Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 206: 115264, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36174767

RESUMO

Prenatal dexamethasone exposure (PDE) can lead to offspring long bone dysplasia and continue to postnatal, and this is an important cause of fetal-derived osteoporosis. Studies have confirmed that intrauterine endogenous GC overexposure mediates multiple organ dysplasia and adult-related disease susceptibility in offspring through the glucocorticoid-insulin-like growth factor1 (GC-IGF1) axis. However, it remains unknown if exogenous dexamethasone can regulate bone development in offspring through the GC-IGF1 axis. We determined that the PDE fetal rats exhibited poor osteogenic differentiation, decreased bone mass that continued to adolescence, and increased susceptibility to osteoporosis in adulthood. Concurrently, PDE decreased the serum corticosterone concentration and IGF1 expression in offspring before and after birth, while the increased serum corticosterone concentration induced by chronic stress reversed the inhibition of IGF1 expression induced by PDE. Furthermore, PDE decreased the expression of GRα and miR-130a-5p, increased HDAC4, and decreased H3K27 acetylation in the IGF1 promoter region in bone tissue, and the above changes were negatively compensated after chronic stress. In vitro, a low concentration of corticosterone inhibited the expression of GRα and miR130a-5p, upregulated the expression of HDAC4, inhibited the promoter region H3K27 acetylation, and expression of IGF1 in bone marrow mesenchymal stem cell (BMSCs) osteoblast differentiated cells and inhibited osteogenic differentiation of BMSCs. GRα overexpression, miR-130a-5p mimic treatment, or HDAC4 siRNA exposure reversed the downstream molecular alterations caused by low corticosterone concentrations. In conclusion, PDE-induced intrauterine hypoglucocorticoid exposure could positively program IGF1 expression in bone tissue through the GRα/miR-130a-5p/HDAC4 pathways, thus mediating osteogenic dysdifferentiation and adult osteoporosis susceptibility in male offspring rats.


Assuntos
Dexametasona , MicroRNAs , Osteoporose , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Masculino , Gravidez , Ratos , Corticosterona/efeitos adversos , Dexametasona/toxicidade , Fator de Crescimento Insulin-Like I/genética , MicroRNAs/genética , Osteogênese , Osteoporose/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Wistar , Glucocorticoides
2.
Arthroscopy ; 38(7): 2246-2254.e1, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35093495

RESUMO

PURPOSE: This study aimed to verify whether transplantation of dedifferentiated osteogenic bone marrow mesenchymal stem cells (De-BMSCs) at the tendon-bone interface could result in more bone formation than BMSC transplantation in anterior cruciate ligament (ACL) reconstruction. METHODS: BMSCs from femur and tibia of New Zealand White rabbit were subjected to osteogenic induction and then cultured in osteogenic factor-free medium; the obtained cell population was termed De-BMSCs. Bilateral ACL reconstruction was performed in 48 adult rabbits. Three groups were established: control group with alginate gel injection, BMSCs group with the BMSCs injection, and De-BMSCs group with the De-BMSCs injection. At week 4 and 12 postoperatively, tendon-bone healing by histologic staining, micro-computed tomography examination, and biomechanical test were evaluated. RESULTS: The expression of α1 chain of type I collagen, osteocalcin, and osteopontin at the tendon-bone interface in the De-BMSCs group was greater than in the control or BMSCs group. The bone volume/total volume by micro-computed tomography scan was significantly greater in the De-BMSCs group than that in the control group (P = .013) or BMSCs group (P = .045) at 4 weeks, and greater than that in the control group (P = .014) or BMSCs group (P = .017) at 12 weeks. The tunnel area was significantly smaller in the De-BMSCs group than in the control group (P = .013) or BMSCs group (P = .044) at 12 weeks. The failure load and stiffness in De-BMSCs group were both significantly enhanced at 4 and 12 weeks than control group or De-BMSCs group. CONCLUSIONS: De-BMSCs transplantation can promote bone formation at the tendon-bone interface better than BMSCs transplantation in ACL reconstruction and increase the early biomechanical strength of the reconstructed ACL CLINICAL RELEVANCE: De-BMSCs transplantation is a potential choice for enhancing early bone formation in the tunnel in ACL reconstruction.


Assuntos
Reconstrução do Ligamento Cruzado Anterior , Células-Tronco Mesenquimais , Animais , Reconstrução do Ligamento Cruzado Anterior/métodos , Células da Medula Óssea , Osteogênese , Coelhos , Tendões/metabolismo , Microtomografia por Raio-X
3.
Pharmacol Res ; 175: 105990, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34808367

RESUMO

Epidemiological investigations have shown that individuals treated with dexamethasone during pregnancy have an increased risk of osteoporosis after birth. Our studies reported that peak bone mass was decreased in the prenatal dexamethasone exposure (PDE) offspring before chronic stress, while further decrease was observed after chronic stress. Simultaneously, increase of bone local active corticosterone was observed in the PDE offspring, while further increase was also observed after chronic stress. Moreover, the histone H3 lysine 9 acetylation (H3K9ac) level of 11-beta hydroxysteroid dehydrogenase 2 (11ß-HSD2) and its expression in bone tissue of PDE offspring rats remained lower than the control before and after birth. Injection of 11ß-HSD2 overexpression lentivirus into the bone marrow cavity could partially alleviate the accumulation of bone local active corticosterone and bone loss induced by PDE. In vitro, dexamethasone inhibited the expression of 11ß-HSD2 and aggravated the inhibitory effect of corticosterone on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Overexpression of 11ß-HSD2 partially alleviated the inhibitory effect of corticosterone. Moreover, dexamethasone promoted the nuclear translocation of glucocorticoid receptor (GR), which resulted in the stimulation of 11ß-HSD2 expression due to the binding of GR to the 11ß-HSD2 promoter region directly, as well as increasing H3K9ac level in the 11ß-HSD2 promoter region by recruiting histone deacetylase 11 (HDAC11). Our results indicated that low expression of 11ß-HSD2 in bone tissue is an important mediator for the high susceptibility to osteoporosis in PDE adult offspring.


Assuntos
Anti-Inflamatórios/efeitos adversos , Dexametasona/efeitos adversos , Glucocorticoides/efeitos adversos , Histona Desacetilases/genética , Osteoporose/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Animais , Desenvolvimento Ósseo/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Corticosterona/sangue , Corticosterona/metabolismo , Feminino , Histona Desacetilases/metabolismo , Masculino , Osteoporose/genética , Osteoporose/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Wistar , Estresse Fisiológico
4.
Stem Cell Res Ther ; 12(1): 576, 2021 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-34775995

RESUMO

BACKGROUND: Bone formation plays an important role in early tendon-bone healing after anterior cruciate ligament reconstruction (ACLR). Dedifferentiated osteogenic bone marrow mesenchymal stem cells (De-BMSCs) have enhanced osteogenic potential. This study aimed to investigate the effect of De-BMSCs transplantation on the promotion of bone formation at the tendon-bone interface after ACLR and to further explore the molecular mechanism of the enhanced osteogenic potential of De-BMSCs. METHODS: BMSCs from the femurs and tibias of New Zealand white rabbits were subjected to osteogenic induction and then cultured in medium without osteogenic factors; the obtained cell population was termed De-BMSCs. De-BMSCs were induced to undergo osteo-, chondro- and adipo-differentiation in vitro to examine the characteristics of primitive stem cells. An ACLR model with a semitendinosus tendon was established in rabbits, and the animals were divided into a control group, BMSCs group, and De-BMSCs group. At 12 weeks after surgery, the rabbits in each group were sacrificed to evaluate tendon-bone healing by histologic staining, micro-computed tomography (micro-CT) examination, and biomechanical testing. During osteogenic differentiation of De-BMSCs, an siRNA targeting nuclear factor of activated T-cells 1 (NFATc1) was used to verify the molecular mechanism of the enhanced osteogenic potential of De-BMSCs. RESULTS: De-BMSCs exhibited some properties similar to BMSCs, including multiple differentiation potential and cell surface markers. Bone formation at the tendon-bone interface in the De-BMSCs group was significantly increased, and biomechanical strength was significantly improved. During the osteogenic differentiation of De-BMSCs, the expression of Nanog and NFATc1 was synergistically increased, which promoted the interaction of NFATc1 and Osterix, resulting in increased expression of osteoblast marker genes such as COL1A, OCN, and OPN. CONCLUSIONS: De-BMSCs transplantation could promote bone formation at the tendon-bone interface after ACLR and improve the biomechanical strength of the reconstruction. The Nanog/NFATc1/Osterix signaling pathway mediated the enhanced osteogenic differentiation efficiency of De-BMSCs.


Assuntos
Osteogênese , Cicatrização , Animais , Diferenciação Celular , Coelhos , Transdução de Sinais , Tendões , Microtomografia por Raio-X
5.
BMC Musculoskelet Disord ; 22(1): 787, 2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34517870

RESUMO

BACKGROUND: Venous thromboembolism (VTE) is a potentially fatal complication after arthroplasty. Numerous prophylactic strategies and studies to reduce VTEs have focused on the duration of the hospital stay and on few extramural hospitals. This study aimed to investigate extramural hospital management of VTE after total hip/knee arthroplasty (THA/TKA) in China with a novel survey tool. METHODS: A total of 180 patients undergoing arthroplasty, including 68 THA patients and 112 TKA patients, were enrolled in this study. All patients received anticoagulant treatment management. A survey querying VTE management and adherence, such as therapy information, understanding of anticoagulation, satisfaction with the ability of medical staff, and satisfaction with health care costs, was administered by a questionnaire (TKA/THA Patients' Experience with Anticoagulation in the Post-discharge Period) for quality improvement. RESULTS: The average age of the patients was 65.27 ± 13.62 years. All patients knew their follow-up times. 85 % of them were suggested that re-examine at the next 14 days, and the others at the next 28 days. All patients continued to visit the orthopaedic clinic after discharge without choosing other types of outpatient services, such as an anticoagulant clinic or home visit with a nurse/pharmacist or remote evaluation by telephone. A total of 96.6 % of all patients used new oral anticoagulants, and the most common treatment duration was 2-4 weeks (93.3 %). 48 % informed their physicians that they were taking anticoagulation medications when they visited ophthalmology, dentistry, dermatology, and other departments. The overall rate of satisfaction with anticoagulation management was 81.67 %, and 6.67 % of patients were not unsatisfied with their medical expenses. Patient compliance decreased with increasing follow-up time. Continuous follow-ups after discharge significantly improved patient compliance. CONCLUSIONS: These results elucidate how we can improve the quality of anticoagulation. Continuous follow-up appointments for 30 days after discharge, especially for individuals over 65 years old, significantly improved patient satisfaction and reduced the incidence of VTE and medical costs.


Assuntos
Artroplastia de Quadril , Artroplastia do Joelho , Tromboembolia Venosa , Assistência ao Convalescente , Idoso , Anticoagulantes/uso terapêutico , Artroplastia de Quadril/efeitos adversos , Artroplastia do Joelho/efeitos adversos , China/epidemiologia , Hospitais , Humanos , Pessoa de Meia-Idade , Alta do Paciente , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/prevenção & controle , Inquéritos e Questionários , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/tratamento farmacológico , Tromboembolia Venosa/epidemiologia
6.
FASEB J ; 35(2): e21370, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33734471

RESUMO

Caffeine has developmental toxicity. Prenatal caffeine exposure (PCE) caused intrauterine growth retardation (IUGR) and multiple organ dysplasia. This study intended to explore the effect and mechanism of PCE on long bone development in female fetal rats. In vivo, the PCE group pregnant rats were given different concentrations of caffeine during the gestational Day 9-20. The mRNA expression of osteogenesis-related genes were significantly reduced in PCE group. In the PCE group (120 mg/kg·d), the length and primary center of fetal femur were shorter, and accompanied by H-type blood vessel abundance reducing. Meanwhile, connective tissue growth factor (CTGF) expression decreased in the growth plate of the PCE group (120 mg/kg·d). In contrast, the miR375 expression increased. In vitro, caffeine decreased CTGF and increased miR375 expression in fetal growth plate chondrocytes. After co-culture with caffeine-treated chondrocytes, the tube formation ability for the H-type endothelial cells was decreased. Furthermore, CTGF overexpression or miR375 inhibitor reversed caffeine-induced reduction of tube formation ability, and miR375 inhibitor reversed caffeine-induced CTGF expression inhibition. In summary, PCE decreased the expression of CTGF by miR375, ultimately resulting in H-type blood vessel-related long bone dysplasia.


Assuntos
Desenvolvimento Ósseo , Doenças do Desenvolvimento Ósseo/etiologia , Cafeína/toxicidade , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Endotélio Vascular/efeitos dos fármacos , MicroRNAs/metabolismo , Efeitos Tardios da Exposição Pré-Natal/etiologia , Animais , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Endotélio Vascular/metabolismo , Feminino , MicroRNAs/genética , Gravidez , Ratos , Ratos Wistar , Transdução de Sinais
7.
Biochem Pharmacol ; 185: 114414, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33434537

RESUMO

Dexamethasone is a common synthetic glucocorticoid drug that can promote foetal lung maturity. An increasing number of studies have shown that prenatal dexamethasone exposure (PDE) can cause a variety of short-term and long-term hazards to offspring, including bone development toxicity. H-type vessels are a newly discovered subtype of blood vessels associated with promoted bone formation and maintenance of bone mass. In this study, we aimed to explore whether H-type blood vessels are involved in PDE-induced long bone development toxicity in offspring and its mechanism. In vivo, we injected dexamethasone (0.2 mg/kg.d) subcutaneously at gestational days 9-20 and observed the H-type vessel abundance and bone mass at different time points in the offspring rats. In vitro, we investigated the effect of dexamethasone (0, 20, 100, and 500 nM) on the tube formation function of rat bone marrow-derived endothelial progenitor cells (EPCs) and explored its mechanism. Our results showed that the adult PDE female offspring rats were susceptible to osteoporosis. In addition, PDE inhibited bone mass, H-type vessel formation and the expression of bone platelet-derived growth factor receptor ß (PDGFRß)/focal adhesion kinase (FAK) pathway-related genes in antenatal and postnatal female offspring. Moreover, PDE promoted the expression of bone glucocorticoid receptor (GR), CCAAT and enhancer binding protein α (C/EBPα) and miR-34c in female foetuses. Dexamethasone suppressed the tube formation of rat bone marrow-derived EPCs and the activity of the PDGFRß/FAK pathway, which was mediated by GR/C/EBPα/miR-34c signalling activation. In summary, PDE can cause H-type vessel dysplasia and high susceptibility to osteoporosis in female offspring, and its mechanism is related to the low-activity programming of the PDGFRß/FAK pathway induced by GR/C/EBPα/miR-34c signalling activation. This study enhances the understanding of the molecular mechanism of dexamethasone-induced bone development toxicity and provides new insights for exploring the early intervention and therapeutic targets of foetal-derived osteoporosis.


Assuntos
Dexametasona/toxicidade , Fêmur/irrigação sanguínea , Fêmur/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Osteoporose/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Dexametasona/administração & dosagem , Feminino , Fêmur/efeitos dos fármacos , Masculino , Osteoporose/induzido quimicamente , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
8.
Br J Pharmacol ; 177(20): 4683-4700, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32757222

RESUMO

BACKGROUND AND PURPOSE: Prenatal caffeine exposure (PCE) can cause developmental toxicity of long bones in offspring, but the long-term effects and the underlying mechanism have not been fully clarified. Here, we investigated the effects of PCE peak bone mass accumulation and osteoporosis susceptibility in offspring and its intrauterine programming mechanism. EXPERIMENTAL APPROACH: Pregnant Wistar rats were administrated intragastrically with saline or caffeine (120 mg·kg-1 ·day-1 ) on gestational days 9-20. The serum and bone samples were collected from the fetal and postnatal offspring for bone mass, genes expression and corticosterone analysis. Then, rat bone marrow mesenchymal stem cells (BMSCs) were treated with corticosterone in vitro to confirm the molecular mechanism. KEY RESULTS: PCE caused fetal bone dysplasia in male and female offspring. In adulthood, PCE reduced peak bone mass and increased osteoporosis susceptibility in male offspring but not in females. Meanwhile, PCE only decreased the H3K9ac and expression levels of 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2) before and after birth in the male offspring but not in the females. Moreover, the high level of corticosterone induced by PCE down-regulated the H3K9ac and expression levels of 11ß-HSD2 through promoting glucocorticoid receptor (GR; NR3C1) into the nucleus of bone marrow mesenchymal stem cells (BMSCs) and recruiting histone deacetylase 11 (HDAC11) binding to 11ß-HSD2 promoter region, which further enhanced the effect of corticosterone on suppressing osteogenic function of BMSCs. CONCLUSION AND IMPLICATIONS: PCE caused osteoporosis susceptibility in male adult offspring, which attributed to the low-functional programming of 11ß-HSD2 induced by corticosterone via GR/HDAC11 signalling.


Assuntos
Osteoporose , Efeitos Tardios da Exposição Pré-Natal , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2 , 11-beta-Hidroxiesteroide Desidrogenases , Animais , Cafeína/toxicidade , Feminino , Glucocorticoides , Masculino , Osteoporose/induzido quimicamente , Gravidez , Ratos , Ratos Wistar
9.
Bone ; 133: 115245, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31962170

RESUMO

Prenatal dexamethasone exposure (PDE) induces developmental toxicities of multi-organs and susceptibility to multi-diseases in offspring. However, the effects of PDE on osteoarthritis susceptibility in adult offspring and its mechanism have not been reported. In the present study, we treated pregnant Wistar rats with dexamethasone (0.2 mg/kg) daily on gestational days (GD) 9-20. Some pregnant rats were sacrificed on GD20, and the rest were delivered to obtain the postnatal offspring. The adult female offspring rats were performed with ovariectomy or sham operation during postnatal weeks 22-28. We found that PDE led to osteoarthritis phenotypes in articular cartilage and an increase in modified Mankin's score, but reduced the cartilage thickness in female adult offspring rats, which were more evident after ovariectomy. Moreover, PDE reduced the bone mass of subchondral bone in female adult offspring, which was aggravated by ovariectomy. The correlation analysis results indicated that the osteoarthritic phenotype and cartilage thickness were closely associated with the decreased bone mass of subchondral bone induced by PDE. Further, PDE retarded the development of primary and secondary ossification centers, then led to subchondral bone dysplasia, which could be partly mediated by the inhibited osteogenic function before and after birth. Collectively, the subchondral bone dysplasia partly participated in osteoarthritis susceptibility induced by PDE in female offspring rats.


Assuntos
Doenças do Desenvolvimento Ósseo , Cartilagem Articular , Osteoartrite , Efeitos Tardios da Exposição Pré-Natal , Animais , Dexametasona/toxicidade , Feminino , Osteoartrite/induzido quimicamente , Gravidez , Ratos , Ratos Wistar
10.
Food Chem Toxicol ; 136: 111083, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31887396

RESUMO

Prenatal ethanol exposure induces developmental toxicities of multiple organs in offspring. Here, we investigate the effects of prenatal ethanol exposure on bone mass in postnatal offspring and explore its intrauterine programming mechanism. We found that prenatal ethanol exposure could induce bone dysplasia in fetuses and postnatal osteopenia in female offspring, accompanied by the sustained activation of the local renin-angiotensin systems (RAS) and inhibition of bone formation. Additionally, we also found that histone 3 lysine 9 acetylation (H3K9ac) and H3K27ac levels in the promoter region of angiotensin-converting enzyme (ACE) were increased in female offspring exposed to ethanol during pregnancy. In vitro, ethanol suppressed the formation of mineralized nodules and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), which was blocked by enalapril. Furthermore, ethanol promoted the expression and nuclear translocation of early growth response factor 1 (Egr1), which participated in the promotion of histone acetylation of ACE and subsequent RAS activation, by recruiting p300 and binding to the ACE promoter region directly. These findings indicate that the sustained activation of the local RAS might participate in bone dysplasia in fetus and postnatal osteopenia in the female offspring, while the Egr1/p300/ACE signal might be a key promoter of the sustained activation of the local RAS of the long bone.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Etanol/efeitos adversos , Exposição Materna/efeitos adversos , Peptidil Dipeptidase A/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Animais , Doenças Ósseas Metabólicas/etiologia , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/fisiopatologia , Proteína 1 de Resposta de Crescimento Precoce/genética , Feminino , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Osteogênese , Peptidil Dipeptidase A/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Wistar , Fatores Sexuais , Fatores de Transcrição de p300-CBP/genética
11.
FASEB J ; 33(11): 12972-12982, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31500447

RESUMO

Prenatal nicotine exposure (PNE) induces developmental toxicity in offspring. However, the long-term harmful effects on bone development and the intrauterine programming mechanism attributed to PNE remain unclear. In the present research, pregnant Wistar rats were injected subcutaneously with nicotine (2 mg/kg/d) to obtain and analyze bone samples from the fetal and adult offspring. Bone marrow mesenchymal stem cells (BMSCs) were treated with nicotine during osteogenic differentiation to clarify the related molecular mechanisms. The results indicated that PNE led to bone dysplasia in the fetuses and reduced bone mass in the adult offspring, which was mediated by the sustained activation of the local bone renin angiotensin system (RAS) and suppressed osteogenic differentiation before and after birth. In vitro, nicotine suppressed BMSCs' osteogenic function through promoting angiotensin-converting enzyme (ACE) expression and activating RAS. Furthermore, nicotine induced histone acetylase p300 into the nuclei of the BMSCs by acting on the α4ß2-nicotinic acetylcholine receptor (α4ß2-nAChR), leading to the increased histone 3 lysine 9 acetylation level of ACE and RAS activation. Taken together, the sustained activation of local bone RAS mediated prenatal nicotine-induced osteopenia in adult offspring via the α4ß2-nAChR-p300-ACE pathway.-Xiao, H., Wen, Y., Pan, Z., Shangguan, Y., Magdalou, J., Wang, H., Chen, L. Nicotine exposure during pregnancy programs osteopenia in male offspring rats via α4ß2-nAChR-p300-ACE pathway.


Assuntos
Doenças Ósseas Metabólicas/induzido quimicamente , Antígenos CD18/metabolismo , Integrina alfa4/metabolismo , Exposição Materna , Nicotina/administração & dosagem , Peptidil Dipeptidase A/metabolismo , Receptores Nicotínicos/metabolismo , Animais , Feminino , Masculino , Gravidez , Ratos , Ratos Wistar
12.
Orthop Surg ; 11(2): 229-235, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30945802

RESUMO

OBJECTIVES: To explore the safety and efficacy of the enhanced recovery after surgery (ERAS) program for elderly total knee arthroplasty (TKA) patients. METHODS: A prospective controlled study was conducted for patients older than 65 years, who would undergo unilateral TKA with a minimum follow-up of 2 years. Patients were divided into an ERAS group (n = 106) and a traditional group (n = 141) based on the patients' willingness to participate in the ERAS program. Baseline parameters of American Society of Anesthesiologists classification and comorbidity were recorded. Complication, mortality, knee function assessment using knee society score and knee range of motion, and perioperative clinical outcomes were compared between the two groups. RESULTS: There were no significant differences between the two groups in terms of baseline parameters. Although no significant differences were found in postoperative nausea and vomiting, urinary tract infection, deep venous thrombosis, pulmonary embolism, wound delayed healing, superficial infection, and deep infection, there were significantly fewer total complications in the ERAS group (26/106 vs 52/141; P = 0.039). No significant difference was found in short-term mortality (1/106 vs 3/141; P = 0.836) between the two groups. There were no significant differences in preoperative visual analogue scale (VAS), knee society score (KSS), and range of motion (ROM) between the two groups. Lower VAS scores were found in the ERAS group at time of postoperative day (POD) 1 (P = 0.012) and POD 5 (P = 0.020); no significant differences were observed at time of postoperative month (POM) 1 and final follow-up. Higher KSS scores were found in the ERAS group at time of POD 1 (P = 0.013), and POD 5 (P = 0.011), no significant differences were observed at time of POM 1 and final follow-up. Increased ROM degree was found in the ERAS group at time of POD 1 (P = 0.021); no significant differences were observed at time of POD 5, POM 1 and final follow-up. Decreased intraoperative blood loss (P < 0.001), total blood loss (P < 0.001), transfusion rate (P = 0.004), and length of stay (P < 0.001) were found in the ERAS group; no significant differences were found in operative time and hospitalization costs between the two groups. CONCLUSION: The ERAS program is safer and more efficacious in elderly TKA patients compared to the traditional pathway. It could effectively relieve perioperative pain and improve joint function, and reduce blood transfusion, length of stay, and total complications without increasing short-term mortality.


Assuntos
Artroplastia do Joelho/reabilitação , Cuidados Pós-Operatórios/métodos , Idoso , Feminino , Humanos , Masculino , Medição da Dor , Dor Pós-Operatória/epidemiologia , Estudos Prospectivos , Amplitude de Movimento Articular , Recuperação de Função Fisiológica , Resultado do Tratamento
13.
Toxicol Appl Pharmacol ; 363: 1-10, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30423288

RESUMO

This study was aimed to investigate whether and how prenatal caffeine exposure (PCE) could induce osteopenia in the adult offspring. Pregnant rats were treated with prenatal caffeine 12 mg/100 g body weight per day from pregnant day 9 to 20, while rat bone marrow mesenchymal stem cells (BMSCs) were treated with exogenous corticosterone during osteogenic induction. Shorter femur and primary ossification center was observed in the PCE offspring, as well as less bone trabecular and poor biomechanical intensity. Local gene expression of glucocorticoid receptor (GR) and angiotensin converting enzyme (ACE), as well as angiotensin 2 content, was found to be stimulated, while the expression of bone gamma-carboxyglutamate protein (BGLAP), alkaline phosphatase (ALP) and bone sialoprotein (BSP) was found to be suppressed, with hypomethylation of ACE promoter. Corticosterone (1250 nM) suppressed osteogenic differentiation of BMSCs and gene expression of BGLAP, ALP and BSP, which was attenuated by enalapril, while it stimulated ACE mRNA expression and induced hypomethylation of ACE promoter, which was attenuated by mifepristone. It indicated that PCE caused bone growth retardation and adult osteopenia in offspring, which might be triggered by the activation of local RAS induced by excessive maternal glucocorticoid, while the hypomethylation of ACE gene might be the key point of the sustained activation of the local RAS.


Assuntos
Doenças Ósseas Metabólicas/induzido quimicamente , Cafeína/efeitos adversos , Glucocorticoides/metabolismo , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Sistema Renina-Angiotensina/fisiologia , Animais , Doenças Ósseas Metabólicas/patologia , Células da Medula Óssea , Osso e Ossos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/fisiologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Masculino , Células-Tronco Mesenquimais , Osteogênese/efeitos dos fármacos , Peptidil Dipeptidase A/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Cultura Primária de Células , Ratos , Ratos Wistar , Receptores de Glucocorticoides/metabolismo , Organismos Livres de Patógenos Específicos
14.
Am J Pathol ; 188(12): 2863-2876, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273601

RESUMO

Epidemiologic investigations suggest that excessive intake of caffeine during pregnancy is one of the risk factors for osteoporosis in adult offspring. However, the phenomena and mechanisms have remained obscure. This study found that prenatal caffeine exposure (PCE) leads to persistent bone dysplasia in gestational day 20 and postnatal week 12 offspring rats and increases the susceptibility to osteoporosis in postnatal week 28 offspring rats. In the embryonic period, PCE increases the concentration of serum corticosterone and inhibits the expression of insulin-like growth factor-1 (IGF1) and osteogenic differentiation genes. After birth, the recovery of IGF1 expression in PCE offspring is unable to completely compensate osteogenic function, and chronic stress can lead to a further decrease in IGF1 expression. In vitro experiments found that corticosterone instead of caffeine restrains mineralized nodule formation and osteoblast differentiation by inhibiting IGF1 expression. The corticosterone inhibits H3K9 and H3K14 histone acetylation of IGF1 in osteoblasts through glucocorticoid receptor and CCAAT and enhancer binding protein α, respectively. In conclusion, glucocorticoid instead of caffeine inhibits bone IGF1 expression via glucocorticoid receptor and CCAAT and enhancer binding protein α and mediates the PCE-induced bone dysplasia and bone mass reduction in offspring fetal rats, which may contribute to osteoporosis susceptibility in adulthood.


Assuntos
Cafeína/toxicidade , Reprogramação Celular/efeitos dos fármacos , Retardo do Crescimento Fetal/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Osteoporose/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Estimulantes do Sistema Nervoso Central/toxicidade , Feminino , Retardo do Crescimento Fetal/induzido quimicamente , Retardo do Crescimento Fetal/metabolismo , Glucocorticoides/efeitos adversos , Osteogênese , Osteoporose/induzido quimicamente , Osteoporose/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Wistar
15.
Toxicol Lett ; 295: 249-255, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29981921

RESUMO

This study investigated the mechanisms underlying the retarded development of long bone in fetus by prenatal nicotine exposure (PNE) which had been demonstrated by our previous work. Nicotine (2.0 mg/kg.d) or saline was injected subcutaneously into pregnant rats every morning from gestational day (GD) 9 to 20. Fetal femurs or tibias were harvested for analysis on GD 20. We found massive accumulation of hypertrophic chondrocytes and a delayed formation of primary ossification center (POC) in the fetal femur or tibia of rat fetus after PNE, which was accompanied by a decreased amount of osteoclasts in the POC and up-regulated expression of osteoprotegerin (OPG) but by no obvious change in the expression of receptor activator of NF-κB ligand (RANKL). In primary osteoblastic cells, both nicotine (0, 162, 1620, 16,200 ng/ml) and corticosterone (0, 50, 250, 1250 nM) promoted the mRNA expression of OPG but concentration-dependently suppressed that of RANKL. Furthermore, blocking α4ß2-nicotinic acetylcholine receptor (α4ß2-nAChR) or glucocorticoid receptor rescued the above effects of nicotine and corticosterone, respectively. In conclusion, retarded osteoclastogenesis may contribute to delayed endochondral ossification in long bone in fetal rats with PNE. The adverse effects of PNE may be mediated via the direct effect of nicotine and indirect effect of maternal corticosterone on osteoblastic cells.


Assuntos
Desenvolvimento Ósseo/efeitos dos fármacos , Fêmur/efeitos dos fármacos , Nicotina/toxicidade , Agonistas Nicotínicos/toxicidade , Osteogênese/efeitos dos fármacos , Tíbia/efeitos dos fármacos , Animais , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Corticosterona/toxicidade , Feminino , Fêmur/embriologia , Fêmur/metabolismo , Idade Gestacional , Exposição Materna/efeitos adversos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteoprotegerina/metabolismo , Gravidez , Ligante RANK/metabolismo , Ratos Wistar , Receptores de Glucocorticoides/metabolismo , Receptores Nicotínicos/metabolismo , Tíbia/embriologia , Tíbia/metabolismo
16.
Cell Death Dis ; 9(6): 638, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844424

RESUMO

Prenatal dexamethasone exposure (PDE) induces developmental toxicities of multiple organs in offspring. Here, we verified the intergenerational effect of low peak bone mass induced by PDE and investigated its intrauterine programming mechanism. Pregnant rats were injected subcutaneously with 0.2 mg/kg/d dexamethasone from gestation day (GD) 9 to 20. Some pregnant rats were killed for the fetuses on GD20, and the rest went on to spontaneous labor to produce the first-generation (F1) offspring. The adult F1 male offspring were mated with normal females to produce the F2 offspring. In vivo, PDE leads to low peak bone mass in F1 male offspring rats at postnatal week (PW) 28. Furthermore, PDE reduced the bone mass in F1 male offspring from GD20 to PW12. Meanwhile, the osteogenic differentiation was suppressed and the local renin-angiotensin system (RAS) was activated continuously by PDE. Moreover, the histone 3 lysine 27 acetylation (H3K27ac) level in angiotensin-converting enzyme (ACE) promoter region was increased by PDE from GD20 to PW12. Likewise, PDE induced the low peak bone mass and the activated local RAS in F2 male offspring. Meaningfully, the H3K27ac level of ACE was increased by PDE in the F2 offspring. In vitro, dexamethasone inhibited bone marrow mesenchymal stem cells (BMSCs) osteogenic differentiation and promoted RAS activation. Furthermore, dexamethasone recruited CCAAT/enhancer-binding protein α and p300 into the BMSCs nucleus by activating glucocorticoid receptor, which cooperatively increased the H3K27ac level in the ACE promoter region. In conclusion, PDE induced the low peak bone mass and its intergenerational effect, which was mediated by sustained activation of RAS via increasing H3K27ac level of ACE.


Assuntos
Osso e Ossos/patologia , Dexametasona/efeitos adversos , Histonas/metabolismo , Lisina/metabolismo , Peptidil Dipeptidase A/metabolismo , Efeitos Tardios da Exposição Pré-Natal/enzimologia , Acetilação , Animais , Diferenciação Celular/efeitos dos fármacos , Cruzamentos Genéticos , Feminino , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Linhagem , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos Wistar , Sistema Renina-Angiotensina/efeitos dos fármacos
17.
Cell Death Dis ; 8(10): e3157, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29072695

RESUMO

Our previous studies discovered that prenatal caffeine exposure (PCE) could induce intrauterine growth retardation (IUGR) and long-bone dysplasia in offspring rats, accompanied by maternal glucocorticoid over-exposure. This study is to explore whether intrauterine high glucocorticoid level can cause endochondral ossification retardation and clarify its molecular mechanism in PCE fetal rats. Pregnant Wistar rats were intragastrically administered 30 and 120 mg/kg day of caffeine during gestational days (GDs) 9-20, then collected fetal serum and femurs at GD20. In vitro, primary chondrocytes were treated with corticosterone (0-1250 nM), caffeine (0-100 µM), mitogen-inducible gene 6 (Mig-6) siRNA and epidermal growth factor receptor (EGFR) siRNA, respectively, or together. Results showed that the hypertrophic chondrocytes zone (HZ) of PCE fetal femur was widened. Meanwhile, the expression levels of chondrocytes terminal differentiation genes in the HZ were decreased, and the chondrocytes apoptosis rate in the HZ was decreased too. Furthermore, PCE upregulated Mig-6 and suppressed EGFR expression in the HZ. In vitro, a high-concentration corticosterone (1250 nM) upregulated Mig-6 expression, inhibit EGFR/c-Jun N-terminal kinase (JNK) signaling pathway and terminal differentiation genes expression in chondrocytes and reduced cell apoptosis, and these above alterations could be partly reversed step-by-step after Mig-6 and EGFR knockdown. However, caffeine concentration dependently increased chondrocyte apoptosis without significant changes in the expression of terminal differentiation genes. Collectively, PCE caused endochondral ossification retardation in the female fetal rats, and its main mechanism was associated with glucocorticoid (rather than caffeine)-mediated chondrocyte terminal differentiation suppression by the upregulation of Mig-6 and then inhibition of EGFR/JNK pathway-mediated chondrocyte apoptosis.


Assuntos
Cafeína/efeitos adversos , Retardo do Crescimento Fetal/etiologia , Glucocorticoides/efeitos adversos , Osteogênese/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Diferenciação Celular , Feminino , Retardo do Crescimento Fetal/patologia , Feto , Gravidez , Ratos , Ratos Wistar
18.
Toxicol Appl Pharmacol ; 305: 234-241, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27338645

RESUMO

Prenatal ethanol exposure (PEE) inhibits longitudinal growth of fetal bones, but the underlying mechanisms remain unknown. In this study, we aimed to investigate how PEE induces the retardation of long bone development in fetal rats. Pregnant Wistar rats were treated with ethanol or distilled water (control group) by gavage from gestational day (GD) 9 to 20. Fetuses were delivered by cesarean section on GD20. Fetal sera were collected for assessing corticosterone (CORT) level. Fetal long bones were harvested for histochemical, immunohistochemical and gene expression analysis. Primary chondrocytes were treated with ethanol or CORT for analyzing genes expression. PEE fetuses showed a significant reduction in birth weight and body length. The serum CORT concentration in PEE group was significantly increased, while the body weight, body length and femur length all were significantly decreased in the PEE group. The length of the epiphyseal hypertrophy zone was enlarged, whereas the length of the primary ossification center was significantly reduced in PEE fetuses. TUNEL assay showed reduced apoptosis in the PEE group. Further, the gene expression of osteoprotegerin (OPG) was markedly up-regulated. In vitro experiments showed that CORT (but not ethanol) treatment significantly activated the expression of OPG, while the application of glucocorticoid receptor inhibitor, mifepristone, attenuated these change induced by CORT. These results indicated that PEE-induced glucocorticoid over-exposure enhanced the expression of OPG in fetal epiphyseal cartilage and further lead to the suppressed osteoclast differentiation in the chondro-osseous junction and consequently inhibited the endochondral ossification in long bones of fetal rats.


Assuntos
Etanol/toxicidade , Fêmur/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Feminino , Fêmur/crescimento & desenvolvimento , Feto , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Gravidez , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...