Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
1.
JAMA Intern Med ; 184(3): 242-251, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38252426

RESUMO

Importance: Use of buprenorphine or methadone to treat opioid use disorder is recommended in pregnancy; however, their teratogenic potential is largely unknown. Objective: To compare the risk of congenital malformations following in utero exposure to buprenorphine vs methadone. Design, Setting, and Participants: This population-based cohort study used health care utilization data from publicly insured Medicaid beneficiaries in the US from 2000 to 2018. A total of 13 360 pregnancies with enrollment from 90 days prior to pregnancy start through 1 month after delivery and first trimester use of buprenorphine or methadone were included and linked to infants. Data were analyzed from July to December 2022. Exposure: A pharmacy dispensing of buprenorphine or a code for administration of methadone in the first trimester. Main Outcomes and Measures: Primary outcomes included major malformations overall and malformations previously associated with opioids (any cardiac malformations, ventricular septal defect, secundum atrial septal defect/nonprematurity-related patent foramen ovale, neural tube defects, clubfoot, and oral clefts). Secondary outcomes included other organ system-specific malformations. Risk differences and risk ratios (RRs) were estimated comparing buprenorphine with methadone, adjusting for confounders with propensity score overlap weights. Results: The cohort included 9514 pregnancies with first-trimester buprenorphine exposure (mean [SD] maternal age, 28.4 [4.6] years) and 3846 with methadone exposure (mean [SD] maternal age, 28.8 [4.7] years). The risk of malformations overall was 50.9 (95% CI, 46.5-55.3) per 1000 pregnancies for buprenorphine and 60.6 (95% CI, 53.0-68.1) per 1000 pregnancies for methadone. After confounding adjustment, buprenorphine was associated with a lower risk of malformations compared with methadone (RR, 0.82; 95% CI, 0.69-0.97). Risk was lower with buprenorphine for cardiac malformations (RR, 0.63; 95% CI, 0.47-0.85), including both ventricular septal defect (RR, 0.62; 95% CI, 0.39-0.98) and secundum atrial septal defect/nonprematurity-related patent foramen ovale (RR, 0.54; 95% CI, 0.30-0.97), oral clefts (RR, 0.65; 95% CI, 0.35-1.19), and clubfoot (RR, 0.55; 95% CI, 0.32-0.94). Results for neural tube defects were uncertain given low event counts. In secondary analyses, buprenorphine was associated with a decreased risk of central nervous system, urinary, and limb malformations but a greater risk of gastrointestinal malformations compared with methadone. These findings were consistent in sensitivity and bias analyses. Conclusions and Relevance: In this cohort study, the risk of most malformations previously associated with opioid exposure was lower in buprenorphine-exposed infants compared with methadone-exposed infants, independent of measured confounders. Malformation risk is one factor that informs the individualized patient decision regarding medications for opioid use disorder in pregnancy.


Assuntos
Buprenorfina , Pé Torto Equinovaro , Forame Oval Patente , Cardiopatias Congênitas , Comunicação Interventricular , Defeitos do Tubo Neural , Transtornos Relacionados ao Uso de Opioides , Complicações na Gravidez , Gravidez , Lactente , Feminino , Humanos , Adulto , Metadona/efeitos adversos , Buprenorfina/efeitos adversos , Primeiro Trimestre da Gravidez , Estudos de Coortes , Pé Torto Equinovaro/complicações , Pé Torto Equinovaro/tratamento farmacológico , Forame Oval Patente/complicações , Forame Oval Patente/tratamento farmacológico , Complicações na Gravidez/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/epidemiologia , Cardiopatias Congênitas/complicações , Defeitos do Tubo Neural/complicações , Defeitos do Tubo Neural/tratamento farmacológico , Comunicação Interventricular/complicações , Comunicação Interventricular/tratamento farmacológico
2.
Mymensingh Med J ; 32(4): 1123-1132, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37777911

RESUMO

Etiology of congenital heart defects are complex and possibly lie within the interaction of environmental exposures and inherited factors. Exploration of the contribution of environmental risk factors that are potentially modifiable impeded the prevention of CHDs. This study was conducted to evaluate the environmental risk factors of CHD. It was a case control study, conducted from July 2018 to June 2019 in Paediatric Cardiology department of Dhaka Shishu (Children) Hospital, Bangladesh. Parents of the children with CHDs visiting the out-patient department were considered as case. Control was taken from parents of the children not having congenital heart disease. Data were collected by face-to-face interview using a structured questionnaire containing all the variables of interest and analyzed by using SPSS version 21.0. Majority of the respondents were from rural area (86.9% and 80.0% in case and control group respectively) and CHD was found significantly higher in rural population (p<0.05). Consanguinity was present in 8.9% in case group and CHD was found significantly higher among children born to consanguineous parents (p<0.05). Most of the mother (65.4%) had completed primary level of education however 11.9% mother was illiterate in case group. CHD was found significantly higher among illiterate mothers (p<0.05). Most of the respondents belonged to lower and lower-middle class family (83.1% and 75.7% in case and control group respectively) and CHD was found significantly higher among them (p<0.05). Mothers exposed to passive smoking and in stress during pregnancy period, CHD was significantly higher (p<0.05). No significant association was found between maternal drug use and infection during pregnancy period with CHD (p<0.05). Maternal illiteracy, residing in rural areas, low and lower- middle class socioeconomic status, consanguineous marriage, exposed to passive smoking and stress during pregnancy period have been significantly associated with CHDs.


Assuntos
Cardiopatias Congênitas , Poluição por Fumaça de Tabaco , Feminino , Gravidez , Humanos , Criança , Estudos de Casos e Controles , Poluição por Fumaça de Tabaco/efeitos adversos , Bangladesh/epidemiologia , Cardiopatias Congênitas/etiologia , Cardiopatias Congênitas/induzido quimicamente , Fatores de Risco
3.
Artigo em Inglês | MEDLINE | ID: mdl-36740268

RESUMO

BACKGROUND: Few prospective studies have investigated the association between paternal occupational exposures and risk of infant congenital heart defects (CHDs). We investigated the associations between paternal occupational exposures, frequency of use, and concurrent or sequential exposure to a mixture of compounds and the risk of infant CHDs. METHODS: Our study examined 28,866 participants in the Japan Environment and Children's Study. Logistic regression analysis was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) associated with paternal occupational exposures during the 3 months until pregnancy was noticed after adjustment for potential confounding factors of the infant CHDs. CHD diagnosis was ascertained from medical record. RESULTS: In total, 175 were diagnosed with infant CHDs. The number of fathers who were exposed to the following substances at least once a month were: 11,533 for photo copying machine/laser printer, 10,326 for permanent marker, 8,226 for soluble paint/inkjet printer, 6,188 for kerosene/petroleum/benzene/gasoline, 4,173 for organic solvents, 3,433 for chlorine bleach/germicide, 2,962 for engine oil, 2,931 for insecticide, 2,460 for medical sterilizing disinfectant, 1,786 for welding fumes, 1,614 for dyestuffs, 1,247 for any products containing lead-like solder, 986 for herbicide, 919 for radiation/radioactive substances/isotopes, 837 for lead-free solder, 341 for microbes, 319 for formalin/formaldehyde, 301 for agricultural chemical not listed above or unidentified, 196 for general anesthetic for surgery at hospital, 171 for anti-cancer drug, 147 for chromium/arsenic/cadmium, 88 for mercury and 833 for other chemical substances. Paternal occupational exposure regularly to photo copying machine or laser printer and soluble paint/inkjet printer were associated with higher risks of infant CHDs: the adjusted ORs (95%CIs) were 1.38 (1.00-1.91) and 1.60 (1.08-2.37), respectively. The higher risks were also observed for occasional exposure to engine oil, any products containing lead-like solder lead-free solder, and microbes; the adjusted ORs (95%CIs) were 1.68 (1.02-2.77), 2.03 (1.06-3.88), 3.45 (1.85-6.43), and 4.51, (1.63-12.49), respectively. CONCLUSIONS: Periconceptional paternal occupational exposure was associated with a higher risk of infant CHDs. Further studies using biomarkers of the association between paternal occupational exposure and infant CHDs are warranted.


Assuntos
Cardiopatias Congênitas , Exposição Ocupacional , Masculino , Humanos , Lactente , Criança , Japão/epidemiologia , Estudos Prospectivos , Fatores de Risco , Estudos de Casos e Controles , Exposição Ocupacional/efeitos adversos , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/epidemiologia , Pai
4.
JAMA Psychiatry ; 80(2): 156-166, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36477338

RESUMO

Importance: Psychiatric disorders are common among female individuals of reproductive age. While antipsychotic medication use is increasing, the safety of such medications in pregnancy is an area with large evidence gaps. Objective: To evaluate the risk of first-trimester antipsychotic exposure with respect to congenital malformations, focusing on individual drugs and specific malformation subtypes. Design, Setting, and Participants: This cohort study used data from nationwide health registers from the 5 Nordic countries and the US and spanned 1996 to 2018. The Nordic cohort included all pregnancies resulting in singleton live-born infants, and the US cohort consisted of publicly insured mothers linked to their live-born infants nested in the nationwide Medicaid Analytic eXtract. Data were analyzed from November 2020 to April 2022. Exposures: One or more first-trimester dispensing of any atypical, any typical, and individual antipsychotic drugs. Main Outcomes and Measures: Any major congenital malformation and specific malformation subtypes previously suggested to be associated with antipsychotic exposure in utero: cardiovascular malformations, oral clefts, neural tube defects, hip dysplasia, limb reduction defects, anorectal atresia/stenosis, gastroschisis, hydrocephalus, other specific brain anomalies, and esophageal disorders. Propensity score stratification was used to control for potential confounders. Pooled adjusted estimates were calculated using indirect standardization. Results: A total of 6 455 324 unexposed mothers (mean maternal age range across countries: 24-31 years), 21 751 mothers exposed to atypical antipsychotic drugs (mean age range, 26-31 years), and 6371 mothers exposed to typical antipsychotic drugs (mean age range, 27-32 years) were included in the study cohort. Prevalence of any major malformation was 2.7% (95% CI, 2.7%-2.8%) in unexposed infants, 4.3% (95% CI, 4.1%-4.6%) in infants with atypical antipsychotic drug exposure, and 3.1% (95% CI, 2.7%-3.5%) in infants with typical antipsychotic drug exposure in utero. Among the most prevalent exposure-outcome combinations, adjusted relative risks (aRR) were generally close to the null. One exception was olanzapine exposure and oral cleft (aRR, 2.1 [95% CI, 1.1-4.3]); however, estimates varied across sensitivity analyses. Among moderately prevalent combinations, increased risks were observed for gastroschisis and other specific brain anomalies after atypical antipsychotic exposure (aRR, 1.5 [95% CI, 0.8-2.6] and 1.9 [95% CI, 1.1-3.0]) and for cardiac malformations after chlorprothixene exposure (aRR, 1.6 [95% CI, 1.0-2.7]). While the association direction was consistent across sensitivity analyses, confidence intervals were wide, prohibiting firm conclusions. Conclusions and Relevance: In this study, considering the evidence from primary and sensitivity analyses and inevitable statistical noise for very rare exposure-outcome combinations, in utero antipsychotic exposure generally was not meaningfully associated with an increased risk of malformations. The observed increased risks of oral clefts associated with olanzapine, gastroschisis, and other specific brain anomalies with atypical antipsychotics and cardiac malformations with chlorprothixene requires confirmation as evidence continues to accumulate.


Assuntos
Anormalidades Induzidas por Medicamentos , Antipsicóticos , Gastrosquise , Cardiopatias Congênitas , Gravidez , Lactente , Feminino , Humanos , Adulto Jovem , Adulto , Antipsicóticos/efeitos adversos , Estudos de Coortes , Olanzapina , Clorprotixeno , Gastrosquise/complicações , Anormalidades Induzidas por Medicamentos/epidemiologia , Anormalidades Induzidas por Medicamentos/etiologia , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/epidemiologia , Países Escandinavos e Nórdicos/epidemiologia
5.
Cells ; 11(24)2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36552710

RESUMO

Folic acid (FA) is a synthetic and highly stable version of folate, while 6S-5-methyltetrahydrofolate is the predominant form of dietary folate in circulation and is used as a crystalline form of calcium salt (MTHF-Ca). The current study aims to evaluate the toxicity and safety of FA and MTHF-Ca on embryonic development, with a focus on cardiovascular defects. We began to analyze the toxicity of FA and MTHF-Ca in zebrafish from four to seventy-two hours postfertilization and assessed the efficacy of FA and MTHF-Ca in a zebrafish angiogenesis model. We then analyzed the differently expressed genes in in vitro fertilized murine blastocysts cultured with FA and MTHF-Ca. By using gene-expression profiling, we identified a novel gene in mice that encodes an essential eukaryotic translation initiation factor (Eif1ad7). We further applied the morpholino-mediated gene-knockdown approach to explore whether the FA inhibition of this gene (eif1axb in zebrafish) caused cardiac development disorders, which we confirmed with qRT-PCR. We found that FA, but not MTHF-Ca, could inhibit angiogenesis in zebrafish and result in abnormal cardiovascular development, leading to embryonic death owing to the downregulation of eif1axb. MTHF-Ca, however, had no such cardiotoxicity, unlike FA. The current study thereby provides experimental evidence that FA, rather than MTHF-Ca, has cardiovascular toxicity in early embryonic development and suggests that excessive supplementation of FA in perinatal women may be related to the potential risk of cardiovascular disorders, such as congenital heart disease.


Assuntos
Ácido Fólico , Cardiopatias Congênitas , Animais , Feminino , Camundongos , Gravidez , Cálcio , Desenvolvimento Embrionário/efeitos dos fármacos , Ácido Fólico/efeitos adversos , Coração , Peixe-Zebra/genética , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/etiologia
6.
PLoS One ; 17(11): e0277611, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36395323

RESUMO

BACKGROUND: Children with congenital heart defects have an increased risk of neurodevelopmental disability. The impact of environmental chemical exposures during daily life on neurodevelopmental outcomes in toddlers with congenital heart defects is unknown. METHODS: This prospective study investigated the impacts of early childhood exposure to mixtures of environmental chemicals on neurodevelopmental outcomes after cardiac surgery. Outcomes were assessed at 18 months of age using The Bayley Scales of Infant and Toddler Development-III. Urinary concentrations of exposure biomarkers of pesticides, phenols, parabens, and phthalates, and blood levels of lead, mercury, and nicotine were measured at the same time point. Bayesian profile regression and weighted quantile sum regression were utilized to assess associations between mixtures of biomarkers and neurodevelopmental scores. RESULTS: One-hundred and forty infants were enrolled, and 110 (79%) returned at 18 months of age. Six biomarker exposure clusters were identified from the Bayesian profile regression analysis; and the pattern was driven by 15 of the 30 biomarkers, most notably 13 phthalate biomarkers. Children in the highest exposure cluster had significantly lower adjusted language scores by -9.41 points (95%CI: -17.2, -1.7) and adjusted motor scores by -4.9 points (-9.5, -0.4) compared to the lowest exposure. Weighted quantile sum regression modeling for the overall exposure-response relationship showed a significantly lower adjusted motor score (ß = -2.8 points [2.5th and 97.5th percentile: -6.0, -0.6]). The weighted quantile sum regression index weights for several phthalates, one paraben, and one phenol suggest their relevance for poorer neurodevelopmental outcomes. CONCLUSIONS: Like other children, infants with congenital heart defects are exposed to complex mixtures of environmental chemicals in daily life. Higher exposure biomarker concentrations were associated with significantly worse performance for language and motor skills in this population.


Assuntos
Cardiopatias Congênitas , Lactente , Humanos , Pré-Escolar , Estudos Prospectivos , Teorema de Bayes , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/cirurgia , Parabenos , Fenóis , Biomarcadores
7.
Medicine (Baltimore) ; 101(41): e30145, 2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36254077

RESUMO

The vancomycin dosing range for safe and effective treatment remains uncertain for children who had corrective surgery for a congenital heart disease (CHD). We aimed to determine the vancomycin dosing requirements for this subgroup of patients. This prospective cohort study included children younger than 14 years old with CHD who received intravenous vancomycin for at least 3 days at the Pediatric Cardiology section of King Abdulaziz Medical City, Riyadh. In total, 140 pediatric patients with CHD were included with a median age of 0.57 years (interquartile range 0.21-2.2). The mean vancomycin total daily dose (TDD), 37.71 ±â€…6.8 mg/kg/day, was required to achieve a therapeutic trough concentration of 7-20 mg/L. The patient's age group and the care setting were significant predictors of the vancomycin dosing needs. Neonates required significantly lower doses of 34 ±â€…6.03 mg/kg/day (P = .002), and young children higher doses of 43.97 ±â€…9.4 mg/kg/day (P = .003). The dosage requirements were independent of the type of cardiac lesion, cardiopulmonary surgery exposure, sex, and BMI percentile. However, the patients in the pediatric cardiac ward required higher doses of vancomycin 41.08 ±â€…7.06 mg/kg/day (P = .039). After the treatment, 11 (8.5%) patients had an elevated Scr, and 3 (2.3%) patients developed AKI; however, none of the patients' sociodemographic factors or clinical variables, or vancomycin therapy characteristics was significantly associated with the renal dysfunction. Overall, the vancomycin TDD requirements are lower in pediatric post-cardiac surgery compared to non-cardiac patients and are modulated by several factors.


Assuntos
Cardiopatias Congênitas , Vancomicina , Adolescente , Antibacterianos , Criança , Pré-Escolar , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/cirurgia , Humanos , Lactente , Recém-Nascido , Estudos Prospectivos , Estudos Retrospectivos
8.
Sci Rep ; 12(1): 14973, 2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056058

RESUMO

There is a gap in knowledge how maternal exposure to environmental tobacco smoke (ETS) is associated with offspring congenital heart defects (CHDs). In this case-control study, we collected data on 749 fetuses with CHDs and 880 fetuses without any congenital anomalies to examine the association of maternal ETS with fetal CHDs and the potentially moderating effect by maternal hazardous and noxious substances (HNS), periconceptional folate intake and paternal smoking. Maternal exposure to ETS in first trimester was associated with increased risk of CHDs in a dose-response gradient, with the AORs (95% CI) were1.38 (1.00-1.92), 1.60 (1.07-2.41), and 4.94 (2.43-10.05) for ETS < 1 h/day, 1-2 h/day, and ≥ 2 h/day, respectively. With the doubly unexposed group as reference categories, AORs for maternal ETS exposure ≥ 2 h/day in the absence of folate intake, in the presence of HNS exposure or paternal smoking, were 7.21, 11.43, and 8.83, respectively. Significant additive interaction between ETS exposure and maternal folate intake on CHDs was detected. Maternal ETS exposure during first trimester may increase the risk of offspring CHDs in a dose-response shape, and such effect may be modified by maternal folate intake or other potential factors.


Assuntos
Cardiopatias Congênitas , Poluição por Fumaça de Tabaco , Estudos de Casos e Controles , China/epidemiologia , Feminino , Ácido Fólico , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/etiologia , Humanos , Masculino , Exposição Materna/efeitos adversos , Fatores de Risco , Fumaça , Nicotiana , Poluição por Fumaça de Tabaco/efeitos adversos
9.
J Cell Mol Med ; 26(11): 3223-3234, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35521669

RESUMO

Maternal cigarette smoking is a risk factor for congenital heart defects (CHDs). Nicotine replacement therapies are often offered to pregnant women following failed attempts of smoking cessation. However, the impact of nicotine on embryonic heart development is not well understood. In the present study, the effects of maternal nicotine exposure (MNE) during pregnancy on foetal heart morphogenesis were studied. Adult female mice were treated with nicotine using subcutaneous osmotic pumps at 0.75 or 1.5 mg/kg/day and subsequently bred with male mice. Our results show that MNE dose-dependently increased CHDs in foetal mice. CHDs included atrial and ventricular septal defects, double outlet right ventricle, unguarded tricuspid orifice, hypoplastic left ventricle, thickened aortic and pulmonary valves, and ventricular hypertrophy. MNE also significantly reduced coronary artery size and vessel abundance in foetal hearts. Moreover, MNE resulted in higher levels of oxidative stress and altered the expression of key cardiogenic regulators in the developing heart. Nicotine exposure reduced epicardial-to-mesenchymal transition in foetal hearts. In conclusion, MNE induces CHDs and coronary artery malformation in mice. These findings provide insight into the adverse outcomes of foetuses by MNE during pregnancy.


Assuntos
Cardiopatias Congênitas , Efeitos Tardios da Exposição Pré-Natal , Abandono do Hábito de Fumar , Animais , Feminino , Cardiopatias Congênitas/induzido quimicamente , Humanos , Masculino , Camundongos , Nicotina/efeitos adversos , Gravidez , Dispositivos para o Abandono do Uso de Tabaco/efeitos adversos
10.
BMC Pregnancy Childbirth ; 22(1): 88, 2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35100977

RESUMO

BACKGROUND: MTHFD1 gene may affect the embryonic development by elevated homocysteine levels, DNA synthesis and DNA methylation, but limited number of genetic variants of MTHFD1 gene was focused on the association with congenital heart disease (CHD). This study examined the role of MTHFD1 gene and maternal smoking on infant CHD risk, and investigated their interaction effects in Chinese populations. METHODS: A case-control study of 464 mothers of CHD infants and 504 mothers of health controls was performed. The exposures of interest were maternal tobacco exposure, single nucleotide polymorphisms (SNPs) of maternal MTHFD1 gene. The logistic regression model was used for accessing the strength of association. RESULTS: Mothers exposed to secondhand smoke during 3 months before pregnancy (adjusted odds ratio [aOR] = 1.56; 95% confidence interval [CI]: 1.13-2.15) and in the first trimester of pregnancy (aOR = 2.24; 95%CI: 1.57-3.20) were observed an increased risk of CHD. Our study also found that polymorphisms of maternal MTHFD1 gene at rs1950902 (AA vs. GG: aOR = 1.73, 95% CI: 1.01-2.97), rs2236222 (GG vs. AA: aOR = 2.38, 95% CI: 1.38-4.12), rs1256142 (GA vs.GG: aOR = 1.57, 95% CI: 1.01-2.45) and rs11849530 (GG vs. AA: aOR = 1.68, 95% CI: 1.02-2.77) were significantly associated with higher risk of CHD. However, we did not observe a significant association between maternal MTHFD1 rs2236225 and offspring CHD risk. Furthermore, we found the different degrees of interaction effects between polymorphisms of the MTHFD1 gene including rs1950902, rs2236222, rs1256142, rs11849530 and rs2236225, and maternal tobacco exposure. CONCLUSIONS: Maternal polymorphisms of MTHFD1 gene, maternal tobacco exposure and their interactions are significantly associated with the risk of CHD in offspring in Han Chinese populations. However, more studies in different ethnic populations with a larger sample and prospective designs are required to confirm our findings. TRIAL REGISTRATION: Registration number: ChiCTR1800016635 .


Assuntos
Cardiopatias Congênitas/genética , Doenças do Recém-Nascido/genética , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Antígenos de Histocompatibilidade Menor/genética , Polimorfismo de Nucleotídeo Único , Adulto , Povo Asiático , Estudos de Casos e Controles , China/epidemiologia , Feminino , Cardiopatias Congênitas/induzido quimicamente , Humanos , Recém-Nascido , Doenças do Recém-Nascido/induzido quimicamente , Modelos Logísticos , Exposição Materna/efeitos adversos , Gravidez , Poluição por Fumaça de Tabaco/efeitos adversos , Fumar Tabaco/efeitos adversos
11.
Environ Sci Pollut Res Int ; 29(18): 26950-26959, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34865185

RESUMO

To investigate the correlation between maternal manganese and iron concentrations and the risk of CHD among their infant. A multi-center hospital-based case control study was conducted in China. There were 322 cases and 333 controls have been selected from pregnant women who received prenatal examinations. Correlations between CHDs and maternal manganese and iron concentrations were estimated by conditional logistic regression. Moreover, the interaction between manganese and iron on CHDs was analyzed. Compared with the controls, mothers whose hair manganese concentration was 3.01 µg/g or more were more likely to have a child with CHD than those with a lower concentration. The adjusted OR was 2.68 (95%CI = 1.44-4.99). The results suggested that mothers whose iron content was 52.95 µg/g or more had a significantly higher risk of having a child with CHD (aOR = 2.87, 95%CI = 1.54-5.37). No interaction between maternal manganese and iron concentrations was observed in the multiplicative or additive model. The concurrently existing high concentration of manganese and iron may bring higher risk of CHD (OR = 7.02). Women with excessive manganese concentrations have a significantly increased risk of having offspring with CHDs. The high maternal iron status also correlates with CHDs. The concurrently existing high concentration of manganese and iron may bring higher risk of CHD.


Assuntos
Cardiopatias Congênitas , Manganês , Estudos de Casos e Controles , Criança , China , Feminino , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/epidemiologia , Humanos , Ferro , Gravidez , Fatores de Risco
12.
Toxicology ; 462: 152965, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34597721

RESUMO

Benzo[a]pyrene (BaP), a prototypical polycyclic aromatic hydrocarbon, is widely present in the environment. BaP-induced heart defects have been frequently reported, but the underlying molecular mechanisms remain elusive. Here, we found that BaP increased heart malformations in zebrafish embryos in a concentration-dependent manner, which were attenuated by supplementation with either CH223191 (CH), an aryl hydrocarbon receptor (AHR) inhibitor, or N-acetyl-l-cysteine (NAC), a reactive oxygen species (ROS) scavenger. While CH and NAC both inhibited BaP-induced ROS generation, NAC had no effect on BaP-induced AHR activation. We further demonstrated that BaP increased mitochondrial ROS, decreased mitochondrial membrane potential, and caused endogenous apoptosis, with all these effects being counteracted by supplementation with either CH or NAC. Resveratrol (RSV), a natural AHR antagonist and ROS scavenger, also counteracted the heart malformations caused by BaP. Further experiments showed that RSV attenuated BaP-induced oxidative stress, mitochondrial damage and apoptosis, but had no significant effect on AHR activation. In conclusion, our findings show that BaP induces oxidative stress via AHR activation, which causes mitochondria-mediated intrinsic apoptosis, resulting in heart malformations in zebrafish embryos, and that RSV had a protective effect against BaP-induced heart defects mainly by inhibiting oxidative stress rather than through antagonism of AHR activity.


Assuntos
Benzo(a)pireno/toxicidade , Cardiopatias Congênitas/prevenção & controle , Receptores de Hidrocarboneto Arílico/metabolismo , Resveratrol/farmacologia , Acetilcisteína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Compostos Azo/farmacologia , Benzo(a)pireno/administração & dosagem , Relação Dose-Resposta a Droga , Cardiopatias Congênitas/induzido quimicamente , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Pirazóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Peixe-Zebra
13.
BJOG ; 128(12): 1949-1957, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34036715

RESUMO

OBJECTIVE: Studies restricted to live births may underestimate severe teratogenic effects. We address the limitation by including data from both prenatal and postnatal diagnoses of cardiac malformations. DESIGN: Register-based study. SETTING: Denmark. POPULATION: 364 012 singleton pregnancies from 2007 to 2014. METHODS: We used data from five nationwide registries. Exposure to antidepressants was measured using redeemed prescriptions. MAIN OUTCOME MEASURES: Pregnancies with cardiac malformations that end in miscarriage, termination, stillbirth, postnatal death or cardiac surgery <1 year of birth were classified as severe cardiac malformations (SCM). Propensity scores with adjusted prevalence ratios (PRs) were calculated. RESULTS: SCM were reported in 972 of 364 012 pregnancies overall and in 16 of 4105 exposed. For venlafaxine, the PR for SCM was 2.13 (95% confidence interval [CI] 0.89-5.13), 1.73 (95% CI 1.08-2.77) for other cardiac malformations, and there was a cluster of hypoplastic left heart syndromes (HLHS) (crude PR 17.4 [95% CI 6.41-47.2]), none of which ended in a live birth. For HLHS, the absolute risk increase was 4.4/1000 and the number needed to harm was 225. For selective serotonin reuptake inhibitors, the PRs were 1.09 (95% CI 0.52-2.30) and 1.38 (95% CI 1.00-1.92) for SCM and other cardiac malformations, respectively. CONCLUSIONS: Pregnancy exposure to venlafaxine is associated with an increased risk of severe cardiac malformations but with a low absolute risk. Potential mechanisms include direct effects or confounding by indication. Venlafaxine exposure is a marker for risk pregnancies for which fetal echocardiography may be considered. TWEETABLE ABSTRACT: Exposure to venlafaxine is associated with an increased risk of cardiac malformations but with a low absolute risk.


Assuntos
Anormalidades Induzidas por Medicamentos/epidemiologia , Antidepressivos/efeitos adversos , Cardiopatias Congênitas/induzido quimicamente , Complicações na Gravidez/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Adulto , Dinamarca/epidemiologia , Feminino , Cardiopatias Congênitas/epidemiologia , Humanos , Recém-Nascido , Masculino , Gravidez , Complicações na Gravidez/psicologia , Cuidado Pré-Natal/estatística & dados numéricos , Sistema de Registros , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Cloridrato de Venlafaxina/efeitos adversos
14.
Toxicol In Vitro ; 72: 105088, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33429043

RESUMO

The present study investigated the developmental toxicity of diethylstilbestrol (DES) in the zebrafish embryotoxicity test (ZET). This was done to investigate whether the ZET would better capture the developmental toxicity of DES than the embryonic stem cells test (EST) that was previously shown to underpredict the DES-induced developmental toxicity as compared to in vivo data, potentially because the EST does not capture late events in the developmental process. The ZET results showed DES-induced growth retardation, cumulative mortality and dysmorphisms (i.e. induction of pericardial edema) in zebrafish embryos while the endogenous ERα agonist 17ß-estradiol (E2) showed only growth retardation and cumulative mortality with lower potency compared to DES. Furthermore, the DES-induced pericardial edema formation in zebrafish embryos could be counteracted by co-exposure with ERα antagonist fulvestrant, indicating that the ZET captures the role of ERα in the mode of action underlying the developmental toxicity of DES. Altogether, it is concluded that the ZET differentiates DES from E2 with respect to their developmental toxicity effects, while confirming the role of ERα in mediating the developmental toxicity of DES. Furthermore, comparison to in vivo data revealed that, like the EST, in a quantitative way also the ZET did not capture the relatively high in vivo potency of DES as a developmental toxicant.


Assuntos
Carcinógenos/toxicidade , Dietilestilbestrol/toxicidade , Embrião não Mamífero/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Estradiol/toxicidade , Estrogênios/toxicidade , Teratogênicos/toxicidade , Peixe-Zebra/anormalidades , Animais , Embrião não Mamífero/anormalidades , Feminino , Cabeça/anormalidades , Cardiopatias Congênitas/induzido quimicamente , Masculino , Cauda/anormalidades , Cauda/efeitos dos fármacos , Testes de Toxicidade , Saco Vitelino/anormalidades , Saco Vitelino/efeitos dos fármacos
15.
Alcohol Clin Exp Res ; 45(1): 69-78, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33206417

RESUMO

BACKGROUND: Fetal alcohol spectrum disorder (FASD) is caused by prenatal alcohol exposure (PAE), the intake of ethanol (C2 H5 OH) during pregnancy. Features of FASD cover a range of structural and functional defects including congenital heart defects (CHDs). Folic acid and choline, contributors of methyl groups to one-carbon metabolism (OCM), prevent CHDs in humans. Using our avian model of FASD, we have previously reported that betaine, another methyl donor downstream of choline, prevents CHDs. The CHD preventions are substantial but incomplete. Ethanol causes oxidative stress as well as depleting methyl groups for OCM to support DNA methylation and other epigenetic alterations. To identify more compounds that can safely and effectively prevent CHDs and other effects of PAE, we tested glutathione (GSH), a compound that regulates OCM and is known as a "master antioxidant." METHODS/RESULTS: Quail embryos injected with a single dose of ethanol at gastrulation exhibited congenital defects including CHDs similar to those identified in FASD individuals. GSH injected simultaneously with ethanol not only prevented CHDs, but also improved survival and prevented other PAE-induced defects. Assays of hearts at 8 days (HH stage 34) of quail development, when the heart normally has developed 4-chambers, showed that this single dose of PAE reduced global DNA methylation. GSH supplementation concurrent with PAE normalized global DNA methylation levels. The same assays performed on quail hearts at 3 days (HH stage 19-20) of development, showed no difference in global DNA methylation between controls, ethanol-treated, GSH alone, and GSH plus ethanol-treated cohorts. CONCLUSIONS: GSH supplementation shows promise to inhibit effects of PAE by improving survival, reducing the incidence of morphological defects including CHDs, and preventing global hypomethylation of DNA in heart tissues.


Assuntos
Metilação de DNA/efeitos dos fármacos , Transtornos do Espectro Alcoólico Fetal/prevenção & controle , Glutationa/uso terapêutico , Cardiopatias Congênitas/prevenção & controle , Efeitos Tardios da Exposição Pré-Natal , Consumo de Bebidas Alcoólicas/efeitos adversos , Animais , Depressores do Sistema Nervoso Central/efeitos adversos , Avaliação Pré-Clínica de Medicamentos , Etanol/efeitos adversos , Feminino , Glutationa/farmacologia , Cardiopatias Congênitas/induzido quimicamente , Gravidez , Codorniz
16.
Toxicol Appl Pharmacol ; 398: 115029, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32376357

RESUMO

Resveratrol (RSV), a natural polyphenolic compound commonly found in food, has antioxidant and aryl hydrocarbon receptor (AHR) antagonist effects. We have recently demonstrated that AHR mediated reactive oxygen species (ROS) generation contributes to the cardiac developmental toxicity of ambient fine particle matter (PM2.5). Thus, we hypothesized that RSV protects against the cardiac developmental toxicity of PM2.5 by inhibiting ROS generation and AHR activity. To test this concept, we exposed zebrafish embryos to extractable organic matter (EOM) from PM2.5 in the presence or absence of RSV. We found that RSV significantly counteracted EOM-induced cardiac malformations in zebrafish embryos. The EOM-induced ROS production, DNA damage and apoptosis in the heart of zebrafish embryos were also counteracted by RSV supplementation. Furthermore, RSV attenuated EOM-induced changes in the expression of genes involved in cardiac development (nkx2.5, sox9b, axin2), oxidative stress (nrf2a, nrf2b, gstp1, gstp2, sod1, sod2, cat) and apoptosis (p53, bax). However, RSV did not suppress EOM-induced AHR activity. In conclusion, our data indicates that RSV protects against the PM2.5-induced heart malformations by inhibiting oxidative stress rather than through AHR antagonism.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/tratamento farmacológico , Material Particulado/efeitos adversos , Substâncias Protetoras/farmacologia , Resveratrol/farmacologia , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/metabolismo , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Coração/efeitos dos fármacos , Cardiopatias Congênitas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
17.
Environ Res ; 186: 109550, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32335433

RESUMO

BACKGROUND: Although there is evidence in experimental model systems that exposure to polycyclic aromatic hydrocarbons (PAHs) is linked with congenital heart defects (CHDs), few studies have examined the association in humans. We conducted a case-control study to examine the association between maternal exposure to PAHs and CHDs in offspring using data from the National Birth Defects Prevention Study (NBDPS) (1997-2011). METHODS: We obtained detailed information on maternal occupation during the month before to three months after conception. Expert raters, masked to case-control status, assessed job descriptions to assign categorical levels of exposure. Categories were quantitatively mapped to estimate cumulative exposure to PAHs, incorporating exposure intensity, frequency, work duration, and work hours. Quartiles were generated for cumulative maternal exposure to PAHs. Crude and adjusted odds ratios (ORs) with 95% confidence intervals (CIs) were estimated using unconditional logistic regression for quartiles of PAH exposure and six CHD groupings (e.g. conotruncal) and specific subtypes (e.g. tetralogy of Fallot [ToF]). Final models were adjusted for maternal age, race/ethnicity, education, smoking, anticonvulsant use, folic acid supplementation, and study center. RESULTS: There were 4,775 case and 7,734 control infants eligible for the study. The prevalence of occupational exposure to PAHs was 10.2% among both case and control mothers. In adjusted analysis, compared to mothers with no occupational PAH exposure, those in the highest quartile of exposure were more likely to have offspring in the conotruncal heart defects group (OR 1.41; 95% CI 1.00-2.00), and with ToF (OR 1.83; 95% CI 1.21-2.78). CONCLUSIONS: Women in the highest quartile of estimated cumulative occupational PAH exposure during early pregnancy were more likely to have offspring with conotruncal heart defects, specifically ToF, compared to women with no occupational PAH exposure. Other comparisons between PAHs and other CHDs subgroups did not show any statistically precise associations.


Assuntos
Cardiopatias Congênitas , Exposição Ocupacional , Hidrocarbonetos Policíclicos Aromáticos , Estudos de Casos e Controles , Feminino , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/epidemiologia , Humanos , Lactente , Exposição Materna/efeitos adversos , Exposição Ocupacional/efeitos adversos , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Gravidez , Fatores de Risco
18.
Toxicology ; 435: 152422, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32112805

RESUMO

Ribavirin has been proven to be an antiviral treatment, whereas there are still risks of hemolysis and congenital malformation. Abnormal cardiac development contributes to the occurrence and development of many heart diseases. However, there is so far no evidence that ribavirin induces human cardiac developmental toxicity. Herein, we employed the cardiac differentiation model of human induced pluripotent stem cells (hiPSCs) to determine the impact of ribavirin on heart development. Our data showed that ribavirin at clinically high concentrations (5 and 10 µM) significantly inhibited the proliferation and differentiation of hiPSCs from mesoderm to cardiac progenitor cells and cardiac progenitor cells to cardiomyocytes, but not from pluripotent status to mesoderm. Meanwhile, DCFH-DA staining revealed that ribavirin could increase ROS content in the mid-phase of differentiation. In addition, ribavirin treatment (1, 5 and 10 µM) remarkably caused DNA damage which was shown by the increase of γH2AX-positive cells and upregulation of the p53 during the differentiation of hiPSCs from mesoderm to cardiac progenitor cells. Moreover, exposuring to ribavirin (5 and 10 µM) markedly upregulated the expression of lncRNAs Gas5 in both mid-phase and late phase of differentiation and HBL1 in the mid-phase. In conclusion, our results suggest that ribavirin is detrimental in cardiac differentiation of hiPSCs, which may be associated with DNA damage, upregulated p53 and increased Gas5. It may provide the evidence for the rational clinical application of ribavirin.


Assuntos
Antivirais/toxicidade , Diferenciação Celular/efeitos dos fármacos , Cardiopatias Congênitas/induzido quimicamente , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Ribavirina/toxicidade , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Histonas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Medição de Risco , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Pharmacoepidemiol Drug Saf ; 29(9): 1151-1158, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31863536

RESUMO

PURPOSE: To evaluate the use of data from population-based surveys such as the National Health and Nutrition Examination Survey (NHANES) for external adjustment for confounders imperfectly measured in health care databases in the United States. METHODS: Our example study used Medicaid Analytic eXtract (MAX) data to estimate the relative risk (RR) for prenatal serotonin-norepinephrine reuptake inhibitors (SNRIs) exposure and cardiac defects. Smoking and obesity are known confounders poorly captured in databases. NHANES collects information on lifestyle factors, depression, and prescription medications. External adjustment requires information on the prevalence of confounders and their association with SNRI use; which was obtained from the NHANES. It also requires estimates of their association with the outcome, which were based on the literature and allowed us to correct the RR using sensitivity analyses. RESULTS: In MAX, the RR for the association between prenatal SNRI exposure and cardiac defects was 1.51 unadjusted and 1.20 adjusted for measured confounders and restricted to women with depression. In NHANES, among women of childbearing age with depression, the prevalence of smoking was 60.2% (95% Confidence Interval 43.2, 74.3) for SNRI users and 44.1% (39.6, 48.8) for nonusers of antidepressants. The corresponding estimates for obesity were 59.2% (43.2, 74.3) and 40.5% (35.9, 45.0), respectively. If the associations between smoking and obesity with cardiac defects are independent from each other and from other measured confounders, additional adjustment for smoking and obesity would move the RR from 1.20 to around 1.10. CONCLUSION: National surveys like NHANES are readily available sources of information on potential confounders and they can be used to assess and improve the validity of RR estimates from observational studies missing data on known risk factors.


Assuntos
Fatores de Confusão Epidemiológicos , Bases de Dados Factuais/estatística & dados numéricos , Inquéritos Nutricionais/estatística & dados numéricos , Farmacoepidemiologia/métodos , Medicamentos sob Prescrição/efeitos adversos , Antidepressivos/efeitos adversos , Depressão/tratamento farmacológico , Prescrições de Medicamentos/estatística & dados numéricos , Feminino , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/epidemiologia , Humanos , Estilo de Vida , Obesidade/epidemiologia , Aceitação pelo Paciente de Cuidados de Saúde/estatística & dados numéricos , Farmacoepidemiologia/estatística & dados numéricos , Gravidez , Complicações na Gravidez/tratamento farmacológico , Prevalência , Probabilidade , Fatores de Risco , Inibidores da Recaptação de Serotonina e Norepinefrina/efeitos adversos , Fumar/epidemiologia , Estados Unidos/epidemiologia
20.
J Hazard Mater ; 385: 121521, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-31699484

RESUMO

Trichloroethylene (TCE), a widely used chlorinated solvent, is a common environmental pollutant. Current evidence shows that TCE could induce heart defects during embryonic development, but the underlining mechanism(s) remain unclear. Since activation of the aryl hydrocarbon receptor (AHR) could induce oxidative stress, we hypothesized that AHR-mediated oxidative stress may play a role in the cardiac developmental toxicity of TCE. In this study, we found that the reactive oxygen species (ROS) scavenger, N-Acetyl-L-cysteine (NAC), and AHR inhibitors, CH223191 (CH) and StemRegenin 1, significantly counteracted the TCE-induced heart malformations in zebrafish embryos. Moreover, both CH and NAC suppressed TCE-induced ROS and 8-OHdG (8-hydroxy-2' -deoxyguanosine). TCE did not affect ahr2 and cyp1a expression, but increased cyp1b1 expression, which was restored by CH supplementation. CH also attenuated the TCE-induced mRNA expression changes of Nrf2 signalling genes (nrf2b, gstp2, sod2, ho1, nqo1) and cardiac differentiation genes (gata4, hand2, c-fos, sox9b). In addition, the TCE enhanced SOD activity was attenuated by CH. Morpholino knockdown confirmed that AHR mediated the TCE-induced ROS and 8-OHdG generation in the heart of zebrafish embryos. In conclusion, our results suggest that AHR mediates TCE-induced oxidative stress, leading to DNA damage and heart malformations in zebrafish embryos.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Cardiopatias Congênitas/embriologia , Receptores de Hidrocarboneto Arílico/metabolismo , Tricloroetileno/toxicidade , Proteínas de Peixe-Zebra/metabolismo , Acetilcisteína/farmacologia , Animais , Compostos Azo/farmacologia , Cardiotoxicidade/embriologia , Dano ao DNA/efeitos dos fármacos , Coração/embriologia , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Purinas/farmacologia , Pirazóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA