Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Genet ; 13: 938334, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36267398

RESUMO

Purpose: Observational research has indicated the presence of a causal relationship between sleep disturbances and low back pain (LBP). However, the link may have been biased by confounding factors. The purpose of this study was to examine the potential causal association of insomnia and daytime sleepiness with LBP by using mendelian randomization (MR). Methods: Genome-wide association study (GWAS) summary statistics of insomnia were obtained from a large-scale GWAS meta-analysis (n = 1,331,010; individuals from UK Biobank and 23andMe) or UK Biobank alone (n = 453,379). The summary statistics of daytime sleepiness were from UK Biobank (n = 452,071) and LBP were provided by the FinnGen Release 6 (210,645 individuals with 16,356 LBP cases and 194,289 controls) or UK Biobank (5,423 cases versus 355,771 controls). Linkage disequilibrium score (LDSC) regression and bidirectional MR analysis was employed to estimate genetic correlation and causal relationship. In the MR analysis, the inverse variance weighted method (IVW) was utilized as the main analysis procedure, while MR-Egger, Weighted median and Robust adjusted profile score (RAPS) were utilized for supplementary analyses. Results: LDSC analysis showed that LBP were significantly genetically correlated with insomnia (rg = 0.57, p = 2.26e-25) and daytime sleepiness (rg = 0.18, p = 0.001). The MR analysis revealed that genetically predicted insomnia was significantly associated with an increased risk of LBP (OR = 1.250, 95% CI: 1.186-1.318; p = 1.69e-16). However, the reverse causality was not confirmed. No evidence was identified supporting causality of daytime sleepiness and LBP. Conclusion: This study demonstrates a putative causal link of insomnia on LBP and a null causal effect of LBP on insomnia. Furthermore, a causal link between daytime sleepiness and LBP were not reported. This finding may stimulate new strategies for patient management in clinical practice, benefiting public health.

2.
Front Surg ; 9: 1030999, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36684180

RESUMO

Background: Thoracic ossification of the posterior longitudinal ligament (TOPLL) requires surgery for spinal cord decompression. Traditional open surgery is extremely invasive and has various complications. Unilateral biportal endoscopy (UBE) is a newly developed technique for spine surgery, especially in the lumbar region, but rare in the thoracic spine. In this study, we first used a different percutaneous UBE "cave-in" decompression technique for the treatment of beak-type TOPLL. Methods: A 31-year-old female with distinct zonesthesia and numbness below the T3 dermatome caused by beak-type TOPLL (T2-T3) underwent a two-step UBE decompression procedure. In the first step, the ipsilateral lamina, left facet joint, partial transverse process, and pedicles of T2 and T3 were removed. In the second step, a cave was created by removing the posterior third of the vertebral body (T2-T3). The eggshell-like TOPLL was excised by forceps, and the dural sac was decompressed. All procedures are performed under endoscopic guidance. A drainage tube was inserted, and the incisions were closed after compliance with the decompression scope via a C-arm. The patient's preoperative and postoperative radiological and clinical results were evaluated. Results: Postoperative CT and MR films conformed complete decompression of the spinal cord. The patient's lower extremity muscle strength was greatly improved, and no complications occurred. The mJOA score improved from 5 to 7, with a recovery rate of 33.3%. Conclusion: UBE spinal decompression for TOPLL showed favorable clinical and radiological results and offers the advantages of minimal soft tissue dissection, shorter hospital stays, and a faster return to daily life activities.

4.
Oncotarget ; 8(48): 83949-83961, 2017 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-29137395

RESUMO

Osteoclast over-activation and inflammation responses promote peri-implant osteolysis (PIO), which is the leading cause of aseptic artificial joint loosening. We examined the effect of lithium chloride (LiCl) on wear debris-induced osteoclastogenesis and inflammation. Fifty-Six C57BL/6J male mice were randomly distributed into four groups: sham control (sham, treated with phosphate buffered saline [PBS]), vehicle (treated with titanium/PBS), low-LiCl (L-LiCl, titanium: 50 mg/kg LiCl) and high-LiCl (H-LiCl, titanium: 200 mg/kg LiCl). After 14 days, mouse calvaria were harvested for micro-computed tomography and histomorphological and molecular analyses. Bone marrow-derived macrophages (BMMs) were extracted to examine osteoclast differentiation, and the RAW264.7 cell line was used to investigate osteoclastogenesis mechanisms. LiCl reduced the number of osteoclasts, debris-induced osteolysis, and the expression of inflammatory factors, thereby preventing bone loss in vivo. In vitro, LiCl inhibited osteoclastogenesis and osteoclastic bone resorption by inhibiting the RANKL-induced NF-κB signaling pathway. LiCl's activation of the canonical Wnt/ß-catenin signaling pathway was not associated with LiCl's inhibition of osteoclastogenesis. These results suggest that LiCl may be an effective agent for treatment of osteolytic diseases caused by chronic inflammation and over-activation of osteoclasts.

5.
Acta Biomater ; 62: 362-371, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28867647

RESUMO

Wear debris-induced peri-implant osteolysis challenges the longevity of implants. The host response to wear debris causes chronic inflammation, promotes bone resorption, and impairs bone formation. We previously demonstrated that melatonin enhances bone formation and attenuates wear debris-induced bone loss in vivo. However, whether melatonin inhibits chronic inflammation and bone resorption at sites of wear debris-induced osteolysis remains unclear. In this study, we examined the potential inhibitory effects of melatonin on titanium particle-induced inflammatory osteolysis in a murine calvarial model and on RANKL-induced osteoclastic formation in bone marrow-derived macrophages. We found that the exogenous administration of melatonin significantly inhibited wear debris-induced bone resorption and the expression of inflammatory cytokines in vivo. Additionally, melatonin inhibited RANKL-induced osteoclast differentiation, F-actin ring formation, and osteoclastic resorption in a concentration-dependent manner in vitro. We also showed that melatonin blocked the phosphorylation of IκB-α and p65, but not IKKα, and significantly inhibited the expression of NFATc1 and c-Fos. However, melatonin had no effect on MAPK or PI3K/AKT signaling pathways. These results provide novel mechanistic insight into the anti-inflammatory and anti-bone resorptive effects of melatonin on wear debris-induced bone loss and provide an evidence-based rationale for the protective effects of melatonin as a treatment for peri-implant osteolysis. STATEMENT OF SIGNIFICANCE: Wear debris-induced chronic inflammation, osteoclastic activation and osteoblastic inhibition have been identified as critical factors of peri-implant bone loss. We previously demonstrated that melatonin, a bioactive indolamine secreted mainly by the pineal gland, activates Wnt/ß-catenin signaling pathway and enhances bone regeneration at osteolytic site in vivo. In the current study, we further demonstrated that melatonin significantly suppresses wear debris-induced bone resorption and inflammatory cytokine expression in vivo. In addition, melatonin inhibits receptor activator of nuclear factor kappa-B ligand induced osteoclast formation and osteoclastic bone resorption in vitro. Meanwhile, we found that melatonin mediates its anti-inflammation and anti-bone resorption effects by abrogating nuclear factor kappa-B activation. These results further support the protective effects of melatonin on wear debris-induced peri-implant bone loss, and strongly suggest that melatonin could be considered as a potential candidate for the prevention and treatment of wear debris-induced osteolysis and subsequent aseptic loosening.


Assuntos
Reabsorção Óssea , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melatonina/farmacologia , NF-kappa B/metabolismo , Osteoclastos/metabolismo , Próteses e Implantes/efeitos adversos , Titânio/efeitos adversos , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Reabsorção Óssea/induzido quimicamente , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Osteoclastos/patologia , Ligante RANK/metabolismo , Titânio/farmacologia
6.
Acta Biomater ; 51: 513-525, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28088671

RESUMO

Wear debris-induced inhibition of bone regeneration and extensive bone resorption were common features in peri-prosthetic osteolysis (PPO). Here, we investigated the effect of melatonin on titanium particle-stimulated osteolysis in a murine calvariae model and mouse-mesenchymal-stem cells (mMSCs) culture system. Melatonin inhibited titanium particle-induced osteolysis and increased bone formation at osteolytic sites, confirmed by radiological and histomorphometric data. Furthermore, osteoclast numbers decreased dramatically in the low- and high-melatonin administration mice, as respectively, compared with the untreated animals. Melatonin alleviated titanium particle-induced depression of osteoblastic differentiation and mineralization in mMSCs. Mechanistically, melatonin was found to reduce the degradation of ß-catenin, levels of which were decreased in presence of titanium particles both in vivo and in vitro. To further ensure whether the protective effect of melatonin was mediated by the Wnt/ß-catenin signaling pathway, ICG-001, a selective ß-catenin inhibitor, was added to the melatonin-treated groups and was found to attenuate the effect of melatonin on mMSC mineralization. We also demonstrated that melatonin modulated the balance between receptor activator of nuclear factor kappa-B ligand and osteoprotegerin via activation of Wnt/ß-catenin signaling pathway. These findings strongly suggest that melatonin represents a promising candidate in the treatment of PPO. STATEMENT OF SIGNIFICANCE: Peri-prosthetic osteolysis, initiated by wear debris-induced inhibition of bone regeneration and extensive bone resorption, is the leading cause for implant failure and reason for revision surgery. In the current study, we demonstrated for the first time that melatonin can induce bone regeneration and reduce bone resorption at osteolytic sites caused by titanium-particle stimulation. These effects might be mediated by activating Wnt/ß-catenin signaling pathway and enhancing osteogenic differentiation. Meanwhile, the ability of melatonin to modulate the balance between receptor activator of nuclear factor kappa-B ligand and osteoprotegerin mediated by Wnt/ß-catenin signaling pathway, thereby suppressing osteoclastogenesis, may be implicated in the protective effects of melatonin on titanium-particle-induced bone resorption. These results suggested that melatonin can be considered as a promising therapeutic agent for the prevention and treatment of peri-prosthetic osteolysis.


Assuntos
Melatonina/uso terapêutico , Osteólise/induzido quimicamente , Osteólise/tratamento farmacológico , Titânio/efeitos adversos , Via de Sinalização Wnt , Animais , Regeneração Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Melatonina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Osteólise/metabolismo , Osteólise/patologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Crânio/patologia , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
7.
Acta Biomater ; 48: 479-488, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27838465

RESUMO

Peri-implant osteolysis (PIO) and the following aseptic loosening is the leading cause of implant failure. Emerging evidence suggests that receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast formation and osteoclastic bone resorption are responsible for particle-stimulated PIO. Here, we explored the effect of theaflavin-3,3'-digallate (TF3) on titanium particle-induced osteolysis in vivo and in vitro. Twenty-eight male C57BL/6 mice were randomly separated into four groups: sham control (sham), titanium particles only (titanium), titanium particles with low TF3 concentration (low-TF3, 1mg/kg TF3), and titanium particles with high TF3 concentration (high-TF3, 10mg/kg TF3). Two weeks later, micro-computed tomography and histological analysis were performed. Bone-marrow-derived macrophages and RAW264.7 murine macrophages were applied to examine osteoclast formation and differentiation. TF3 significantly inhibited titanium particle-induced osteolysis and prevented bone destruction compared with titanium group. Interestingly, the number of mature osteoclasts reduced after treatment with TF3 in vivo, suggesting osteoclast formation might be inhibited by TF3. In vitro, TF3 suppressed osteoclast formation, polarization and osteoclastic bone resorption by specifically targeting the RANKL-induced ERK signal pathway. Collectively, these results suggest that TF3, a natural active compound derived from black tea, is a promising candidate for the treatment of osteoclast-related osteolytic diseases, such as wear debris-induced PIO. STATEMENT OF SIGNIFICANCE: Total joint arthroplasty is widely accepted for the treatment of end-stage joint diseases. However, it is reported that aseptic loosening, initiated by peri-implant osteolysis, is the major reason for prosthesis failure. Although the pathophysiology of PIO remains unclear, increasing evidence indicates that osteoclasts are excessively activated at the implant site by wear debris from materials. Here, we demonstrated that theaflavin-3,3'-digallate, a natural active compound derived from black tea, inhibited osteoclast formation and osteoclastic bone resorption mainly via suppressing the ERK pathway. Moreover, the findings of this study have confirmed for the first time that theaflavin-3,3'-digallate has a protective effect on particle-induced osteolysis in a mouse calvarial model, thus preventing bone loss. These results indicate that theaflavin-3,3'-digallate may be a suitable therapeutic agent to treat wear debris-induced peri-implant osteolysis.


Assuntos
Biflavonoides/farmacologia , Catequina/análogos & derivados , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Osteólise/induzido quimicamente , Osteólise/patologia , Titânio/efeitos adversos , Animais , Biflavonoides/uso terapêutico , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Catequina/farmacologia , Catequina/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/efeitos dos fármacos , Osteólise/tratamento farmacológico , Ligante RANK/farmacologia , Células RAW 264.7 , Crânio/efeitos dos fármacos , Crânio/patologia
8.
Sci Rep ; 6: 36251, 2016 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-27796351

RESUMO

Wear-particle-induced chronic inflammation and osteoclastogenesis have been identified as critical factors of aseptic loosening. Although strontium is known to be involved in osteoclast differentiation, its effect on particle-induced inflammatory osteolysis remains unclear. In this study, we investigate the potential impact and underling mechanism of strontium on particle-induced osteoclast activation and chronic inflammation in vivo and in vitro. As expected, strontium significantly inhibited titanium particle-induced inflammatory infiltration and prevented bone loss in a murine calvarial osteolysis model. Interestingly, the number of mature osteoclasts decreased after treatment with strontium in vivo, suggesting osteoclast formation might be inhibited by strontium. Additionally, low receptor activator of nuclear factor-κB ligand (RANKL), tumor necrosis factor-α, interleukin-1ß, interleukin-6 and p65 immunochemistry staining were observed in strontium-treatment groups. In vitro, strontium obviously decreased osteoclast formation, osteoclastogenesis-related gene expression, osteoclastic bone resorption and pro-inflammatory cytokine expression in bone-marrow-derived macrophages in a dose-dependent manner. Furthermore, we demonstrated that strontium impaired osteoclastogenesis by blocking RANKL-induced activation of NF-κB pathway. In conclusion, our study demonstrated that strontium can significantly inhibit particle-induced osteoclast activation and inflammatory bone loss by disturbing the NF-κB pathway, and is an effective therapeutic agent for the treatment of wear particle-induced aseptic loosening.


Assuntos
Reabsorção Óssea/tratamento farmacológico , NF-kappa B/imunologia , Osteoclastos/imunologia , Transdução de Sinais/efeitos dos fármacos , Estrôncio/farmacologia , Titânio/toxicidade , Animais , Reabsorção Óssea/induzido quimicamente , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Citocinas/imunologia , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Osteoclastos/patologia , Transdução de Sinais/imunologia
9.
Sci Rep ; 6: 23827, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-27029606

RESUMO

Wear-debris-induced periprosthetic osteolysis (PIO) is a common clinical condition following total joint arthroplasty, which can cause implant instability and failure. The host response to wear debris promotes bone resorption and impairs bone formation. We previously demonstrated that icariin suppressed wear-debris-induced osteoclastogenesis and attenuated particle-induced osteolysis in vivo. Whether icariin promotes bone formation in a wear-debris-induced osteolytic site remains unclear. Here, we demonstrated that icariin significantly attenuated titanium-particle inhibition of osteogenic differentiation of mesenchymal stem cells (MSCs). Additionally, icariin increased bone mass and decreased bone loss in titanium-particle-induced osteolytic sites. Mechanistically, icariin inhibited decreased ß-catenin stability induced by titanium particles in vivo and in vitro. To confirm icariin mediated its bone-protective effects via the Wnt/ß-catenin signaling pathway, we demonstrated that ICG-001, a selective Wnt/ß-catenin inhibitor, attenuated the effects of icariin on MSC mineralization in vitro and bone formation in vivo. Therefore, icariin could induce osteogenic differentiation of MSCs and promote new bone formation at a titanium-particle-induced osteolytic site via activation of the Wnt/ß-catenin signaling pathway. These results further support the protective effects of icariin on particle-induced bone loss and provide novel mechanistic insights into the recognized bone-anabolic effects of icariin and an evidence-based rationale for its use in PIO treatment.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Flavonoides/farmacologia , Osteogênese/efeitos dos fármacos , Osteólise/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Titânio/efeitos adversos , beta Catenina/agonistas , Administração Tópica , Animais , Interface Osso-Implante/patologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Diferenciação Celular , Feminino , Flavonoides/antagonistas & inibidores , Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteólise/induzido quimicamente , Osteólise/metabolismo , Osteólise/patologia , Cultura Primária de Células , Pirimidinonas/farmacologia , Crânio/efeitos dos fármacos , Crânio/metabolismo , Crânio/cirurgia , beta Catenina/antagonistas & inibidores , beta Catenina/genética , beta Catenina/metabolismo
10.
Biomaterials ; 80: 1-10, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26695376

RESUMO

Chronic inflammation and extensive osteoclast formation play critical roles in wear-debris-induced peri-implant osteolysis. We investigated the potential impact of dopamine on titanium-particle-induced inflammatory osteolysis in vivo and in vitro. Twenty-eight C57BL/6J mice were randomly assigned to four groups: sham control (PBS treatment), titanium (titanium/PBS treatment), low- (titanium/2 µg kg(-1) day(-1) dopamine) and high-dopamine (titanium/10 µg kg(-1) day(-1) dopamine). After 2 weeks, mouse calvariae were collected for micro-computed tomography (micro-CT) and histomorphometry analysis. Bone-marrow-derived macrophages (BMMs) were isolated to assess osteoclast differentiation. Dopamine significantly reduced titanium-particle-induced osteolysis compared with the titanium group as confirmed by micro-CT and histomorphometric data. Osteoclast numbers were 34.9% and 59.7% (both p < 0.01) lower in the low- and high-dopamine-treatment groups, respectively, than in the titanium group. Additionally, low RANKL, tumor necrosis factor-α, interleukin-1ß and interleukin-6 immunochemistry staining were noted in dopamine-treatment groups. Dopamine markedly inhibited osteoclast formation, osteoclastogenesis-related gene expression and pro-inflammatory cytokine expression in BMMs in a dose-dependent manner. Moreover, the resorption area was decreased with 10(-9) M and 10(-8) M dopamine to 40.0% and 14.5% (both p < 0.01), respectively. Furthermore, the inhibitory effect of dopamine was reversed by the D2-like-receptor antagonist haloperidol but not by the D1-like-receptor antagonist SCH23390. These results suggest that dopamine therapy could be developed into an effective and safe method for osteolysis-related disease caused by chronic inflammation and excessive osteoclast formation.


Assuntos
Dopamina/uso terapêutico , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Osteólise/induzido quimicamente , Osteólise/tratamento farmacológico , Titânio/efeitos adversos , Animais , Células Cultivadas , Dopamina/administração & dosagem , Inflamação/patologia , Interleucina-1beta/análise , Interleucina-6/análise , Masculino , Camundongos Endogâmicos C57BL , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteólise/patologia , Ligante RANK/análise , Receptores de Dopamina D2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Crânio/efeitos dos fármacos , Crânio/patologia , Fator de Necrose Tumoral alfa/análise , Microtomografia por Raio-X
11.
Biomaterials ; 69: 12-21, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26275858

RESUMO

Aseptic loosening is associated with the development of wear debris-induced peri-implant osteolytic bone disease caused by an increased osteoclastic bone resorption and decreased osteoblastic bone formation. However, no effective measures for the prevention and treatment of peri-implant osteolysis currently exist. The aim of this study was to determine whether lithium chloride (LiCl), a selective inhibitor of glycogen synthetase kinase 3 beta (GSK-3ß), mitigates wear debris-induced osteolysis in a murine calvarial model of osteolysis. GSK-3ß is activated by titanium (Ti) particles, and implantation of Ti particles on the calvarial surface in C57BL/6 mice resulted in osteolysis caused by an increase in the number of osteoclasts and a decrease in the number of osteoblasts. Mice implanted with Ti particles were gavage-fed LiCl (50 or 200 mg kg(-1)d(-1)), 6 days per week for 2 weeks. The LiCl treatment significantly inhibited GSK-3ß activity and increased ß-catenin and axin-2 expression in a dose-dependent manner, dramatically mitigating the Ti particle-induced suppression of osteoblast numbers and the expression of bone formation markers. Finally, we demonstrated that inhibition of GSK-3ß suppresses osteoclast differentiation and reduces the severity of Ti particle-induced osteolysis. The results of this study indicate that Ti particle-induced osteolysis is partly dependent on GSK-3ß and, therefore, the canonical Wnt signaling pathway. This suggests that selective inhibitors of GSK-3ß such as LiCl may help prevent and treat wear debris-induced osteolysis.


Assuntos
Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Cloreto de Lítio/uso terapêutico , Osteólise/tratamento farmacológico , Osteólise/enzimologia , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Endogâmicos C57BL , Osteólise/etiologia , Osteólise/patologia , Próteses e Implantes/efeitos adversos , Crânio/efeitos dos fármacos , Crânio/enzimologia , Crânio/patologia , Titânio/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA