Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 578
Filter
Add more filters

Publication year range
1.
J Bone Miner Metab ; 42(2): 242-252, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38498197

ABSTRACT

INTRODUCTION: This study was to investigate the correlations between pyrethroid exposure and bone mineral density (BMD) and osteopenia. MATERIALS AND METHODS: This cross-sectional study included 1389 participants over 50 years of age drawn from the 2007-2010 and 2013-2014 National Health and Nutrition Examination Survey (NHANES). Three pyrethroid metabolites, 3-phenoxybenzoic acid (3-PBA), trans-3-(2,2-dichlorovinyl)-2,2-dimethyl-cyclopropane-1-carboxylic acid (trans-DCCA), and 4-fluoro-3-phenoxybenzoic acid (4-F-3PBA) were used as indicators of pyrethroid exposure. Low BMD was defined as T-score < - 1.0, including osteopenia. Weighted multivariable linear regression analysis or logistic regression analysis was utilized to evaluate the correlation between pyrethroid exposure and BMD and low BMD. Bayesian kernel machine regression (BKMR) model was utilized to analyze the correlation between pyrethroids mixed exposure and low BMD. RESULTS: There were 648 (48.41%) patients with low BMD. In individual pyrethroid metabolite analysis, both tertile 2 and tertile 3 of trans-DCCA were negatively related to total femur, femur neck, and total spine BMD [coefficient (ß) = - 0.041 to - 0.028; all P < 0.05]. Both tertile 2 and tertile 3 of 4-F-3PBA were negatively related to total femur BMD (P < 0.05). Only tertile 2 [odds ratio (OR) = 1.63; 95% CI = 1.07, 2.48] and tertile 3 (OR = 1.65; 95% CI = 1.10, 2.50) of trans-DCCA was correlated with an increased risk of low BMD. The BKMR analysis indicated that there was a positive tendency between mixed pyrethroids exposure and low BMD. CONCLUSION: In conclusion, pyrethroids exposure was negatively correlated with BMD levels, and the associations of pyrethroids with BMD and low BMD varied by specific pyrethroids, pyrethroid concentrations, and bone sites.


Subject(s)
Benzoates , Bone Diseases, Metabolic , Insecticides , Phenyl Ethers , Pyrethrins , Adult , Humans , Middle Aged , Pyrethrins/adverse effects , Pyrethrins/analysis , Pyrethrins/metabolism , Insecticides/adverse effects , Insecticides/analysis , Insecticides/metabolism , Nutrition Surveys , Cross-Sectional Studies , Bone Density , Bayes Theorem , Environmental Exposure/adverse effects , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/epidemiology
2.
J Infect Dis ; 228(6): 742-750, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37225667

ABSTRACT

BACKGROUND: Bone mineral density (BMD) loss may be accelerated in people with HIV (PLWH). It is unknown whether a polygenic risk score (PRS) is associated with low BMD in PLWH. METHODS: Swiss HIV Cohort Study participants of self-reported European descent underwent ≥2 per-protocol dual x-ray absorptiometry (DXA) measurements ≥2 years apart (2011-2020). Univariable and multivariable odds ratios (ORs) for DXA-defined osteoporosis were based on traditional and HIV-related risk factors and a genome-wide PRS built from 9413 single-nucleotide polymorphisms associated with low BMD in the general population. Controls were free from osteoporosis/osteopenia on all DXA measurements. RESULTS: We included 438 participants: 149 with osteoporosis and 289 controls (median age, 53 years; 82% male, 95% with suppressed HIV RNA). Participants with unfavorable osteoporosis PRS (top vs bottom quintile) had univariable and multivariable-adjusted osteoporosis ORs of 4.76 (95% CI, 2.34-9.67) and 4.13 (1.86-9.18), respectively. For comparison, hepatitis C seropositivity, 5-year tenofovir disoproxil fumarate exposure, and parent history of hip fracture yielded univariable osteoporosis ORs of 2.26 (1.37-3.74), 1.84 (1.40-2.43), and 1.54 (0.82-2.9). CONCLUSIONS: In PLWH in Switzerland, osteoporosis was independently associated with a BMD-associated PRS after adjustment for established risk factors, including exposure to tenofovir disoproxil fumarate.


Subject(s)
Bone Diseases, Metabolic , HIV Infections , Osteoporosis , Humans , Male , Middle Aged , Female , Cohort Studies , HIV , Switzerland/epidemiology , Osteoporosis/epidemiology , Osteoporosis/genetics , Osteoporosis/chemically induced , HIV Infections/complications , HIV Infections/epidemiology , Risk Factors , Bone Density/genetics , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/epidemiology , Tenofovir/adverse effects
3.
Osteoporos Int ; 34(2): 291-297, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36402867

ABSTRACT

In patients with non-metastatic prostate cancer, treated with radiation therapy and androgen deprivation therapy for 3 years and DMAB on average for 5 years, BMD was in the normal or osteopenic range. Discontinuation of DMAB led to a bone loss of 2-5%. In men with osteopenia, the bone loss was prevented by zoledronate. PURPOSE: Patients with prostate cancer receiving androgen deprivation therapy (ADT) are treated with denosumab (DMAB) to prevent fractures and preserve bone mass. We wanted to investigate the change in BMD in men with non-metastatic prostate cancer discontinuing DMAB. METHODS: We conducted a retrospective cohort study based on medical records from patients referred to the Department of Endocrinology from the Department of Urology, Aarhus University Hospital between June 1, 2018, and June 1, 2021. We retrieved information on biochemistry and DXA performed 0-6 months after the last DMAB injection and a second DXA performed approximately 12 months after the first. In case of a BMD T-score ≤ - 1 at the lumbar spine or total hip at the first DXA, the patients were treated with zoledronate. The primary endpoint was change in lumbar spine BMD. RESULTS: We included 50 patients with non-metastatic prostate cancer. The mean DMAB treatment duration was 5 ± 0.1 years. Among the patients treated with zoledronate (n = 9), BMD was maintained at the spine and femoral neck after a mean of 16 months. We found a significant decrease in BMD; - 4.9 ± 4.2%, - 1.9 ± 3.5%, and - 2.4 ± 3.6% at the spine, total hip, and femoral neck between the first and second DXA in the patients not treated with zoledronate (n = 24) (p ≤ 0.01 for all). One patient who did not receive ZOL sustained multiple fragility vertebral fractures after DMAB discontinuation. CONCLUSION: In men with non-metastatic prostate cancer, discontinuation of DMAB after stopping ADT led to an average bone loss of 2-5%. Zoledronate prevented bone loss in men with osteopenia.


Subject(s)
Bone Density Conservation Agents , Bone Diseases, Metabolic , Prostatic Neoplasms , Male , Humans , Denosumab/adverse effects , Bone Density Conservation Agents/adverse effects , Zoledronic Acid/therapeutic use , Retrospective Studies , Androgen Antagonists/adverse effects , Androgens , Prostatic Neoplasms/complications , Prostatic Neoplasms/drug therapy , Bone Density , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/drug therapy
4.
Calcif Tissue Int ; 112(1): 34-44, 2023 01.
Article in English | MEDLINE | ID: mdl-36287217

ABSTRACT

There is limited evidence on the use of romosozumab (ROMO) in the treatment of osteoporosis in patients on hemodialysis (HD); thus, we aimed to investigate this topic. This prospective, observational, single-center cohort study included 13 prior osteoporosis treatment-naïve patients on HD with osteoporosis. They first received ROMO once monthly for 12 months (210 mg; subcutaneously once every month). Thereafter, they received denosumab (DENO) for an additional 12 months (60 mg; subcutaneously once every 6 months). We examined the incidence of new fractures; treatment safety; and temporal changes in the bone mineral density (BMD), bone metabolism markers, and vascular calcification. No new cases of fractures were noted. The median one-year percentage changes (from the baseline) in the BMDs at the lumbar spine (LS), total hip (TH), and femoral neck (FN) were + 9.0%, + 2.5%, and + 4.7%, respectively. These changes were maintained for 24 months. The corresponding relative changes from the baseline to 24 months thereafter were + 14.9%, + 5.4%, and + 6.5%, respectively. The percentage changes in TH BMD and FN BMD were negatively correlated with baseline BMD. Coronary artery and thoracic aorta calcification scores increased slightly from baseline to 12 months thereafter. However, fatal events (cardiovascular disease-associated and all-cause deaths) did not occur during ROMO treatment. Effectiveness of ROMO was better in patients who had severe osteoporosis with low TH BMD, low FN BMD, and high tartrate-resistant acid phosphatase 5b level at ROMO initiation.


Subject(s)
Bone Density Conservation Agents , Bone Diseases, Metabolic , Fractures, Bone , Osteoporosis, Postmenopausal , Osteoporosis , Humans , Female , Denosumab/pharmacology , Denosumab/therapeutic use , Prospective Studies , Cohort Studies , Osteoporosis/drug therapy , Osteoporosis/chemically induced , Bone Density , Fractures, Bone/epidemiology , Bone Diseases, Metabolic/chemically induced , Renal Dialysis
5.
J Periodontal Res ; 58(2): 283-295, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36575324

ABSTRACT

BACKGROUND AND OBJECTIVES: Periodontitis is a highly prevalent disease in psychiatric patients, including those undergoing symptomatic treatment with second-generation antipsychotics. Some of these drugs, such as clozapine (CLO) and olanzapine (OLA), have prominent metabolic effects such as weight gain, hyperglycemia, and dyslipidemia, which are risk factors for periodontitis. In addition to the metabolic effects, there are reports of changes in salivary flow, gingival bleeding, and caries. In this context, we aimed to evaluate if the metabolic effects of OLA and CLO alter periodontal parameters in an animal model of periodontitis without the environmental and psychosocial biases inherent to human diseases. METHODS: In the first set of experiments, male and female adult Wistar rats received oral administration of CLO, OLA, or vehicle for 45 days. They were evaluated for body mass composition and weight gain, blood glucose parameters (fasting and glucose tolerance and insulin resistance tests), and lipid profile (HDL, total cholesterol, and triglycerides). In a second set of experiments, the same measurements were performed in female rats exposed to the antipsychotics for 45 days and ligature-induced periodontitis on the 30th day of treatment. Macroscopic measurements of exposed roots, microtomography in the furcation region of the first molar, and histological evaluation of the region between the first and second molars were evaluated to assess bone loss. Additionally, gingival measurements of myeloperoxidase activity and pro-inflammatory cytokine TNF-α were made. RESULTS: Only females exposed to OLA had more significant weight gain than controls. They also exhibited differences in glucose metabolism. Ligature-induced periodontitis produced intense bone retraction without changing the density of the remaining structures. The bone loss was even higher in rats with periodontitis treated with OLA or CLO and was accompanied by a local increase in TNF-α caused by CLO. These animals, however, did not exhibit the same metabolic impairments observed for animals without periodontitis. CONCLUSION: The use of clozapine and olanzapine may be a risk factor for periodontal disease, independent of systemic metabolic alterations.


Subject(s)
Antipsychotic Agents , Bone Diseases, Metabolic , Clozapine , Periodontitis , Humans , Adult , Rats , Male , Female , Animals , Antipsychotic Agents/adverse effects , Clozapine/adverse effects , Olanzapine/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Rats, Wistar , Periodontitis/complications , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/drug therapy , Weight Gain
6.
J Antimicrob Chemother ; 77(7): 1974-1979, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35512339

ABSTRACT

BACKGROUND: Tenofovir disoproxil fumarate, particularly when given with a ritonavir-boosted PI, reduces bone mineral density (BMD) and increases bone turnover markers (BTMs). Ritonavir-boosted atazanavir plus lamivudine is a feasible simplified option. We evaluated whether switching from a triple ritonavir-boosted PI plus tenofovir disoproxil fumarate to a two-drug regimen of lamivudine plus ritonavir-boosted atazanavir would improve BMD. METHODS: Single-arm pilot study. Virologically suppressed patients on tenofovir disoproxil fumarate plus lamivudine or emtricitabine plus ritonavir-boosted PI with low BMD, without previous resistance mutations and/or virological failure to study drugs were switched to 100/300 mg of ritonavir-boosted atazanavir plus 300 mg of lamivudine once daily. The primary endpoint was BMD change by DXA at Week 48. RESULTS: There were 31 patients, 4 (13%) female, and median age was 40 years. Seven participants (22.5%) had osteoporosis. At 48 weeks, mean (SD) changes in spine and hip BMD were +0.01 (0.03) (P = 0.0239) and +0.013 (0.03) g/cm2 (P = 0.0046), respectively. Mean (SD) T-score changes were +0.1 (0.23) (P = 0.0089) and +0.25 (0.76) (P = 0.0197), respectively. N-telopeptide and urine tenofovir disoproxil fumarate toxicity markers showed significant improvements. One participant withdrew from the study and two were lost to follow-up. There were no virological failures, or serious or grade 3-4 adverse events. CONCLUSIONS: Switching from a tenofovir disoproxil fumarate plus ritonavir-boosted PI triple therapy to a lamivudine plus ritonavir-boosted atazanavir two-drug regimen in virologically suppressed HIV-infected adults with low BMD was safe, increased low BMD and reduced plasma markers of bone turnover and urine markers of tenofovir disoproxil fumarate toxicity over 48 weeks.


Subject(s)
Anti-HIV Agents , Bone Diseases, Metabolic , Drug Substitution , HIV Infections , HIV Protease Inhibitors , Adult , Anti-HIV Agents/therapeutic use , Atazanavir Sulfate/therapeutic use , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/prevention & control , Emtricitabine/therapeutic use , Female , HIV Infections/complications , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Humans , Lamivudine/therapeutic use , Male , Pilot Projects , Ritonavir/adverse effects , Ritonavir/therapeutic use , Tenofovir/adverse effects , Tenofovir/therapeutic use
7.
Mol Pharm ; 19(11): 4000-4009, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36042532

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability among children and young adults in the United States. In this manuscript, we assessed the utility of an N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer-based dexamethasone (Dex) prodrug (P-Dex) in the treatment of TBI. Using a controlled cortical impact TBI mouse model, P-Dex was found to passively target and sustain at the traumatic/inflammatory brain tissue for over 14 days after systemic administration. The histological evidence supports P-Dex's therapeutic potential in ameliorating neuroinflammation and mitigating neurodegeneration. Behaviorally, the P-Dex-treated animals showed statistically significant improvement in balance recovery. A trend of neurological severity score improvement at the early time point post-TBI was also noted but did not achieve statistical significance. While probing the potential glucocorticoid side effects that may associate with P-Dex treatment, we discovered that the TBI mice develop osteopenia. Interestingly, the P-Dex-treated TBI mice demonstrated higher bone mineral density and better bone microarchitecture parameters when compared to free Dex and the saline control, revealing the osteoprotective effect of P-Dex in addition to its neuronal protection benefits post-TBI.


Subject(s)
Bone Diseases, Metabolic , Brain Injuries, Traumatic , Prodrugs , Mice , Animals , Prodrugs/therapeutic use , Dexamethasone/therapeutic use , Neuroinflammatory Diseases , Macromolecular Substances , Brain Injuries, Traumatic/drug therapy , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/drug therapy , Disease Models, Animal
8.
Int J Clin Oncol ; 27(9): 1439-1449, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35908137

ABSTRACT

BACKGROUND: This study was performed to assess the impact of osteopenia on chemotherapy-induced neutropenia and the prognosis for patients treated with FOLFOXIRI for colorectal cancer. METHODS: In total, 77 patients who underwent FOLFOXIRI for un-resectable metastatic and advanced colorectal cancer were retrospectively evaluated. Osteopenia was evaluated by the bone mineral density, which was measured using the average pixel density of the trabecular bone in the 11th thoracic vertebra by computed tomography before the introduction of chemotherapy. The relationship between osteopenia and neutropenia was evaluated. Progression-free survival and overall survival of patients with osteopenia and patients with neutropenia were evaluated. RESULTS: Grade ≥ 3 neutropenia was significantly more common in patients with than without osteopenia (p = 0.002). The multivariate analysis showed that osteopenia was a significant independent predictive factor for grade ≥ 3 neutropenia (p = 0.016). There was no significant difference in progression-free survival or overall survival between patients with and without osteopenia. Patients with grade ≥ 3 neutropenia tended to have a higher progression-free survival rate than others (p = 0.059). The overall survival rate was significantly higher in patients with grade ≥ 3 neutropenia than in others (p = 0.011). CONCLUSION: Osteopenia might be a predictor of chemotherapy-induced neutropenia, and neutropenia might be a prognostic factor for progression-free survival and overall survival in patients with colorectal cancer treated with FOLFOXIRI.


Subject(s)
Antineoplastic Agents , Bone Diseases, Metabolic , Colorectal Neoplasms , Neutropenia , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/drug therapy , Camptothecin/analogs & derivatives , Colorectal Neoplasms/pathology , Fluorouracil/adverse effects , Humans , Leucovorin/adverse effects , Neutropenia/chemically induced , Organoplatinum Compounds , Retrospective Studies
9.
BMC Pediatr ; 22(1): 437, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864501

ABSTRACT

BACKGROUND: Caffeine is a routinely prescribed pharmacological active compound in neonatal intensive care units (NICU) for treating apnea of prematurity (AOP), which also decreases the risk of bronchopulmonary dysplasia and cerebral palsy in neonates. Caffeine-induced excessive calcium loss can promote the development of metabolic bone disease (MBD) in preterm neonates. This study aimed to evaluate the effect of the caffeine regimen on the development of osteopenia of prematurity (OOP), using serum alkaline phosphatase (serum-ALP) concentrations as a surrogate marker at the 4th week of life. METHODS: This retrospective cohort study was conducted including neonates of < 32 weeks gestational age (GA) and birth weight < 1500 g, admitted to NICU from April-2017 to December-2018 and received caffeine therapy till 28 days of life for AOP. Based on serum-ALP levels, formed the high and low-ALP groups. Neonatal characteristics, caffeine regimen, risk factors for OOP, including duration of parenteral nutrition (PN), exposure to medicines associated with MBD, and intake of essential vitamins and minerals, were compared in both groups. Predictors of OOP were analyzed through logistic regression. RESULTS: From the total of 268 participants, 52 (19%) developed OOP, mostly female (61.5%). In the high ALP group, the serum-ALP levels were significantly higher than in the low-ALP group (725.0 ± 143.8 vs 273.6 ± 55.0 units/L, p < 0.001). The high-ALP group received significantly (p < 0.001) higher daily and cumulative caffeine doses and were associated with a higher likelihood of developing OOP in this study cohort [cumulative dose (mg) (AOR = 1.082 95% CI 1.011 to 1.157) and daily dose (mg/kg/day) (AOR = 2.892 95% CI 1.392 to 6.007)]. Smaller GA was found directly related to OOP. Among the other medical risk factors, phosphorus intake was significantly low in the high-ALP group. No, significant relationship between duration of PN and use of steroids and diuretics, and intake of vitamins and minerals were identified. CONCLUSION: The daily and cumulative doses of caffeine and smaller GA are associated with the development of OOP in this study cohort. Clinical randomized control studies are needed to validate the outcomes and determine the range of safest and most effective caffeine doses for treating AOP in preterm neonates.


Subject(s)
Bone Diseases, Metabolic , Rickets , Sleep Apnea Syndromes , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/drug therapy , Caffeine/adverse effects , Cohort Studies , Female , Humans , Infant, Newborn , Infant, Premature , Infant, Very Low Birth Weight , Male , Retrospective Studies , Vitamins
10.
Int J Environ Health Res ; 32(7): 1489-1499, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33660557

ABSTRACT

To investigate the potential association between BMP2 single nucleotide polymorphisms (SNPs) and brick-tea-type skeletal fluorosis risk in cross-sectional case-control study conducted in Sinkiang and Qinghai, China, a total of 598 individuals, including 308 Tibetans and 290 Kazakhs, were enrolled. Using the standard WS/192-2008 (China), 221 skeletal fluorosis cases were diagnosed, including 123 Tibetans and 98 Kazakhs. Logistic regressions 2 analysis did not find the association between SNPs (Rs235764, Rs235739 and Rs996544) and skeletal fluorosis. Genetic models, linkage disequilibrium (LD) and haplotype analysis were not found to be associated with risk of skeletal fluorosis after adjustment by age and sex (P>0.05).Our data suggested that Rs 235764, Rs 235739 and Rs 996544 were not linked susceptibility for skeletal fluorosis in our cross-sectional case-control study.


Subject(s)
Bone Diseases, Metabolic , Bone Morphogenetic Protein 2/genetics , Tea/chemistry , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/genetics , Case-Control Studies , China/epidemiology , Cross-Sectional Studies , Fluorides/analysis , Fluorides/toxicity , Humans , Polymorphism, Single Nucleotide , Tibet/epidemiology
11.
J Evid Based Dent Pract ; 22(3): 101721, 2022 09.
Article in English | MEDLINE | ID: mdl-36162899

ABSTRACT

AIM: This systematic review and meta-analysis sought to determine if there was a significantly enhanced risk of peri­implant marginal bone loss (MBL) due to the increased number of cigarettes smoked per day. MATERIALS AND METHODS: Six databases, including Medline, Embase, Cochrane Library, Web of Science, Scopus, and ProQuest, were searched until February, 2021. The search terms "dental implant, oral implant, smoking, smoker, non-smoker, marginal bone loss and crestal bone loss" were used in combination to seek the articles providing data for MBL related to the smoking habit. Articles were excluded if the quantity of cigarettes smoked per day was not reported. Random-effects meta-analyses were used to pool the estimates of mean difference (MD) with 95% confidence intervals (CI). RESULTS: Eight studies were included for qualitative and 5 for quantitative synthesis. The meta-analyses revealed higher levels of MBL in patients who smoked <10 or >10 cigarettes/day than in non-smokers (<10: (MD -0.33, 95% CI -0.69-0.03 and >10: MD -0.58, 95% CI -0.96- -0.19). There was a significant risk of MBL between patients who smoked >10 and <10 cigarettes/day (MD -0.23, 95% CI -0.47-0.01). CONCLUSION: It seems the risk of MBL is steadily increasing as daily smoking increases.


Subject(s)
Alveolar Bone Loss , Bone Diseases, Metabolic , Dental Implants , Alveolar Bone Loss/chemically induced , Bone Diseases, Metabolic/chemically induced , Dental Implantation, Endosseous , Dental Implants/adverse effects , Humans , Smokers , Smoking/adverse effects , Tobacco Smoking
12.
FASEB J ; 34(4): 5348-5362, 2020 04.
Article in English | MEDLINE | ID: mdl-32072664

ABSTRACT

Alcohol consumption is regarded as one of the leading risk factors for secondary osteopenia. Coupled angiogenesis and osteogenesis via distinct type-H vessels orchestrates subtle biological processes of bone homeostasis. The dysfunction of angiogenesis and osteogenesis contributes to decreased bone mass during the development of osteopenia. Herein, we identified microRNA-136-3p was remarkedly downregulated in the mouse model of alcohol-induced osteopenia. Following the alcohol administration, downregulated microRNA-136-3p significantly suppressed vascularization and osteogenic differentiation in human umbilical vein endothelial cells (HUVECs) and bone mesenchymal stem cells (BMSCs), respectively. Furthermore, microRNA-136-3p could target phosphatase and tensin homolog deleted on chromosome ten (PTEN) in both HUVECs and BMSCs, thus substantially modulating the capacity of vessel formation and osteogenic differentiation. In the mouse model, microRNA-136-3p Agomir ameliorated alcohol-induced osteopenia, with the concomitant restoration of bone mass and type-H vessel formation. For the first time, this study demonstrated the pivotal role of microRNA-136-3p/PTEN axis in regulations of vascularization and bone formation, which might become the potential therapeutic target of alcohol-induced bone loss.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Ethanol/toxicity , Gene Expression Regulation , MicroRNAs/genetics , Neovascularization, Pathologic/prevention & control , Osteogenesis , PTEN Phosphohydrolase/metabolism , Animals , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Cell Differentiation , Central Nervous System Depressants/toxicity , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/genetics
13.
J Pediatr Hematol Oncol ; 43(5): 172-175, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32815875

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate levels of vitamin D, bone mineral density (BMD), and radiograph features at diagnosis and after 6 months of chemotherapy in patients with acute lymphoblastic leukemia (ALL). Vitamin D levels were also correlated with BMD and radiograph features. MATERIALS AND METHODS: 25-Hydroxy vitamin D [25(OH)D] levels, BMD, and radiograph features were assessed in 50 newly diagnosed patients of ALL in the age group of 2 to 14 years. A total of 30 age-matched and sex-matched children were recruited as controls. Vitamin D deficiency was defined as 25(OH)D <10 ng/mL, Vitamin D insufficiency as 10 to 29 ng/mL, and Vitamin D sufficiency as ≥30 ng/mL. Enzyme immunoassay (EIA) was used for the quantitative measurement of 25(OH)D levels in plasma and a LUNAR DPX NT bone densitometer was used for the assessment of BMD. RESULTS: The mean age of the patients was 6.3 years, with a male:female ratio of 1.38:1. The mean 25(OH)D levels were 31.90±16.90 ng/mL in patients at diagnosis against 41.63±20.50 ng/mL in controls (P=0.02). Levels were 18.50±11.10 ng/mL postchemotherapy (P=0.00). Female sex was a risk factor for deficient 25(OH)D levels. There was a significant decrease in BMD postchemotherapy in the age groups of 5 to 10 and above 10 years at the femoral neck. Osteopenic changes were observed in more number of patients after 6 months of chemotherapy. There was a significant correlation between vitamin D levels, BMD, and osteopenic changes. CONCLUSIONS: Vitamin D deficiency was common among ALL patients, which worsened after chemotherapy. This had a significant correlation with BMD and osteopenic changes in radiograph.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Density/drug effects , Bone and Bones/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Agents/adverse effects , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/metabolism , Bone and Bones/drug effects , Child , Female , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D/metabolism , Vitamin D Deficiency/blood , Vitamin D Deficiency/metabolism
14.
Clin Orthop Relat Res ; 479(11): 2493-2501, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34077400

ABSTRACT

BACKGROUND: Neoadjuvant chemotherapy in patients with primary osteosarcoma improves survival rates, but it also causes side effects in various organs including bone. Low bone mineral density (BMD) can occur owing partly to chemotherapy or limited mobility. This can cause a higher risk of fractures compared with those who do not receive such treatment. Changes in BMD alone cannot explain the propensity of fractures. Studying microarchitectural changes of bone might help to understand the effect. QUESTIONS/PURPOSES: (1) Do patients who were treated for osteosarcoma (more than 20 years previously) have low BMD? (2) Do these patients experience more fractures than controls who do not have osteosarcoma? (3) What differences in bone microarchitecture are present between patients treated for high-grade osteosarcoma and individuals who have never had osteosarcoma? METHODS: We contacted 48 patients who were treated for osteosarcoma and who participated in an earlier study. These patients underwent multimodal treatment including chemotherapy more than 20 years ago. Of the original patient group, 60% (29 of 48) were missing, leaving 40% (19 of 48) available for inclusion in this study; all 19 agreed to participate. There were nine men and 10 women with a mean age of 46 ± 4 years and a mean time from surgery to examination of 28 ± 3 years. BMD was measured by dual-energy x-ray absorptiometry, and any fracture history was assessed using a questionnaire. Additionally, high-resolution peripheral quantitative CT was performed to compare the groups in terms of microarchitectural changes, such as cortical and trabecular area, cortical and trabecular thickness, cortical porosity, and endocortical perimeter. Participants in the control group were selected from a cohort consisting of a population-based random sample of 499 healthy adult women and men. Osteoporosis or low BMD was not an exclusion criterion for entering this study; however, the patients in the control group were selected based on a normal BMD (that is, T score > -1.0 at both the spine and hip). Also, the participants were matched based on age and sex. Differences between patients and controls were assessed using the Wilcoxon rank sum test for continuous variables and a chi-square test for categorical variables. A multiple regression analysis was performed. Model assumptions were checked using histograms and quantile-quantile plots of residuals. RESULTS: Twelve of 19 patients who were treated for osteosarcoma had either osteopenia (eight patients) or osteoporosis (four patients). More patients with osteosarcoma reported sustaining fractures (11 of 19 patients) than did control patients (2 of 19 controls; p < 0.001). Among all microarchitectural parameters, only the endocortical perimeter was increased in patients compared with the control group (75 ± 15 mm versus 62 ± 18 mm; p = 0.04); we found no differences between the groups in terms of cortical and trabecular area, cortical and trabecular thickness, or cortical porosity. CONCLUSION: Although patients who were treated for osteosarcoma had osteopenic or osteoporotic BMD and a higher proportion of patients experienced fractures than did patients in the control group, we could not confirm differences in microarchitectural parameters using high-resolution peripheral quantitative CT. Therefore, it seems that bone geometry and microstructural parameters are not likely the cause of the increased proportion of fractures observed in our patients who were treated for osteosarcoma. Until we learn more about the bone changes associated with chemotherapy in patients with osteosarcoma, we recommend that patients undergo regular BMD testing, and we recommend that physicians consider osteoporosis treatment in patients with low BMD. These data might provide the impetus for future multicenter prospective studies examining the association between chemotherapy and bone microarchitecture. LEVEL OF EVIDENCE: Level III, therapeutic study.


Subject(s)
Bone Diseases, Metabolic/chemically induced , Fractures, Bone/chemically induced , Neoadjuvant Therapy/adverse effects , Osteoporosis/chemically induced , Osteosarcoma/therapy , Absorptiometry, Photon , Adult , Bone Density , Cancellous Bone/diagnostic imaging , Cancellous Bone/physiopathology , Cancellous Bone/ultrastructure , Combined Modality Therapy , Cortical Bone/diagnostic imaging , Cortical Bone/physiopathology , Cortical Bone/ultrastructure , Female , Follow-Up Studies , Humans , Male , Middle Aged , Osteosarcoma/physiopathology , Tomography, X-Ray Computed
15.
Biochem Biophys Res Commun ; 527(1): 270-275, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446379

ABSTRACT

Aldehyde dehydrogenase 2 (ALDH2) plays major roles in aldehyde detoxification and in the catalysis of amino acids. ALDH2∗2, a dominant-negative transgenic expressing aldehyde dehydrogenase 2 (ALDH2) protein, is produced by a single nucleotide polymorphism (rs671) and is involved in the development of osteoporosis and hip fracture with aging. In a previous study, transgenic mice expressing Aldh2∗2(Aldh2∗2 Tg) osteoblastic cells or acetaldehyde -treated MC3T3-E1 showed impaired osteoblastogenesis and caused osteoporosis [1]. In this study, we demonstrated the effects of astaxanthin for differentiation to osteoblasts of MC3T3-E1 by the addition of acetaldehyde and Aldh2∗2 Tg mesenchymal stem cells in bone marrow. Astaxanthin restores the inhibited osteoblastogenesis by acetaldehyde in MC 3T3-E1 and in bone marrow mesenchymal stem cells of Aldh2∗2 Tg mice. Additionally, astaxanthin administration improved femur bone density in Aldh2∗2 Tg mice. Furthermore, astaxanthin improved cell survival and mitochondrial function in acetaldehyde-treated MC 3T3-E1 cells. Our results suggested that astaxanthin had restorative effects on osteoblast formation and provide new insight into the regulation of osteoporosis and suggest a novel strategy to promote bone formation in osteopenic diseases caused by impaired acetaldehyde metabolism.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/metabolism , Bone Diseases, Metabolic/drug therapy , Osteoclasts/drug effects , 3T3 Cells , Acetaldehyde/antagonists & inhibitors , Acetaldehyde/pharmacology , Administration, Oral , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mutation , Osteoclasts/metabolism , Osteogenesis/drug effects , Oxidative Stress/drug effects , Xanthophylls/administration & dosage , Xanthophylls/pharmacology
16.
Breast Cancer Res Treat ; 180(3): 675-685, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32124136

ABSTRACT

PURPOSE: The phase III DATA study compared 6 and 3 years of adjuvant anastrozole following 2-3 years of tamoxifen in postmenopausal breast cancer patients. This pre-planned side-study assessed the relationship between a reduced bone mineral density (BMD) and distant recurrence-free survival (DRFS), and evaluated the effect of bisphosphonates on DRFS. METHODS: We selected all patients with a BMD measurement within 3 years after randomisation (landmark) without any DRFS events. Kaplan-Meier methods and Cox proportional hazards models were used for analyses. RESULTS: Of 1860 eligible patients, 1142 had a DEXA scan before the landmark. The BMD was normal in 436 (38.2%) and showed osteopenia in 565 (49.5%) and osteoporosis in 141 (12.3%) patients. After a median follow-up of 5.0 years from the landmark, neither osteopenia nor osteoporosis (compared with normal BMD) were associated with DRFS in both the 6-year [osteopenia HR 0.82 (95% CI 0.45-1.49), osteoporosis HR 1.10 (95% CI 0.26-4.67)] and the 3-year arm [osteopenia HR 0.75 (95% CI 0.40-1.42), osteoporosis HR 1.86 (95% CI 0.43-8.01)]. Moreover, bisphosphonate use did not impact DRFS. CONCLUSION: No association was observed between a reduced BMD and DRFS. Neither did we observe an impact of bisphosphonates on DRFS.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Density/drug effects , Bone Diseases, Metabolic/mortality , Breast Neoplasms/drug therapy , Diphosphonates/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Osteoporosis/mortality , Antineoplastic Agents, Hormonal/adverse effects , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/pathology , Breast Neoplasms/pathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/pathology , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Recurrence, Local/pathology , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Osteoporosis/pathology , Prognosis , Survival Rate , Tamoxifen/adverse effects
17.
Osteoporos Int ; 31(9): 1671-1682, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32361950

ABSTRACT

The relationship between heavy metal exposure and risk of osteopenia or osteoporosis has biological plausibility, yet it remains inconclusive; therefore, we conducted a systematic review and meta-analysis to evaluate the associations between exposure to heavy metals (i.e., cadmium, lead, and mercury) and the risk of osteopenia or osteoporosis. Databases of MEDLINE, Embase, Scopus, and Web of Science were searched through November 2019, to identify studies that evaluated the relationship between exposure to cadmium, lead, and mercury and risk of osteopenia or osteoporosis in adults. Fourteen eligible studies were included. Effect sizes expressed as pooled odds ratios (OR) and 95% confidence intervals (CI) were estimated using weighted random-effect models. Exposure to cadmium (OR = 1.35; 95% CI: 1.17 to 1.56; P ≤ 0.001) and lead (OR = 1.15; 95% CI: 1.00 to 1.32; P = 0.05) was associated with an increased risk of osteopenia or osteoporosis, unlike mercury. Subgroup analyses showed cadmium exposure increased the risk of osteopenia or osteoporosis in older (> 65 yrs.; OR = 1.43; 95%CI: 1.08 to 1.88, P = 0.01) compared with younger (18-65 yrs.; OR = 1.24; 95% CI: 1.02 to 1.52, P = 0.03) adults. Also, lead exposure increased the risk in men (OR = 1.55; 95% CI: 1.15 to 2.09, P = 0.007) unlike in women. By contrast to urinary levels, blood (OR = 1.26; 95% CI: 1.08 to 1.47, P = 0.003) and dietary (OR = 1.46; 95% CI: 1.28 to 1.67, P < 0.001) levels of cadmium were associated with an increased risk of osteopenia or osteoporosis. Exposure to cadmium and lead may be associated with an increased risk of osteopenia or osteoporosis, although high heterogeneity was detected.


Subject(s)
Bone Diseases, Metabolic , Mercury , Metals, Heavy , Osteoporosis , Adult , Aged , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/epidemiology , Cadmium/toxicity , Female , Humans , Male , Mercury/toxicity , Metals, Heavy/toxicity , Osteoporosis/chemically induced , Osteoporosis/epidemiology
18.
FASEB J ; 33(11): 12972-12982, 2019 11.
Article in English | MEDLINE | ID: mdl-31500447

ABSTRACT

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.


Subject(s)
Bone Diseases, Metabolic/chemically induced , CD18 Antigens/metabolism , Integrin alpha4/metabolism , Maternal Exposure , Nicotine/administration & dosage , Peptidyl-Dipeptidase A/metabolism , Receptors, Nicotinic/metabolism , Animals , Female , Male , Pregnancy , Rats , Rats, Wistar
19.
Skeletal Radiol ; 49(3): 345-357, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31501957

ABSTRACT

Skeletal fluorosis is a rare toxic osteopathy characterized by massive bone fixation of fluoride. The disease occurs as an endemic problem in some parts of the world and is the result of prolonged ingestion or rarely by inhalation of high amounts of fluoride. Radiographic presentation is mainly characterized by bone changes with osteocondensation and later ossification of many ligaments and interosseous membranes. Skeletal fluorosis is not clinically obvious and can be confused with other rheumatologic disorders. Its severity lies in the development of skeletal deformities and neurological complications. Management of fluorosis generally focuses on symptom treatment.


Subject(s)
Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/diagnostic imaging , Fluoride Poisoning/diagnostic imaging , Bone Diseases, Metabolic/epidemiology , Fluoride Poisoning/epidemiology , Humans , Ossification, Heterotopic/chemically induced , Ossification, Heterotopic/diagnostic imaging , Ossification, Heterotopic/epidemiology , Osteosclerosis/chemically induced , Osteosclerosis/diagnostic imaging , Osteosclerosis/epidemiology
20.
Int J Mol Sci ; 21(7)2020 Apr 04.
Article in English | MEDLINE | ID: mdl-32260461

ABSTRACT

Growing evidence suggests an adverse impact of gut microbiota dysbiosis on human health. However, it remains unclear whether embryonic osteogenesis is affected by maternal gut dysbacteriosis. In this study, we observed that elevated lipopolysaccharide (LPS) levels led to skeletal developmental retardation in an established mouse model of gut microbiota dysbiosis. Using chick embryos exposed to dysbacteriosis-derived LPS, we found restriction in the development of long bones as demonstrated by Alcian blue and alizarin red staining. Micro-CT and histological analysis exhibited decreased trabecular volume, bone mineral density, and collagen production, as well as suppressed osteoblastic gene expression (Ocn, Runx2, Osx, and Dlx5) in chick embryonic phalanges following LPS treatment. Atomic force microscopy manifested decreased roughness of MC3T3-E1 cells and poorly developed matrix vesicles (MVs) in presence of LPS. The expression of the aforementioned osteoblastic genes was suppressed in MC3T3-E1 cells as well. High-throughput RNA sequencing indicated that retinoic acid (RA) may play an important role in LPS-induced osteopenia. The addition of RA suppressed Dlx5 expression in MC3T3-E1 cells, as was also seen when exposed to LPS. Quantitative PCR, Western blot, and immunofluorescent staining showed that retinoic acid receptor α (RARα) was upregulated by LPS or RA treatment, while the expression of DLX5 was downregulated. CYP1B1 expression was increased by LPS treatment in MC3T3-E1 cells, which might be attributed to the increased inflammatory factors and subsequently activated NF-κB signaling. Eventually, blocking RA signals with AGN193109 successfully restored LPS-inhibited osteoblastic gene expression. Taken together, our data reveals that maternal gut microbiota dysbiosis can interfere with bone ossification, in which Dlx5 expression regulated by RA signaling plays an important role.


Subject(s)
Bone Diseases, Metabolic/genetics , Dysbiosis/genetics , Homeodomain Proteins/genetics , Lipopolysaccharides/adverse effects , Tretinoin/metabolism , Animals , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/metabolism , Cell Line , Chick Embryo , Disease Models, Animal , Dysbiosis/chemically induced , Dysbiosis/metabolism , Ectoderm/metabolism , Female , Homeodomain Proteins/metabolism , Mice , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL