPS-induced receptor activator of NF-κB ligand (RANKL) upregulation and osteoprotegerin (OPG) downregulation, respectively. Furthermore, both peptides significantly reduced LPS-induced osteoclastogenesis in the BMC-ST2 co-culture and RANKL-induced osteoclastogenesis in RAW-D cells. In vivo, topical application of these peptides significantly reduced the osteoclast number by downregulating RANKL and upregulating OPG in the periodontal ligament. It is indicated that the novel bLF peptides can be used to treat periodontitis-associated bone destruction.


Assuntos
Lactoferrina , Lipopolissacarídeos , Osteoclastos , Peptídeos , Animais , Lactoferrina/farmacologia , Lactoferrina/química , Lactoferrina/metabolismo , Lipopolissacarídeos/farmacologia , Ratos , Peptídeos/farmacologia , Peptídeos/química , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Masculino , Perda do Osso Alveolar/tratamento farmacológico , Perda do Osso Alveolar/metabolismo , Perda do Osso Alveolar/patologia , Bovinos , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/citologia , Ratos Sprague-Dawley , Osteogênese/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Sítios de Ligação , Técnicas de Cocultura , Osteoprotegerina/metabolismo , Modelos Animais de Doenças
13.
FASEB J ; 38(13): e23776, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38958998

RESUMO

This study aimed to explore how mechanical stress affects osteogenic differentiation via the miR-187-3p/CNR2 pathway. To conduct this study, 24 female C57BL/6 mice, aged 8 weeks, were used and divided into four groups. The Sham and OVX groups did not undergo treadmill exercise, while the Sham + EX and OVX + EX groups received a 8-week treadmill exercise. Post-training, bone marrow and fresh femur samples were collected for further analysis. Molecular biology analysis, histomorphology analysis, and micro-CT analysis were conducted on these samples. Moreover, primary osteoblasts were cultured under osteogenic conditions and divided into GM group and CTS group. The cells in the CTS group underwent a sinusoidal stretching regimen for either 3 or 7 days. The expression of early osteoblast markers (Runx2, OPN, and ALP) was measured to assess differentiation. The study findings revealed that mechanical stress has a regulatory impact on osteoblast differentiation. The expression of miR-187-3p was observed to decrease, facilitating osteogenic differentiation, while the expression of CNR2 increased significantly. These observations suggest that mechanical stress, miR-187-3p, and CNR2 play crucial roles in regulating osteogenic differentiation. Both in vivo and in vitro experiments have confirmed that mechanical stress downregulates miR-187-3p and upregulates CNR2, which leads to the restoration of distal femoral bone mass and enhancement of osteoblast differentiation. Therefore, mechanical stress promotes osteoblasts, resulting in improved osteoporosis through the miR-187-3p/CNR2 signaling pathway. These findings have broad prospect and provide molecular biology guidance for the basic research and clinical application of exercise in the prevention and treatment of PMOP.


Assuntos
Diferenciação Celular , Camundongos Endogâmicos C57BL , MicroRNAs , Osteoblastos , Osteogênese , Osteoporose Pós-Menopausa , Estresse Mecânico , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Osteoblastos/metabolismo , Feminino , Osteoporose Pós-Menopausa/metabolismo , Osteoporose Pós-Menopausa/terapia , Osteoporose Pós-Menopausa/genética , Osteoporose Pós-Menopausa/patologia , Camundongos , Osteogênese/fisiologia , Humanos , Transdução de Sinais , Células Cultivadas
14.
Sci Rep ; 14(1): 15837, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982204

RESUMO

45S5 Bioglass (BG) is composed of a glass network with silicate based on the component and can be doped with various therapeutic ions for the enhancement of hard tissue therapy. Nanoceria (CeO2) has been shown to indicate redox reaction and enhance the biological response. However, few studies focus on the proportion of CeO2-doped and its effect on the cellular bioactivity of CeO2-doped BG (CBG). In this study, we synthesized the CBG series with increasing amounts of doping CeO2 ranging (1 to 12) wt.%. The synthesized CBG series examined the characterization, mineralization capacity, and cellular activity against BG. Our results showed that the CBG series exhibited a glass structure and indicated the redox states between Ce3+ and Ce4+, thus they showed the antioxidant activity by characterization of Ce. The CBG series had a stable glass network structure similar to BG, which showed the preservation of bioactivity by exhibiting mineralization on the surface. In terms of biological response, although the CBG series showed the proliferative activity of pre-osteoblastic cells similar to BG, the CBG series augmented not only the alkaline phosphatase activity but also the osteogenic marker in the mRNA level. As stimulated the osteogenic activity, the CBG series improved the biomineralization. In conclusion, the CBG series might have a potential application for hard tissue therapeutic purposes.


Assuntos
Cerâmica , Cério , Vidro , Oxirredução , Cério/química , Cério/farmacologia , Oxirredução/efeitos dos fármacos , Vidro/química , Camundongos , Cerâmica/química , Cerâmica/farmacologia , Animais , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Proliferação de Células/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Linhagem Celular , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Fosfatase Alcalina/metabolismo
15.
J Cell Mol Med ; 28(13): e18527, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38984969

RESUMO

Infected bone defects (IBDs) are the common condition in the clinical practice of orthopaedics. Although surgery and anti-infective medicine are the firstly chosen treatments, in many cases, patients experience a prolonged bone union process after anti-infective treatment. Epimedium-Curculigo herb pair (ECP) has been proved to be effective for bone repair. However, the mechanisms of ECP in IBDs are insufficiency. In this study, Effect of ECP in IBDs was verified by micro-CT and histological examination. Qualitative and quantitative analysis of the main components in ECP containing medicated serum (ECP-CS) were performed. The network pharmacological approaches were then applied to predict potential pathways for ECP associated with bone repair. In addition, the mechanism of ECP regulating LncRNA MALAT1/miRNA-34a-5p/SMAD2 signalling axis was evaluated by molecular biology experiments. In vivo experiments indicated that ECP could significantly promote bone repair. The results of the chemical components analysis and the pathway identification revealed that TGF-ß signalling pathway was related to ECP. The results of in vitro experiments indicated that ECP-CS could reverse the damage caused by LPS through inhibiting the expressions of LncRNA MALAT1 and SMAD2, and improving the expressions of miR-34a-5p, ALP, RUNX2 and Collagen type І in osteoblasts significantly. This research showed that ECP could regulate the TGF-ß/SMADs signalling pathway to promote bone repair. Meanwhile, ECP could alleviate LPS-induced bone loss by modulating the signalling axis of LncRNA MALAT1/miRNA-34a-5p/ SMAD2 in IBDs.


Assuntos
Epimedium , MicroRNAs , Osteoblastos , RNA Longo não Codificante , Transdução de Sinais , Proteína Smad2 , MicroRNAs/genética , MicroRNAs/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Proteína Smad2/metabolismo , Proteína Smad2/genética , Camundongos , Epimedium/química , Transdução de Sinais/efeitos dos fármacos , Masculino , Regeneração Óssea/efeitos dos fármacos , Humanos , Regulação da Expressão Gênica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética
16.
Biomed Mater ; 19(5)2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38955344

RESUMO

Artificial bone substitutes for bone repair and reconstruction still face enormous challenges. Previous studies have shown that calcium magnesium phosphate cements (CMPCs) possess an excellent bioactive surface, but its clinical application is restricted due to short setting time. This study aimed to develop new CMPC/carboxymethyl chitosan (CMCS) comg of mixed powders of active MgO, calcined MgO and calcium dihydrogen phosphate monohydrate. With this novel strategy, it can adjust the setting time and improve the compressive strength. The results confirmed that CMPC/CMCS composite bone cements were successfully developed with a controllable setting time (18-70 min) and high compressive strength (87 MPa). In addition, the composite bone cements could gradually degrade in PBS with weight loss up to 32% at 28 d. They also promoted the proliferation of pre-osteoblasts, and induced osteogenic differentiation. The findings indicate that CMPC/CMCS composite bone cements hold great promise as a new type of bone repair material in further and in-depth studies.


Assuntos
Materiais Biocompatíveis , Cimentos Ósseos , Fosfatos de Cálcio , Diferenciação Celular , Proliferação de Células , Quitosana , Força Compressiva , Compostos de Magnésio , Teste de Materiais , Osteoblastos , Osteogênese , Quitosana/química , Quitosana/análogos & derivados , Cimentos Ósseos/química , Cimentos Ósseos/farmacologia , Osteogênese/efeitos dos fármacos , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/citologia , Compostos de Magnésio/química , Compostos de Magnésio/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Substitutos Ósseos/química , Substitutos Ósseos/farmacologia , Fosfatos
17.
Sci Rep ; 14(1): 16133, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38997414

RESUMO

Nickel(Ni)-containing materials have been widely used in a wide range of medical applications, including orthopaedics. Despite their excellent properties, there is still a problem with the release of nickel ions into the patient's body, which can cause changes in the behaviour of surrounding cells and tissues. This study aims to evaluate the effects of Ni on bone cells with an emphasis on the determination of Ni localization in cellular compartments in time. For these purposes, one of the most suitable models for studying the effects induced by metal implants was used-the patient's osteoarthritic cells. Thanks to this it was possible to simulate the pathophysiological conditions in the patient's body, as well as to evaluate the response of the cells which come into direct contact with the material after the implantation of the joint replacement. The largest differences in cell viability, proliferation and cell cycle changes occurred between Ni 0.5 mM and 1 mM concentrations. Time-dependent localization of Ni in cells showed that there is a continuous transport of Ni ions between the nucleus and the cytoplasm, as well as between the cell and the environment. Moreover, osteoarthritic osteoblasts showed faster changes in concentration and ability to accumulate more Ni, especially in the nucleus, than physiological osteoblasts. The differences in Ni accumulation process explains the higher sensitivity of patient osteoblasts to Ni and may be crucial in further studies of implant-derived cytotoxic effects.


Assuntos
Proliferação de Células , Sobrevivência Celular , Níquel , Osteoartrite , Osteoblastos , Níquel/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , Humanos , Osteoartrite/metabolismo , Osteoartrite/patologia , Sobrevivência Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Íons/metabolismo , Ciclo Celular/efeitos dos fármacos , Células Cultivadas
18.
BMC Pharmacol Toxicol ; 25(1): 40, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38997762

RESUMO

BACKGROUND: Periapical lesions are characterized by periapical inflammation and damage to periapical tissues and eventually lead to bone resorption and even tooth loss. H2O2 is widely used in root canal therapy for patients with periapical inflammation. Luteolin possesses high anti-inflammatory, antioxidant, and anticancer potential. However, the underlying mechanism of the efficacy of H2O2 and luteolin on oxidative stress and inflammatory tissue has not been previously addressed. We aimed to investigate the anti-inflammatory and antioxidative effects of luteolin on H2O2-induced cellular oxidative inflammation. METHODS: After human osteoblasts (hFOB1.19) were treated with lipopolysaccharide (LPS), luteolin, or H2O2, cell proliferation was analysed by using a cell counting kit-8 (CCK-8), cell apoptosis was measured by using flow cytometry, the production of reactive oxygen species (ROS) was evaluated by using an oxidation-sensitive probe DCFH-DA ROS assay kit, and the expression of genes and proteins was detected by using reverse transcription quantitative polymerase chain reaction (RT‒qPCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA). RESULTS: We demonstrated that inflammation is closely related to oxidative stress and that the oxidative stress level in the inflammatory environment is increased. Luteolin inhibited the H2O2-induced increase in the expression of interleukin-6 (IL-6), interleukin-8 (IL-8) and tumour necrosis factor α (TNF-α) and significantly repressed the H2O2-induced increase in ROS, as well as markedly strengthened superoxide dismutase (SOD) activity in hFOB1.19 cells. Moreover, we detected that luteolin may inhibit H2O2-induced hFOB1.19 cell injury by suppressing the NF-κB pathway. CONCLUSION: We elucidated that luteolin protected human osteoblasts (hFOB1.19) from H2O2-induced cell injury and inhibited the production of proinflammatory cytokines by suppressing the NF-κB signalling pathway. Our findings provide a potential drug for treating H2O2-induced periodontitis and cell injury.


Assuntos
Anti-Inflamatórios , Peróxido de Hidrogênio , Inflamação , Luteolina , Osteoblastos , Estresse Oxidativo , Luteolina/farmacologia , Humanos , Estresse Oxidativo/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Peróxido de Hidrogênio/toxicidade , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Linhagem Celular , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Lipopolissacarídeos/farmacologia , Proliferação de Células/efeitos dos fármacos , Antioxidantes/farmacologia , NF-kappa B/metabolismo , Microambiente Celular/efeitos dos fármacos , Citocinas/metabolismo
19.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 41(3): 604-611, 2024 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-38932548

RESUMO

The surface morphology of titanium metal is an important factor affecting its hydrophilicity and biocompatibility, and exploring the surface treatment strategy of titanium metal is an important way to improve its biocompatibility . In this study , titanium (TA4) was firstly treated by large particle sand blasting and acid etching (SLA) technology, and then the obtained SLA-TA4 was treated by single surface treatments such as alkali-heat, ultraviolet light and plasma bombardment. According to the experimental results, alkali-heat treatment is the best treatment method to improve and maintain surface hydrophilicity of titanium. Then, the nanowire network morphology of titanium surface and its biological property, formed by further surface treatments on the basis of alkali-heat treatment, were investigated. Through the cell adhesion experiment of mouse embryonic osteoblast cells (MC3T3-E1), the ability of titanium material to support cell adhesion and cell spreading was investigated after different surface treatments. The mechanism of biological activity difference of titanium surface formed by different surface treatments was investigated according to the contact angle, pit depth and roughness of the titanium sheet surface. The results showed that the SLA-TA4 titanium sheet after a treatment of alkali heat for 10 h and ultraviolet irradiation for 1 h has the best biological activity and stability. From the perspective of improving surface bioactivity of medical devices, this study has important reference value for relevant researches on surface treatment of titanium implantable medical devices.


Assuntos
Materiais Biocompatíveis , Adesão Celular , Osteoblastos , Próteses e Implantes , Propriedades de Superfície , Titânio , Titânio/química , Animais , Camundongos , Osteoblastos/citologia , Teste de Materiais , Linhagem Celular , Raios Ultravioleta , Interações Hidrofóbicas e Hidrofílicas
20.
Biochem Biophys Res Commun ; 725: 150265, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-38901225

RESUMO

With the substantial increase in the overuse of glucocorticoids (GCs) in clinical medicine, the prevalence of glucocorticoid-induced osteonecrosis of the femoral head (GC-ONFH) continues to rise in recent years. However, the optimal treatment for GC-ONFH remains elusive. Rotating magnetic field (RMF), considered as a non-invasive, safe and effective approach, has been proved to have multiple beneficial biological effects including improving bone diseases. To verify the effects of RMF on GC-ONFH, a lipopolysaccharide (LPS) and methylprednisolone (MPS)-induced invivo rat model, and an MPS-induced invitro cell model have been employed. The results demonstrate that RMF alleviated bone mineral loss and femoral head collapse in GC-ONFH rats. Meanwhile, RMF reduced serum lipid levels, attenuated cystic lesions, raised the expression of anti-apoptotic proteins and osteoprotegerin (OPG), while suppressed the expression of pro-apoptotic proteins and nuclear factor receptor activator-κB (RANK) in GC-ONFH rats. Besides, RMF also facilitated the generation of ALP, attenuated apoptosis and inhibits the expression of pro-apoptotic proteins, facilitated the expression of OPG, and inhibited the expression of RANK in MPS-stimulated MC3T3-E1 cells. Thus, this study indicates that RMF can improve GC-ONFH in rat and cell models, suggesting that RMF have the potential in the treatment of clinical GC-ONFH.


Assuntos
Diferenciação Celular , Necrose da Cabeça do Fêmur , Glucocorticoides , Osteoblastos , Ratos Sprague-Dawley , Animais , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/patologia , Necrose da Cabeça do Fêmur/metabolismo , Necrose da Cabeça do Fêmur/terapia , Ratos , Diferenciação Celular/efeitos dos fármacos , Masculino , Campos Magnéticos , Magnetoterapia/métodos , Cabeça do Fêmur/patologia , Cabeça do Fêmur/metabolismo , Modelos Animais de Doenças , Rotação , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...