Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
FASEB J ; 37(7): e23011, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37249374

RESUMO

Autophagy inhibition is known to be involved in the development of adult osteoarthritis. Dexamethasone, as a synthetic glucocorticoid, is widely used for premature delivery and related pregnancy diseases in clinics. We have previously shown that prenatal dexamethasone exposure (PDE) was associated with increased susceptibility to postnatal osteoarthritis in offspring. However, whether the occurrence of fetal-originated adult osteoarthritis induced by PDE is related to autophagy remains unclear. In this study, we first found that PDE could increase the mRNA and protein expression of cartilage matrix-degrading enzymes (MMP3, MMP13, and ADAMTS5) and decrease the cartilage matrix contents in adult offspring, and the in vitro results suggested that this might be related to the autophagy inhibition of chondrocytes. Further, we demonstrated a persistent autophagy inhibition with autolysosome accumulation, low expression of cathepsin D (CTSD), increased H3K9ac level, and expression of miR-1912-3p in the cartilage of PDE offspring from fetus to adulthood. In vitro experiments showed that dexamethasone inhibited autophagy flux and CTSD expression in fetal chondrocytes, while overexpression of CTSD could alleviate the inhibition of autophagic flux induced by dexamethasone. Finally, we confirmed that dexamethasone increased the H3K9ac level and expression of miR-1912-3p through activation of the glucocorticoid receptor (GR), resulting in the decreased expression of CTSD and inhibition of autophagy flux in fetal chondrocytes. In conclusion, intrauterine miR-1912-3p/CTSD programming-mediated autophagy inhibition promoted the susceptibility to osteoarthritis in PDE adult offspring rats. This study provides new ideas for exploring early prevention and therapeutic targets in fetal-originated osteoarthritis.


Assuntos
MicroRNAs , Osteoartrite , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Humanos , Feminino , Ratos , Masculino , Animais , Ratos Wistar , Catepsina D , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Osteoartrite/induzido quimicamente , Osteoartrite/genética , Osteoartrite/metabolismo , Dexametasona/toxicidade , MicroRNAs/genética , Autofagia
2.
Cell Commun Signal ; 22(1): 62, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-38263047

RESUMO

BACKGROUND: Previous research has reported that prenatal exposure to dexamethasone (PDE) results in organ dysplasia and increased disease susceptibility in offspring. This study aimed to investigate the epigenetic mechanism of metabolic syndrome induced by PDE in offspring. METHODS: Pregnant Wistar rats were administered dexamethasone, and their offspring's serum and liver tissues were analyzed. The hepatocyte differentiation model was established to unveil the molecular mechanism. Neonatal cord blood samples were collected to validate the phenomenon and mechanism. RESULTS: The findings demonstrated that PDE leads to insulin resistance and typical metabolic syndrome traits in adult offspring rats, which originated from fetal liver dysplasia. Additionally, PDE reduced serum corticosterone level and inhibited hepatic insulin-like growth factor 1 (IGF1) signaling in fetal rats. It further revealed that liver dysplasia and functional impairment induced by PDE persist after birth, driven by the continuous downregulation of serum corticosterone and hepatic IGF1 signaling. Both in vitro and in vivo experiments confirmed that low endogenous corticosterone reduces the histone 3 lysine 9 acetylation (H3K27ac) level of IGF1 and its expression by blocking glucocorticoid receptor α, special protein 1, and P300 into the nucleus, resulting in hepatocyte differentiation inhibition and liver dysplasia. Intriguingly, neonatal cord blood samples validated the link between reduced liver function in neonates induced by PDE and decreased serum cortisol and IGF1 levels. CONCLUSIONS: This study demonstrated that low endogenous glucocorticoid level under PDE lead to liver dysplasia by downregulating the H3K27ac level of IGF1 and its expression, ultimately contributing to metabolic syndrome in adult offspring.


Assuntos
Síndrome Metabólica , Feminino , Gravidez , Animais , Ratos , Ratos Wistar , Corticosterona , Epigênese Genética , Hiperplasia , Dexametasona
3.
Acta Pharmacol Sin ; 45(6): 1237-1251, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38472317

RESUMO

Both epidemiological and animal studies suggest that adverse environment during pregnancy can change the offspring development programming, but it is difficult to achieve prenatal early warning. In this study we investigated the impact of prenatal dexamethasone exposure (PDE) on sperm quality and function of blood-testis barrier (BTB) in adult offspring and the underlying mechanisms. Pregnant rats were injected with dexamethasone (0.1, 0.2 and 0.4 mg·kg-1·d-1, s.c.) from GD9 to GD20. After weaning (PW4), the pups were fed with lab chow. At PW12 and PW28, the male offspring were euthanized to collect blood and testes samples. We showed that PDE significantly decreased sperm quality (including quantity and motility) in male offspring, which was associated with impaired BTB and decreased CX43/E-cadherin expression in the testis. We demonstrated that PDE induced morphological abnormalities of fetal testicle and Sertoli cell development originated from intrauterine. By tracing to fetal testicular Sertoli cells, we found that PDE dose-dependently increased expression of histone lysine demethylases (KDM1B), decreasing histone 3 lysine 9 dimethylation (H3K9me2) levels of follistatin-like-3 (FSTL3) promoter region and increased FSTL3 expression, and inhibited TGFß signaling and CX43/E-cadherin expression in offspring before and after birth. These results were validated in TM4 Sertoli cells following dexamethasone treatment. Meanwhile, the H3K9me2 levels of FSTL3 promoter in maternal peripheral blood mononuclear cell (PBMC) and placenta were decreased and its expression increased, which was positively correlated with the changes in offspring testis. Based on analysis of human samples, we found that the H3K9me2 levels of FSTL3 promoter in maternal blood PBMC and placenta were positively correlated with fetal blood testosterone levels after prenatal dexamethasone exposure. We conclude that PDE can reduce sperm quality in adult offspring rats, which is related to the damage of testis BTB via epigenetic modification and change of FSTL3 expression in Sertoli cells. The H3K9me2 levels of the FSTL3 promoter and its expression in the maternal blood PBMC can be used as a prenatal warning marker for fetal testicular dysplasia.


Assuntos
Barreira Hematotesticular , Dexametasona , Efeitos Tardios da Exposição Pré-Natal , Transdução de Sinais , Animais , Masculino , Feminino , Gravidez , Dexametasona/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Barreira Hematotesticular/efeitos dos fármacos , Barreira Hematotesticular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ratos , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ratos Sprague-Dawley , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia
4.
Cell Commun Signal ; 21(1): 294, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37853416

RESUMO

BACKGROUND: Adrenal gland is the synthesis and secretion organ of glucocorticoid, which is crucial to fetal development and postnatal fate. Recently, we found that prenatal dexamethasone exposure (PDE) could cause adrenal dysfunction in offspring rats, but its multigenerational genetic effects and related mechanisms have not been reported. METHODS: The PDE rat model was established, and female filial generation 1 (F1) rats mate with wild males to produce the F2, the same way for the F3. Three generation rats were sacrificed for the related detection. SW-13 cells were used to clarify the epigenetic molecular mechanism. RESULTS: This study confirmed that PDE could activate fetal adrenal glucocorticoid receptor (GR). The activated GR, on the one hand, up-regulated Let-7b (in human cells) to inhibit steroidogenic acute regulatory protein (StAR) expression directly; on the other hand, down-regulated CCCTC binding factor (CTCF) and up-regulated DNA methyltransferase 3a/3b (Dnmt3a/3b), resulting in H19 hypermethylation and low expression. The decreased interaction of H19 and let-7 can further inhibit adrenal steroidogenesis. Additionally, oocytes transmitted the expression change of H19/let-7c axis to the next generation rats. Due to its genetic stability, F2 generation oocytes indirectly exposed to dexamethasone also inhibited H19 expression, which could be inherited to the F3 generation. CONCLUSIONS: This cascade effect of CTCF/H19/Let-7c ultimately resulted in the transgenerational inheritance of adrenal steroidogenesis inhibition of PDE offspring. This study deepens the understanding of the intrauterine origin of adrenal developmental toxicity, and it will provide evidence for the systematic analysis of the transgenerational inheritance effect of acquired traits induced by PDE. Video Abstract.


Assuntos
Efeitos Tardios da Exposição Pré-Natal , Gravidez , Masculino , Ratos , Animais , Feminino , Humanos , Ratos Wistar , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Epigênese Genética , Metilação de DNA , Dexametasona/toxicidade
5.
Cell Biol Toxicol ; 39(5): 2051-2067, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-35246761

RESUMO

Dexamethasone is widely used to treat pregnancy disorders related to premature delivery. However, lots of researches have confirmed that prenatal dexamethasone exposure (PDE) could increase the risk of offspring multiple diseases. This study was designed to elucidate the epigenetic mechanism of adrenal developmental programming and explore its early warning marker in peripheral blood mononuclear cells (PBMC). We found the adrenal morphological and functional changes of PDE male offspring rats before and after birth, which were mainly performed as the decreased serum corticosterone concentration, steroidogenic acute regulatory (StAR) protein expression, and histone 3 lysine 27 acetylation (H3K27ac) level of steroidogenic factor 1 (SF1) promoter region and its expression. Simultaneously, the expressions of glucocorticoid receptor (GR) and histone acetylation enzyme 5 (HDAC5) in the PDE male fetal rats were increased. In vitro, dexamethasone reduced the expression of SF1, StAR, and cortisol production and still increased the expression of GR and HDAC5, the binding between GR and SF1 promoter region, and protein interaction between GR and HDAC5. GR siRNA or HDAC5 siRNA was able to reverse the above roles of dexamethasone. Furthermore, in vivo, we confirmed that H3K27ac levels of SF1 promoter region and its expression in PBMC of the PDE group were decreased before and after birth, showing a positive correlation with the same indexes in adrenal. Meanwhile, in clinical trials, we confirmed that prenatal dexamethasone application decreased H3K27ac of SF1 promoter region and its expression in neonatal PBMC. In conclusion, PDE-caused adrenal insufficiency of male offspring rats was related to adrenal GR activated by dexamethasone in uterus. The activated GR, on the one hand, increased its direct binding to SF1 promoter region to inhibit its expression, on the other hand, upregulated and recruited HDAC5 to decrease H3K27ac level of SF1 promoter region, and strengthened the inhibition of SF1 and subsequent StAR expression.


Assuntos
Insuficiência Adrenal , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Feminino , Humanos , Ratos , Masculino , Animais , Leucócitos Mononucleares , Ratos Wistar , Acetilação , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Histonas/metabolismo , Corticosterona , Dexametasona/farmacologia , Biomarcadores/metabolismo , RNA Interferente Pequeno/metabolismo
6.
Cell Biol Toxicol ; 39(3): 867-883, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-34537908

RESUMO

Prenatal dexamethasone exposure (PDE) induces long-term reproductive toxicity in female offspring. We sought to explore the transgenerational inheritance effects of PDE on diminished ovarian reserve (DOR) in female offspring. Dexamethasone was subcutaneously administered into pregnant Wistar rats from gestational day 9 (GD9) to GD20 to obtain fetal and adult offspring of the F1 generation. F1 adult females were mated with normal males to produce the F2 generation, and the F3 generation. The findings showed decrease of serum levels of anti-Müllerian hormone (AMH) that in the PDE group, decrease in number of primordial follicles, and upregulation of miR-17-5p expression before birth in F1 offspring rats. Expression of cyclin-dependent kinase inhibitor 1B (CDKN1B) and Forkhead Box L2 (FOXL2) were downregulated, and binding of FOXL2 and the CDKN1B promoter region was decreased in PDE groups of the F1, F2, and F3 generations. In vitro intervention experiments showed that glucocorticoid receptor (GR) was involved in activity of dexamethasone. These findings indicate that PDE can activate GR in fetal rat ovary and induce DOR of offspring, and its heritability is mediated by the cascade effect of miR-17-5p/FOXL2/CDKN1B. Increase in miR-17-5p expression in oocytes is the potential molecular basis for transgenerational inheritance of PDE effects.


Assuntos
MicroRNAs , Reserva Ovariana , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Masculino , Humanos , Ratos , Animais , Feminino , Ratos Wistar , Dexametasona/efeitos adversos , Inibidor de Quinase Dependente de Ciclina p27 , Reserva Ovariana/genética , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Oócitos , MicroRNAs/genética , Proteína Forkhead Box L2
7.
Pharmacol Res ; 175: 105990, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34808367

RESUMO

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


Assuntos
Anti-Inflamatórios/efeitos adversos , Dexametasona/efeitos adversos , Glucocorticoides/efeitos adversos , Histona Desacetilases/genética , Osteoporose/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Animais , Desenvolvimento Ósseo/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Corticosterona/sangue , Corticosterona/metabolismo , Feminino , Histona Desacetilases/metabolismo , Masculino , Osteoporose/genética , Osteoporose/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Wistar , Estresse Fisiológico
8.
J Appl Toxicol ; 42(3): 360-370, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34374451

RESUMO

During quantitative real-time polymerase chain reaction (RT-qPCR) data analysis, the selection of optimal housekeeping gene is necessary to ensure the accuracy of results. It is noteworthy that housekeeping genes commonly used in adult studies may not be applicable for fetus. However, the stability analysis of housekeeping gene in fetal kidney has not been reported. This study intends to screen the applicable compound housekeeping genes in rat fetal kidney. In this study, eight housekeeping genes used in kidney studies based on literature reports (GAPDH, ACTB, 18S, HPRT, YWHAZ, HMBS, PPIA, and TBP) were selected as the research object. Their expression levels in the rat fetal kidney in physiological condition and the intrauterine growth retardation (IUGR) model induced by prenatal dexamethasone exposure (PDE) (0.2 mg/kg·day from gestation Days 9 to 20) was measured. Furthermore, these eight housekeeping genes were used to conduct relative quantitative analysis of nephrin expression in the fetal kidney in PDE-induced IUGR model, to compare the influence of choosing different housekeeping gene on data analysis of nephrin expression and to verify the reliability of selected compound housekeeping genes. In this study, stable housekeeping genes of fetal kidney tissues in PDE-induced IUGR model were identified: ACTB, GAPDH, TBP, and HMBS for males; ACTB, YWHAZ, and GAPDH for females. Besides, our results suggest that ACTB + GAPDH were the best compound housekeeping genes for normalization analysis in male fetal kidney studies, and ACTB + YWHAZ in females. This study will provide an experimental evidence basis for the selection of housekeeping genes in the RT-qPCR experiment in renal development toxicology-related models.


Assuntos
Confiabilidade dos Dados , Expressão Gênica , Genes Essenciais , Genética/instrumentação , Rim/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/instrumentação , Animais , Feto , Ratos
9.
Toxicol Appl Pharmacol ; 406: 115243, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32949581

RESUMO

BACKGROUND: In quantitative real-time PCR (qRT-PCR), the expression levels of various adult reference genes may be unstable at different developmental periods and tissues, and will lead to inaccurate detected results. This study aimed to select and identify the optimal panel of reference genes in rat testis at different development periods. METHODS: We detected mRNA expression levels of five common rat testicular reference genes (GAPDH, ß-actin, 18s, RPS16 and RPL19) by qRT-PCR at different developmental periods (fetus, infancy, and adolescence), selected optimal panel of reference genes by combining with stability algorithms, and verified their tissue specificity. Lastly, we observed their mRNA expression alterations under pathological conditions to evaluate the stability and accuracy, and verify testicular dysplasia model. RESULTS: Based on comprehensive analysis, the best panel of reference genes of testis were GAPDH+ß-actin (at fetus) and GAPDH+RPL19 (at infancy and adolescent). Meanwhile, the best panel of reference genes of fetal testis was consistent with placenta and fetal hippocampus but different from fetal liver and kidney. Furthermore, in prenatal dexamethasone exposure (PDE) model, the results were consistent with those under physiological conditions, and the testicular steroidogenesis acute regulatory protein (StAR) was most obviously decreased when using the best panel of reference genes. CONCLUSION: In this study, rat testicular GAPDH+ß-actin for fetuses and GAPDH+RPL19 for infants and adolescents are recommended to be the optimal panel of reference genes. Respectively. The selected panel of reference genes was still stable under PDE condition. This study provided technical and theoretical supports for researches on testicular development toxicology.


Assuntos
Reação em Cadeia da Polimerase em Tempo Real/normas , Testículo/metabolismo , Actinas/genética , Animais , Feminino , Feto , Perfilação da Expressão Gênica , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora) , Hipocampo/embriologia , Hipocampo/metabolismo , Rim/embriologia , Rim/metabolismo , Fígado/embriologia , Fígado/metabolismo , Masculino , Gravidez , RNA Mensageiro/metabolismo , Ratos Wistar , Padrões de Referência , Proteínas Ribossômicas/genética , Testículo/embriologia , Testosterona/sangue , Transcriptoma
10.
Int J Mol Sci ; 17(4): 533, 2016 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-27070590

RESUMO

Increasing evidence has shown that many chronic diseases originate from early life, even before birth, through what are termed as fetal programming effects. Glucocorticoids are frequently used prenatally to accelerate the maturation of the lungs of premature infants. High-fat diets are associated with insulin resistance, but the effects of prenatal glucocorticoid exposure plus a postnatal high-fat diet in diabetes mellitus remain unclear. We administered pregnant Sprague-Dawley rats' intraperitoneal dexamethasone (0.1 mg/kg body weight) or vehicle at gestational days 14-20. Male offspring were administered a normal or high-fat diet starting from weaning. We assessed the effects of prenatal steroid exposure plus postnatal high-fat diet on the liver, pancreas, muscle and fat at postnatal day 120. At 15 and 30 min, sugar levels were higher in the dexamethasone plus high-fat diet (DHF) group than the vehicle plus high-fat diet (VHF) group in the intraperitoneal glucose tolerance test (IPGTT). Serum insulin levels at 15, 30 and 60 min were significantly higher in the VHF group than in the vehicle and normal diet group. Liver insulin receptor and adenosine monophosphate-activated protein kinase mRNA expressions and protein levels were lower in the DHF group. Insulin receptor and insulin receptor substrate-1 mRNA expressions were lower in the epididymal adipose tissue in the VHF and DHF groups. "Programming" of liver or epididymal adipose tissue resulted from prenatal events. Prenatal steroid exposure worsened insulin resistance in animals fed a high-fat diet.


Assuntos
Dexametasona/efeitos adversos , Diabetes Mellitus/etiologia , Dieta Hiperlipídica/efeitos adversos , Glucocorticoides/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Glicemia/análise , Diabetes Mellitus/sangue , Feminino , Teste de Tolerância a Glucose , Insulina/sangue , Resistência à Insulina , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/sangue , Ratos , Ratos Sprague-Dawley
11.
Toxicol Sci ; 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36790081

RESUMO

Decreased sperm quality is the main cause of male infertility. Studies have found that prenatal dexamethasone exposure (PDE) decreases sperm quality in male offspring after birth, but the mechanism is unclear. Wistar pregnant rats were subcutaneously injected with 0.1, 0.2 and 0.4 mg/kg.d dexamethasone at gestational day 9-20. The testes and sperm of first-generation (F1) offspring were collected, and F1 offspring were mated with wild-type female rats to obtain F2. Compared with the control group, F1 offspring in PDE group had lower sperm count and motility after birth, and the deformity rate increased. F2 fetal rats' body length and weight decreased, and the intrauterine growth retardation rate increased. Meanwhile, PDE decreased the expression of connexin 43 (CX43) in offspring testes, while T-box transcription factor 2 (TBX2) promoter region histone 3 lysine 9 acetylation (H3K9ac) level and its expression were increased. Traced back to F1 fetus testes, PDE increased the expression of glucocorticoid receptor (GR) and P300, activated GR protein into the nucleus, and made GR act on the TBX2 promoter region. Further, a series of Sertoli cell interventions confirmed that dexamethasone promoted GR to recruit P300, increased the H3K9ac level of TBX2 promoter region and its expression, and inhibited the expression of CX43. This study confirmed that PDE decreased sperm quality of male offspring, which is related to the epigenetic programming of TBX2/CX43 in the Sertoli cells, provided a theoretical and experimental basis for guiding the rational use of drugs during pregnancy.

12.
J Adv Res ; 47: 123-135, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-35953031

RESUMO

INTRODUCTION: Fetal-originated osteoarthritis is relative to poor cartilage quality and may exhibit transgenerational genetic effects. Previous findings revealed prenatal dexamethasone exposure (PDE) induced poor cartilage quality in offspring. OBJECTIVES: This study focused on further exploring molecular mechanism, heritability, and early intervention of fetal-originated osteoarthritis. METHODS: Pregnant rats (F0) were segregated into control and PDE groups depending upon whether dexamethasone was administered on gestational days (GDs) 9-20. Some female offspring were bred with healthy males during postnatal week (PW) 8 to attain the F2 and F3 generations. The F3-generation rats were administrated with glucosamine intragastrically at PW12 for 6 weeks. The knee cartilages of male and female rats at different time points were harvested to assay their morphologies and functions. Furthermore, primary chondrocytes from the F3-generation rats were isolated to confirm the mechanism and intervention target of glucosamine. RESULTS: Compared with the control, female and male rats in each generation of PDE group showed thinner cartilage thicknesses; shallower and uneven staining; fewer chondrocytes; higher Osteoarthritis Research Society International scores; and lower mRNA and protein expression of SP1, TGFßR1, Smad2, SOX9, ACAN and COL2A1. After F3-generation rats were treated with glucosamine, all of the above changes could be reversed. In primary chondrocytes isolated from the F3-generation rats of PDE group, glucosamine promoted SP1 expression and binding to TGFßR1 promoter to increase the expression of TGFßR1, p-Smad2, SOX9, ACAN and COL2A1, but these were prevented by SB431542 (a potent and selective inhibitor of TGFßR1). CONCLUSIONS: PDE induced chondrodysplasia in offspring and stably inherited in F3-generation rats, which was related to decreased expression of SP1/TGFßR1/Smad2/SOX9 pathway to reduce the cartilage matrix synthesis, without major sex-based variations. Glucosamine could alleviate the poor genetic cartilage quality in offspring induced by PDE by up-regulating SP1/TGFßR1 signaling, which was prevented by a TGFßR1 inhibitor. This study elucidated the molecular mechanism and therapeutic target (TGFßR1) of genetic chondrodysplasia caused by PDE, which provides a research basis for precisely treating fetal-originated osteoarthritis.


Assuntos
Cartilagem Articular , Osteoartrite , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Humanos , Ratos , Masculino , Feminino , Animais , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Wistar , Cartilagem Articular/metabolismo , Osteoartrite/induzido quimicamente , Osteoartrite/tratamento farmacológico , Osteoartrite/genética , Dexametasona/efeitos adversos , Dexametasona/metabolismo , Glucosamina/efeitos adversos , Glucosamina/metabolismo , Fatores de Crescimento Transformadores/efeitos adversos , Fatores de Crescimento Transformadores/metabolismo
13.
Biochem Pharmacol ; 206: 115306, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36326533

RESUMO

Mounting evidence indicates that adverse intrauterine conditions increase offspring's hypercholesterolemia susceptibility in adulthood. This study aimed to confirm prenatal dexamethasone exposure (PDE)-induced hypercholesterolemia susceptibility in female adult offspring rats, and elucidate its intrauterine programming mechanism. Pregnant Wistar rats were injected with dexamethasone subcutaneously (0, 0.1 and 0.2 mg/kg·d) from gestational day (GD) 9 to 20. Serum and liver of the female offspring were collected at GD21 and postnatal week (PW) 12 and 28. PDE offspring showed elevated serum total cholesterol (TCH) levels and a cholesterol phenotype of high cardiovascular disease risk at PW12 and PW28. The histone acetylation levels of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (Hmgcr) and its expression were consistently increased in the PDE offspring both in utero and after birth. Moreover, PDE promoted glucocorticoid receptor (GR) nuclear translocation and miR-133a-3p expression and inhibited sirtuin-1 (Sirt1) expression in the fetal liver. In vitro, dexamethasone increased intracellular and supernatant TCH levels and miR-133a-3p expression, decreased SIRT1 expression, and promoted HMGCR histone acetylation and expression in bone marrow mesenchymal stem cells (BMSCs) hepatoid differentiated cells and HepG2 cell line. GR siRNA, miR-133a-3p inhibitor or SIRT1 overexpression reversed dexamethasone-induced downstream molecular and phenotypic changes. Furthermore, elevated TCH levels in umbilical cord blood and increased HMGCR expression in peripheral blood mononuclear cells (PBMCs) were observed in human female neonates who had received dexamethasone treatment during pregnancy. In conclusion, PDE can cause persistent enhancement of hepatic cholesterol synthesis function before and after birth through GR/miR-133a-3p/Sirt1 pathway, eventually leading to increased hypercholesterolemia susceptibility in female offspring rats.


Assuntos
Hipercolesterolemia , MicroRNAs , Efeitos Tardios da Exposição Pré-Natal , Adulto , Animais , Feminino , Humanos , Gravidez , Ratos , Colesterol , Dexametasona/toxicidade , Epigênese Genética , Histonas/metabolismo , Hipercolesterolemia/induzido quimicamente , Hipercolesterolemia/genética , Leucócitos Mononucleares/metabolismo , MicroRNAs/genética , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Wistar , Receptores de Glucocorticoides/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo
14.
J Reprod Immunol ; 151: 103619, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35367871

RESUMO

Dexamethasone has been widely used in clinical practice to promote fetal lung maturity and reduce neonatal respiratory distress syndrome and perinatal mortality. Nevertheless, its administration is a double-edged sword, as a large number of studies have shown that there are obvious disadvantages in pregnant women and fetal development. In this review, we comprehensively retrospect the latest literature on the toxicological effects and mechanisms of dexamethasone on fetal development, in an attempt to provide a valuable basis for further studies and clinical trials in the future. Overall, prenatal dexamethasone exposure could lead to some adverse consequences on fetal organ systems through intrauterine programming based on the results of current animal and human researches. Potential sequelae include osteoarthritis, hypertension, fatty liver, glomerulosclerosis, depression, diabetes and infertility, some of which can pass on to the next generation. It must be noted that the evidence in humans is preliminary and limited by the small sample size. More studies in large-scale populations are needed to confirm if it raises the risk of sequelae in humans. In addition, we strongly support the application of dexamethasone as a pharmaceutical therapy in pregnant women with coronavirus disease 2019 before a better therapy is developed. However, the adverse side effects that may arise also cannot be ignored.


Assuntos
Tratamento Farmacológico da COVID-19 , Complicações Infecciosas na Gravidez , Síndrome do Desconforto Respiratório do Recém-Nascido , Animais , Dexametasona/efeitos adversos , Feminino , Desenvolvimento Fetal , Humanos , Recém-Nascido , Gravidez
15.
Biochem Pharmacol ; 199: 115018, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35351429

RESUMO

The adverse environment during pregnancy could change epigenetic modifications in germ cells, leading to transgenerational effects. This study aims to explore transgenerational-inheritance of inhibited testicular testosterone synthesis induced by prenatal dexamethasone exposure (PDE) and its mechanism. Wistar pregnant rats were subcutaneously injected with 0.2 mg/kg.d dexamethasone at gestational-day (GD) 9-20. Blood and testes of F1/F3 were obtained by maternal inheritance before and after birth. Meanwhile, Leydig cells were treated with dexamethasone and related interventions to confirm its direct effect and mechanism. The results showed that, in PDE F1 fetus, the serum testosterone level decreased, steroidogenic acute regulatory protein (StAR) decreased, and imprinted miR-466b-3p reduced, histone deacetylase 7 (HDAC7) upregulated, histone 3 lysine 9 acetylation (H3K9ac) level of StAR promoter decreased in testis. Meanwhile, consistent changes were observed in F1/F3 testes. In addition, miR-466b-3p expression in PDE F1/F2 oocytes also decreased. Further, series cell interventions confirmed that dexamethasone could reduce imprinted miR-466b-3p expression, increase HDAC7 expression, and reduce StAR acetylation level and expression by glucocorticoid receptor, finally causing testosterone synthesis inhibition. This study provided an experimental basis for confirming the developmental toxicity in offspring testis induced by PDE and its maternal transgenerational inheritance, offering a potential target for early warning and intervention therapy.


Assuntos
MicroRNAs , Efeitos Tardios da Exposição Pré-Natal , Animais , Dexametasona/farmacologia , Feminino , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Masculino , MicroRNAs/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Wistar , Testículo , Testosterona/metabolismo
16.
Biochem Pharmacol ; 203: 115196, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35917869

RESUMO

An adverse environment during pregnancy leads to intrauterine programming changes in multiple generations, resulting in the multigenerational inheritance of abnormal phenotype. Here, we reported the multigenerational inheritance of poor articular cartilage quality induced by prenatal dexamethasone exposure (PDE) with 0.2 mg/kg·d dexamethasone from gestational day (GD) 9 to GD20 in Wistar rats and investigated its intrauterine epigenetic programming mechanism. For the F1 female offspring at GD20, we found that the matrix synthesis of cartilage was suppressed, the histone 3 lysine 9 acetylation (H3K9ac) level and mRNA expression of the TGFß signaling pathway were decreased, and the expression of histone deacetylase (HDAC) 2 was increased in the cartilage. Meaningfully, the similar changes were also found in the F1-F3 female adult offspring. Furthermore, PDE decreased the expression of miR-92a-3p in the oocytes of the F1-F2 offspring and in the cartilage of the F1-F3 generations. In vitro, the effect of dexamethasone on chondrocytes revealed that it inhibited the expression of miR-92a-3p through activating and binding glucocorticoid receptor, and reduced the H3K9ac level in the promoter of the TGFß signaling pathway through the increased HDAC2. In conclusion, PDE induces the multigenerational inheritance of poor articular cartilage quality in female adult offspring; the potential mechanism involves the intergenerational effect of low miR-92a-3p expression in oocytes and low functional programming of TGFß signaling pathway induced by decreased H3K9ac level via upregulating HDAC2. This study provides a new perspective to explain the multi-generation inheritance of PDE-induced organ dysplasia in adult offspring.


Assuntos
Cartilagem Articular , MicroRNAs , Efeitos Tardios da Exposição Pré-Natal , Animais , Cartilagem Articular/metabolismo , Dexametasona/toxicidade , Feminino , Humanos , MicroRNAs/metabolismo , Oócitos/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Wistar , Fator de Crescimento Transformador beta/metabolismo
17.
Biochem Pharmacol ; 206: 115264, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36174767

RESUMO

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


Assuntos
Dexametasona , MicroRNAs , Osteoporose , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Masculino , Gravidez , Ratos , Corticosterona/efeitos adversos , Dexametasona/toxicidade , Fator de Crescimento Insulin-Like I/genética , MicroRNAs/genética , Osteogênese , Osteoporose/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Wistar , Glucocorticoides
18.
Sci Total Environ ; 826: 154095, 2022 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-35219660

RESUMO

The prevalence of diabetes in children and adolescents has been rising gradually, which is relevant to adverse environment during development, especially prepartum. We aimed to explore the effects of prenatal dexamethasone exposure (PDE) on ß-cell function and glucose homeostasis in juvenile offspring rats. Pregnant Wistar rats were subcutaneously administered with dexamethasone [0.1, 0.2, 0.4mg/(kg.d)] from gestational day 9 to 20. PDE impaired glucose tolerance in the male offspring rather than the females. In male offspring, PDE impaired the development and function of ß-cells, accompanied with lower H3K9ac, H3K14ac and H3K27ac levels in the promoter region of angiotensin-converting enzyme 2 (ACE2) as well as suppressed ACE2 expression. Meanwhile, PDE increased expression of glucocorticoid receptor (GR) and histone deacetylase 3 (HDAC3) in fetal pancreas. Dexamethasone also inhibited ACE2 expression and insulin production in vitro. Recombinant expression of ACE2 restored insulin production inhibited by dexamethasone. In addition, dexamethasone activated GR and HDAC3, increased protein interaction of GR with HDAC3, and promoted the binding of GR-HDAC3 complex to ACE2 promoter region. Both RU486 and TSA abolished dexamethasone-induced decline of histone acetylation and ACE2 expression. In summary, suppression of ACE2 is involved in PDE induced ß-cell dysfunction and glucose intolerance in juvenile male offspring rats.


Assuntos
Intolerância à Glucose , Insulinas , Efeitos Tardios da Exposição Pré-Natal , Adolescente , Enzima de Conversão de Angiotensina 2 , Animais , Dexametasona/toxicidade , Repressão Epigenética , Feminino , Intolerância à Glucose/induzido quimicamente , Humanos , Insulinas/metabolismo , Insulinas/toxicidade , Masculino , Pâncreas/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Wistar , Receptores de Glucocorticoides
19.
Toxicology ; 449: 152664, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33359579

RESUMO

As a synthetic glucocorticoid, dexamethasone has been widely used in the clinical treatment of premature birth and related pregnant diseases, but its clinical use is still controversial due to developmental toxicity. This study aimed to confirm the proliferation inhibitory effect of pregnant dexamethasone exposure (PDE) on fetal liver development and elucidate its molecular mechanism. In vitro studies, we found that dexamethasone inhibited hepatocyte proliferation through autophagy activated by glucocorticoid receptor (GR)-forkhead protein O1 (FOXO1) pathway. Subsequently, in vivo, we confirmed in a PDE rat model that male fetal liver proliferation was inhibited, and the expression of the GR-FOXO1 pathway and autophagy were increased. Taken together, PDE induces autophagy by activating the GR-FOXO1 pathway, which leads to fetal liver proliferation inhibition and dysplasia in offspring rats. This study confirmed that dexamethasone activates cell autophagy in utero through the GR-FOXO1 pathway, thereby inhibiting hepatocyte proliferation and liver development, which provides theoretical basis for understanding the developmental toxicity of dexamethasone and guiding the rational clinical use.


Assuntos
Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dexametasona/toxicidade , Desenvolvimento Fetal/efeitos dos fármacos , Fígado/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Autofagia/fisiologia , Proliferação de Células/fisiologia , Dexametasona/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Desenvolvimento Fetal/fisiologia , Glucocorticoides/toxicidade , Fígado/embriologia , Fígado/patologia , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Wistar
20.
Reprod Toxicol ; 105: 211-220, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34537367

RESUMO

The choice of reference gene is crucial for quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) assay. To screen and determine the suitable reference genes in fetal rat pancreas, we selected eight candidate reference genes (Gapdh, Actb, Rn18 s, B2m, Rpl13a, Tbp, Ywhaz and Ubc), and evaluated the constancy of gene expression from fetal rat pancreases in non-pathological situation and prenatal dexamethasone exposure (PDE) model, using four algorithms: GeNorm, NormFinder, Bestkeeper and Comparative ΔCt method. In addition, the alteration of mRNA levels of pancreatic insulin was compared between control and PDE groups to validate the reliability of selected reference genes for data normalization of RT-qPCR. The comprehensive ranking of reference genes under physiological condition was as follow: Gapdh > Actb > Ywhaz > Ubc > Rn18s > Rpl13a > B2m > Tbp (female); Actb > Ywhaz > Gapdh > Ubc > B2m > Rpl13a > Rn18 s | Tbp (male). The top ranking reference genes were also stably expressed in PDE fetal pancreas. The best reference gene combinations are: Ywhaz+Actb for female and Ywhaz+Gapdh for male fetal rat pancreas, respectively. Compared with low ranking or single reference gene, the change trend of insulin mRNA normalized by the best reference gene combination between control and PDE groups was more significant and consistent with that of serum insulin level. In conclusion, our results provided the optimal combination of stable reference genes for RT-qPCR assay in pancreatic developmental toxicity study.


Assuntos
Feto , Expressão Gênica , Pâncreas , Reação em Cadeia da Polimerase Via Transcriptase Reversa/normas , Animais , Feminino , Insulina/sangue , Masculino , Ratos Wistar , Padrões de Referência , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA