1
|
Li S, Lu T, Lin Z, Zhang Y, Zhou X, Li M, Miao H, Yang Z, Han X. Supplementation with probiotics co-cultivation improves the reproductive performance in a sow-piglet model by mother-infant microbiota transmission and placental mTOR signaling. World J Microbiol Biotechnol 2024; 41:13. [PMID: 39704872 DOI: 10.1007/s11274-024-04222-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Maternal nutritional supplementation has a profound effect on the growth and development of offspring. FAM® is produced by co-cultivation of Lactobacillus acidophilus and Bacillus subtilis and has been demonstrated to potentially alleviate diarrhea, improve growth performance and the intestinal barrier integrity of weaned piglets. This study aimed to explore how maternal FAM improves the reproductive performance through mother-infant microbiota, colostrum and placenta. A total of 40 pregnant sows (Landrace × Large White) on d 85 of gestation with a similar parity were randomly divided into two groups (n = 20): the control group (Con, basal diet) and the FAM group (FAM, basal diet supplemented with 0.2% FAM). The experimental period was from d 85 of gestation to d 21 of lactation. The results revealed that maternal supplementation with FAM significantly decreased the number of weak-born litters and the incidence of diarrhea, as well as increasing birth weight and average weaning weight, accompanied by increased levels of colostrum nutrient composition and immunoglobulins. In addition, FAM modulated the structure of mother-infant microbiota and promoted the vertical transmission of beneficial bacteria, such as Verrucomicrobiota and Akkermansia. Furthermore, FAM contributed to improving the expression of GLU and AA transporters in the placenta, and increasing the activity of the mTOR signaling pathway. Collectively, maternal supplementation with FAM during late pregnancy and lactation could improve reproductive performance through the transmission of beneficial mother-infant microbiota and placental mTOR signaling pathway and promote fetal development.
Collapse
Affiliation(s)
- Suchen Li
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Tingting Lu
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Zhixin Lin
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Yuanyuan Zhang
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Xinchen Zhou
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Meng Li
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Hui Miao
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Zhiren Yang
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Xinyan Han
- Hainan Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Zha X, Fang M, Zhong W, Chen L, Feng H, Zhang M, Wang H, Zhang Y. Dose-, stage- and sex- difference of prenatal prednisone exposure on placental morphological and functional development. Toxicol Lett 2024; 402:68-80. [PMID: 39580039 DOI: 10.1016/j.toxlet.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/25/2024]
Abstract
Prednisone, a synthetic glucocorticoid, is commonly used to treat autoimmune diseases in pregnant women. However, some studies suggest that the use of prednisone during pregnancy may lead to adverse pregnancy outcomes. In this study, we established PPE mouse models at different doses (0.25, 0.5, 1.0 mg/kg·d) and different stages (whole pregnancy, early pregnancy and middle-late pregnancy) and determined outcomes on the placenta and fetus. The results of our study indicated that at the highest dose of 1 mg/kg PPE using a GD 0-18 dosing regime, PPE caused placental morphological changes measured as a decrease in placental weight relative to controls and a decrease in the placenta junctional zone (JZ)/labyrinth zone (LZ) ratio. No changes were observed on the fetuses for number of live, stillborn, and absorbed fetuses between the experimental groups and the control group. In the placentas at some doses, there were decreases in cell proliferation markers measured at the RNA and protein level by Western blot and increased apoptosis. Measures of gene expression at the mRNA level showed altered nutrients (including glucose, amino acid, and cholesterol) transport gene expressions with the most significant change associated with the male placentas at high-dose and whole pregnancy PPE group. It was further found that PPE led to the inhibition of the insulin-like growth factor 2 (IGF2)/insulin-like growth factor 1 receptor (IGF1R) signaling pathway, which was well correlated with the indicators of cell proliferation, syncytialization and nutrient (glucose and amino acid) transport indices. In conclusion, PPE can alter placental morphology and nutrient transport function, with differences in effect related to dose, stage and gender. Differential gene expressions measured for genes of the IGF2/IGF1R signaling pathway suggested this pathway may be involved in the effects seen with PPE. This study provides a theoretical and experimental basis for enhancing the understanding of the effects of prednisone use on placenta during human pregnancy but does not currently raise concerns for human use as effects were not seen on the fetuses and while the effects on cell proliferation are informative they were inconsistent and the differential effects on female and male placentas unexplained suggesting that further work is required to elucidate if these findings have relevance for human use of PPE during pregnancy.
Collapse
Affiliation(s)
- Xiaomeng Zha
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Man Fang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wen Zhong
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liang Chen
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Feng
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Min Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China
| | - Hui Wang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China.
| |
Collapse
|
3
|
Huang S, Yao B, Guo Y, Zhang Y, Li H, Zhang Y, Liu S, Wang X. Human trophoblast organoids for improved prediction of placental ABC transporter-mediated drug transport. Toxicol Appl Pharmacol 2024; 492:117112. [PMID: 39326791 DOI: 10.1016/j.taap.2024.117112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
ATP-binding cassette (ABC) transporters, the important transmembrane efflux transporters, play an irreplaceable role in the placenta barrier. The disposition and drug-drug interaction of clinical drugs are also closely related to the functions of ABC transporters. The trophoblast is a unique feature of the placenta, which is crucial for normal placentation and maintenance during pregnancy. ABC transporters are abundantly expressed in placental syncytiotrophoblast, especially P-gp, BCRP, and MRPs. However, due to the lack of appropriate modeling systems, the molecular mechanisms of regulation between ABC transporters and trophoblast remains unclear. In this report, trophoblast organoids were cultured from human placental villi and developed into three-dimension structures with cavities. Trophoblast organoids exhibited transporter expression and localization comparable to that in villous tissue, indicating their physiological relevance for modeling drug transport. Moreover, fluorescent substrates can accumulate in organoids and be selectively inhibited by inhibitors, indicating the efflux function of ABC transporters (P-gp, BCRP, MRP1, and MRP2) in organoids. Two commonly used hypertension drugs and three antipsychotics were chosen to further validate this drug transport model and demonstrate varying degrees of inhibitory effects on ABC transporters. Overall, a new drug transport model mediated by ABC transporter has been successfully established based on human trophoblast organoids, which can be used to study drug transport in the placenta.
Collapse
Affiliation(s)
- Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanqing Guo
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Haichuan Li
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yi Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
4
|
Zhao C, He L, Li L, Deng F, Zhang M, Wang C, Qiu J, Gao Q. Prenatal glucocorticoids exposure and adverse cardiovascular effects in offspring. Front Endocrinol (Lausanne) 2024; 15:1430334. [PMID: 39351527 PMCID: PMC11439645 DOI: 10.3389/fendo.2024.1430334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Glucocorticoids (GCs) are steroid hormones fundamental to the body's normal physiological functions and are pivotal in fetal growth and development. During gestation, the mother's cortisol concentration (active GCs) escalates to accommodate the requirements of fetal organ development and maturation. A natural placental GCs barrier, primarily facilitated by 11β hydroxysteroid dehydrogenase 2, exists between the mother and fetus. This enzyme transforms biologically active cortisol into biologically inactive corticosterone, thereby mitigating fetal GCs exposure. However, during pregnancy, the mother may be vulnerable to adverse factor exposures such as stress, hypoxia, caffeine, and synthetic GCs use. In these instances, maternal serum GCs levels may surge beyond the protective capacity of the placental GCs barrier. Moreover, these adverse factors could directly compromise the placental GCs barrier, resulting in excessive fetal exposure to GCs. It is well-documented that prenatal GCs exposure can detrimentally impact the offspring's cardiovascular system, particularly in relation to blood pressure, vascular function, and heart function. In this review, we succinctly delineate the alterations in GCs levels during pregnancy and the potential mechanisms driving these changes, and also analyze the possible causes of prenatal GCs exposure. Furthermore, we summarize the current advancements in understanding the adverse effects and mechanisms of prenatal GCs exposure on the offspring's cardiovascular system.
Collapse
Affiliation(s)
- Chenxuan Zhao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei He
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fengying Deng
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Changhong Wang
- Genetics and Prenatal Diagnosis Center, Fuyang People’s Hospital, Fuyang, China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Sze Y, Brunton PJ. How is prenatal stress transmitted from the mother to the fetus? J Exp Biol 2024; 227:jeb246073. [PMID: 38449331 DOI: 10.1242/jeb.246073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo-pituitary-adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11β-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
- Zhejiang University-University of Edinburgh Joint Institute, Haining, Zhejiang 314400, P.R. China
| |
Collapse
|
6
|
Dai Y, Peng Y, Hu W, Liu Y, Wang H. Prenatal amoxicillin exposure induces developmental toxicity in fetal mice and its characteristics. J Environ Sci (China) 2024; 137:287-301. [PMID: 37980015 DOI: 10.1016/j.jes.2023.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 11/20/2023]
Abstract
Amoxicillin, a widely used antibiotic in human and veterinary pharmaceuticals, is now considered as an "emerging contaminant" because it exists widespreadly in the environment and brings a series of adverse outcomes. Currently, systematic studies about the developmental toxicity of amoxicillin are still lacking. We explored the potential effects of amoxicillin exposure on pregnancy outcomes, maternal/fetal serum phenotypes, and fetal multiple organ development in mice, at different doses (75, 150, 300 mg/(kg·day)) during late-pregnancy, or at a dose of 300 mg/(kg·day) during different stages (mid-/late-pregnancy) and courses (single-/multi-course). Results showed that prenatal amoxicillin exposure (PAmE) had no significant influence on the body weights of dams, but it could inhibit the physical development and reduce the survival rate of fetuses, especially during the mid-pregnancy. Meanwhile, PAmE altered multiple maternal/fetal serum phenotypes, especially in fetuses. Fetal multi-organ function results showed that PAmE inhibited testicular/adrenal steroid synthesis, long bone/cartilage and hippocampal development, and enhanced ovarian steroid synthesis and hepatic glycogenesis/lipogenesis, and the order of severity might be gonad (testis, ovary) > liver > others. Further analysis found that PAmE-induced multi-organ developmental and functional alterations had differences in stages, courses and fetal gender, and the most obvious changes might be in high-dose, late-pregnancy and multi-course, but there was no typical rule of a dose-response relationship. In conclusion, this study confirmed that PAmE could cause abnormal development and multi-organ function alterations, which deepens our understanding of the risk of PAmE and provides an experimental basis for further exploration of the long-term harm.
Collapse
Affiliation(s)
- Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yu Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yi Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
7
|
Souza LL, Moura EG, Lisboa PC. Can mothers consume caffeine? The issue of early life exposure and metabolic changes in offspring. Toxicol Lett 2024; 393:96-106. [PMID: 38387763 DOI: 10.1016/j.toxlet.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 01/02/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
Caffeine is a substance with central and metabolic effects. Although it is recommended that its use be limited during pregnancy, many women continue to consume caffeine. Direct and indirect actions of caffeine in fetuses and newborns promote adaptive changes, according to the Developmental Origins of Health and Diseases (DOHaD) concept. In fact, epidemiological and experimental evidence reveals the impact of early caffeine exposure. Here, we reviewed these findings with an emphasis on experimental models with rodents. The similarity of human and rodent caffeine metabolism allows the comprehension of molecular mechanisms affected by prenatal caffeine exposure. Maternal caffeine intake affects the body weight and endocrine system of offspring at birth and has long-term effects on the endocrine system, liver function, glucose and lipid metabolism, the cardiac system, the reproductive system, and behavior. Interestingly, some of these effects are sex dependent. Thus, the dose of caffeine considered safe for pregnant women may not be adequate for the prenatal period.
Collapse
Affiliation(s)
- Luana L Souza
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto G Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia C Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
8
|
Bakhireva LN, Solomon E, Roberts MH, Ma X, Rai R, Wiesel A, Jacobson SW, Weinberg J, Milligan ED. Independent and Combined Effects of Prenatal Alcohol Exposure and Prenatal Stress on Fetal HPA Axis Development. Int J Mol Sci 2024; 25:2690. [PMID: 38473937 PMCID: PMC10932119 DOI: 10.3390/ijms25052690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Prenatal alcohol exposure (PAE) and prenatal stress (PS) are highly prevalent conditions known to affect fetal programming of the hypothalamic-pituitary-adrenal (HPA) axis. The objectives of this study were to assess the effect of light PAE, PS, and PAE-PS interaction on fetal HPA axis activity assessed via placental and umbilical cord blood biomarkers. Participants of the ENRICH-2 cohort were recruited during the second trimester and classified into the PAE and unexposed control groups. PS was assessed by the Perceived Stress Scale. Placental tissue was collected promptly after delivery; gene and protein analysis for 11β-HSD1, 11β-HSD2, and pCRH were conducted by qPCR and ELISA, respectively. Umbilical cord blood was analyzed for cortisone and cortisol. Pearson correlation and multivariable linear regression examined the association of PAE and PS with HPA axis biomarkers. Mean alcohol consumption in the PAE group was ~2 drinks/week. Higher PS was observed in the PAE group (p < 0.01). In multivariable modeling, PS was associated with pCRH gene expression (β = 0.006, p < 0.01), while PAE was associated with 11β-HSD2 protein expression (β = 0.56, p < 0.01). A significant alcohol-by-stress interaction was observed with respect to 11β-HSD2 protein expression (p < 0.01). Results indicate that PAE and PS may independently and in combination affect fetal programming of the HPA axis.
Collapse
Affiliation(s)
- Ludmila N. Bakhireva
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Elizabeth Solomon
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA; (E.S.); (E.D.M.)
| | - Melissa H. Roberts
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Xingya Ma
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Rajani Rai
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Alexandria Wiesel
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Sandra W. Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Erin D. Milligan
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA; (E.S.); (E.D.M.)
| |
Collapse
|
9
|
Chen Y, Wang H. The changes in adrenal developmental programming and homeostasis in offspring induced by glucocorticoids exposure during pregnancy. VITAMINS AND HORMONES 2024; 124:463-490. [PMID: 38408809 DOI: 10.1016/bs.vh.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Clinically, synthetic glucocorticoids are often used to treat maternal and fetal related diseases, such as preterm birth and autoimmune diseases. Although its clinical efficacy is positive, it will expose the fetus to exogenous glucocorticoids. Adverse environments during pregnancy (e.g., exogenous glucocorticoids exposure, malnutrition, infection, hypoxia, and stress) can lead to fetal overexposure to endogenous maternal glucocorticoids. Basal glucocorticoids levels in utero are crucial in determining fetal tissue maturation and its postnatal fate. As the synthesis and secretion organ of glucocorticoids, the adrenal development is crucial for the growth and development of the body. Studies have found that glucocorticoids exposure during pregnancy could cause abnormal fetal adrenal development, which could last after birth or even adulthood. As the key organ of fetal-originated adult disease, the adrenal developmental programming has a profound impact on the health of offspring, which can lead to many chronic diseases in adulthood. However, the aberrant adrenal development in offspring caused by glucocorticoids exposure during pregnancy and its intrauterine programming mechanism have not been systematically clarified. Therefore, this review summarizes recent research progress on the short and long-term hazards of aberrant adrenal development induced by glucocorticoids exposure during pregnancy, which is of great significance for the analysis of aberrant adrenal development and clarify the intrauterine origin mechanism of fetal-originated adult disease.
Collapse
Affiliation(s)
- Yawen Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, P.R. China.
| |
Collapse
|
10
|
Xia X, Chen Y, Qu H, Cao J, Wang H. The high-expression programming of SR-B1 mediates adrenal dysfunction in female offspring induced by prenatal caffeine exposure and its cholesterol accumulation mechanism. Food Funct 2024; 15:716-731. [PMID: 38113052 DOI: 10.1039/d3fo03561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
The cholesterol metabolism and homeostasis of adrenal are important for steroidogenesis. Our previous studies found that prenatal caffeine exposure (PCE) can inhibit adrenal steroidogenesis in offspring, but whether the mechanism is related to local imbalance of cholesterol metabolism remains unknown. Here, we found that PCE inhibited adrenal steroidogenesis and increased the expression of cell pyroptosis and inflammatory-related indicators (NLRP3, caspase-1 and IL-1β) in female adult offspring rats, and at the same time, the cholesterol levels in serum and adrenal gland also significantly increased. In vitro, the high level of cholesterol could inhibit adrenal corticosteroid synthesis through pyroptosis and an inflammatory response. It suggested that the low adrenal steroidogenesis in PCE female adult offspring is related to local cholesterol accumulation-mediated pyroptosis and inflammation. Furthermore, dating back to the intrauterine period, PCE increased the serum CORT level in female fetal rats, and increased the expression of the adrenal cholesterol intake gene SR-B1, which persisted after birth and even into adulthood. At the cellular level, silencing SR-B1 could reverse the increase of intracellular cholesterol content caused by high levels of cortisol in NCI-H295R cells. Finally, we confirmed that high concentrations of glucocorticoids increased the expression and H3K14ac level of the promoter region in SR-B1 by upregulating the GR/SREBP1/p300 pathway in vivo and in vitro. In conclusion, we clarified that the high-expression programming of SR-B1 mediates adrenal dysfunction in PCE female offspring and its cholesterol accumulation mechanism, which provided a favorable basis for finding novel targets to prevent and treat fetal-originated diseases.
Collapse
Affiliation(s)
- Xuan Xia
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yawen Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Qu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| | - Jiangang Cao
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| |
Collapse
|
11
|
Wei L, Wang T, Luo M, Zhang S, Lu M, Zhou X, Cheng X, Wang H, Xu D. A "toxic window" study on the hippocampal development of mice offspring exposed to azithromycin at different doses, courses, and time during pregnancy. Chem Biol Interact 2024; 387:110814. [PMID: 37995777 DOI: 10.1016/j.cbi.2023.110814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Azithromycin, one of the new-generation macrolides, is an effective medicine for the treatment of mycoplasma infection during pregnancy. Epidemiological studies have reported adverse pregnancy outcomes with prenatal azithromycin exposure (PAzE). However, the effect of PAzE on fetal hippocampal development is unclear. This study aimed to explore the effects and potential mechanism of PAzE-induced fetal hippocampal development at different doses, courses, and time. METHOD Pregnant mice were administered azithromycin by gavage at different doses (50, 100 or 200 mg/kg.d), different courses (gestational day (GD)15-17 for three consecutive days, or GD17 once a day) and different time (GD10-12, GD15-17). RESULTS Compared with the control group, morphological development damage of the fetal hippocampus was observed in the PAzE group, with a dysbalance in neuronal proliferation and apoptosis, decreased expression of the neuronal-specific marker Snap25, NeuN, PSD95 and Map2, increased expression of the glial-specific marker Iba1, GFAP, and S-100β, and decreased expression of P2ry12. The PAzE-induced hippocampal developmental deficiency varied based on different doses, courses, and time, and the developmental toxicity was most significant in the late pregnancy, high dose, multi-course group (AZHT). The significant reduction of SOX2 and Wnt, which were related to regulation of neural progenitor cells (NPCs) proliferation in PAzE fetus compared with the control group indicated that the SOX2/Wnt signaling may be involved in PAzE-induced hippocampal developmental toxicity. CONCLUSION In this study, PAzE was associated with hippocampal developmental toxicity in a variety of nerve cells. Hippocampal developmental toxicity due to azithromycin was most significant in the late pregnancy, high-dose (equivalent to maximum clinical dose) and multi-course group (AZHT). The findings provide an experimental and theoretical foundation for guiding the sensible use of medications during pregnancy and effectively assessing the risk of fetal hippocampal developmental toxicity.
Collapse
Affiliation(s)
- Liyi Wei
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Tingting Wang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Mingcui Luo
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuai Zhang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Mengxi Lu
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xinli Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xuelei Cheng
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Dan Xu
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
12
|
Chen Z, Xia LP, Shen L, Xu D, Guo Y, Wang H. Glucocorticoids and intrauterine programming of nonalcoholic fatty liver disease. Metabolism 2024; 150:155713. [PMID: 37914025 DOI: 10.1016/j.metabol.2023.155713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
Accumulating epidemiological and experimental evidence indicates that nonalcoholic fatty liver disease (NAFLD) has an intrauterine origin. Fetuses exposed to adverse prenatal environments (e.g., maternal malnutrition and xenobiotic exposure) are more susceptible to developing NAFLD after birth. Glucocorticoids are crucial triggers of the developmental programming of fetal-origin diseases. Adverse intrauterine environments often lead to fetal overexposure to maternally derived glucocorticoids, which can program fetal hepatic lipid metabolism through epigenetic modifications. Adverse intrauterine environments program the offspring's glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axis, which contributes to postnatal catch-up growth and disturbs glucose and lipid metabolism. These glucocorticoid-driven programming alterations increase susceptibility to NAFLD in the offspring. Notably, after delivery, offspring often face an environment distinct from their in utero life. The mismatch between the intrauterine and postnatal environments can serve as a postnatal hit that further disturbs the programmed endocrine axes, accelerating the onset of NAFLD. In this review, we summarize the current epidemiological and experimental evidence demonstrating that NAFLD has an intrauterine origin and discuss the underlying intrauterine programming mechanisms, focusing on the role of overexposure to maternally derived glucocorticoids. We also briefly discuss potential early life interventions that may be beneficial against fetal-originated NAFLD.
Collapse
Affiliation(s)
- Ze Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Li-Ping Xia
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Lang Shen
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Dan Xu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
13
|
Wang T, Zhang S, Luo M, Lu M, Wei L, Zhou X, Wang H, Xu D. Prenatal caffeine exposure induces autism-like behaviors in offspring under a high-fat diet via the gut microbiota-IL-17A-brain axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115797. [PMID: 38070418 DOI: 10.1016/j.ecoenv.2023.115797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/24/2023] [Accepted: 12/05/2023] [Indexed: 01/12/2024]
Abstract
Prenatal caffeine exposure (PCE) is a significant contributor to intrauterine growth retardation (IUGR) in offspring, which has been linked to an increased susceptibility to autism spectrum disorder (ASD) later in life. Additionally, a high-fat diet (HFD) has been shown to exacerbate ASD-like behaviors, but the underlying mechanisms remain unclear. In this study, we first noted in the rat model of IUGR induced by PCE that male PCE offspring exhibited typical ASD-like behaviors post-birth, in contrast to their female counterparts. The female PCE offspring demonstrated only reduced abilities in free exploration and spatial memory. Importantly, both male and female PCE offspring displayed ASD-like behaviors when exposed to HFD. We further observed that PCE + HFD offspring exhibited damaged intestinal mucus barriers and disturbed gut microbiota, resulting in an increased abundance of Escherichia coli (E. coli). The induced differentiation of colonic Th17 cells by E. coli led to an increased secretion of IL-17A, which entered the hippocampus through peripheral circulation and caused synaptic damage in hippocampal neurons, ultimately resulting in ASD development. Our strain transplantation experiment suggested that E. coli-mediated increase of IL-17A may be the core mechanism of ASD with a fetal origin. In conclusion, PCE and HFD are potential risk factors for ASD, and E. coli-mediated IL-17A may play a crucial role in fetal-originated ASD through the gut-brain axis.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Shuai Zhang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Mingcui Luo
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Mengxi Lu
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Liyi Wei
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinli Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Dan Xu
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
14
|
Volqvartz T, Andersen HHB, Pedersen LH, Larsen A. Obesity in pregnancy-Long-term effects on offspring hypothalamic-pituitary-adrenal axis and associations with placental cortisol metabolism: A systematic review. Eur J Neurosci 2023; 58:4393-4422. [PMID: 37974556 DOI: 10.1111/ejn.16184] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Obesity, affecting one in three pregnant women worldwide, is not only a major obstetric risk factor. The resulting low-grade inflammation may have a long-term impact on the offspring's HPA axis through dysregulation of maternal, placental and fetal corticosteroid metabolism, and children born of obese mothers have increased risk of diabetes and cardiovascular disease. The long-term effects of maternal obesity on offspring neurodevelopment are, however, undetermined and could depend on the specific effects on placental and fetal cortisol metabolism. This systematic review evaluates how maternal obesity affects placental cortisol metabolism and the offspring's HPA axis. Pubmed, Embase and Scopus were searched for original studies on maternal BMI, obesity, and cortisol metabolism and transfer. Fifteen studies were included after the screening of 4556 identified records. Studies were small with heterogeneous exposures and outcomes. Two studies found that maternal obesity reduced placental HSD11β2 activity. In one study, umbilical cord blood cortisol levels were affected by maternal BMI. In three studies, an altered cortisol response was consistently seen among offspring in childhood (n = 2) or adulthood (n = 1). Maternal BMI was not associated with placental HSD11β1 or HSD11β2 mRNA expression, or placental HSD11β2 methylation. In conclusion, high maternal BMI is associated with reduced placental HSD11β2 activity and a dampened cortisol level among offspring, but the data is sparse. Further investigations are needed to clarify whether the HPA axis is affected by prenatal factors including maternal obesity and investigate if adverse effects can be ameliorated by optimising the intrauterine environment.
Collapse
Affiliation(s)
- Tabia Volqvartz
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Lars Henning Pedersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Aarhus, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
15
|
Liu L, Wen Y, Ni Q, Chen L, Wang H. Prenatal ethanol exposure and changes in fetal neuroendocrine metabolic programming. Biol Res 2023; 56:61. [PMID: 37978540 PMCID: PMC10656939 DOI: 10.1186/s40659-023-00473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Prenatal ethanol exposure (PEE) (mainly through maternal alcohol consumption) has become widespread. However, studies suggest that it can cause intrauterine growth retardation (IUGR) and multi-organ developmental toxicity in offspring, and susceptibility to various chronic diseases (such as neuropsychiatric diseases, metabolic syndrome, and related diseases) in adults. Through ethanol's direct effects and its indirect effects mediated by maternal-derived glucocorticoids, PEE alters epigenetic modifications and organ developmental programming during fetal development, which damages the offspring health and increases susceptibility to various chronic diseases after birth. Ethanol directly leads to the developmental toxicity of multiple tissues and organs in many ways. Regarding maternal-derived glucocorticoid-mediated IUGR, developmental programming, and susceptibility to multiple conditions after birth, ethanol induces programmed changes in the neuroendocrine axes of offspring, such as the hypothalamus-pituitary-adrenal (HPA) and glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axes. In addition, the differences in ethanol metabolic enzymes, placental glucocorticoid barrier function, and the sensitivity to glucocorticoids in various tissues and organs mediate the severity and sex differences in the developmental toxicity of ethanol exposure during pregnancy. Offspring exposed to ethanol during pregnancy have a "thrifty phenotype" in the fetal period, and show "catch-up growth" in the case of abundant nutrition after birth; when encountering adverse environments, these offspring are more likely to develop diseases. Here, we review the developmental toxicity, functional alterations in multiple organs, and neuroendocrine metabolic programming mechanisms induced by PEE based on our research and that of other investigators. This should provide new perspectives for the effective prevention and treatment of ethanol developmental toxicity and the early prevention of related fetal-originated diseases.
Collapse
Affiliation(s)
- Liang Liu
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yinxian Wen
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Qubo Ni
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
16
|
Wang T, Chen B, Luo M, Xie L, Lu M, Lu X, Zhang S, Wei L, Zhou X, Yao B, Wang H, Xu D. Microbiota-indole 3-propionic acid-brain axis mediates abnormal synaptic pruning of hippocampal microglia and susceptibility to ASD in IUGR offspring. MICROBIOME 2023; 11:245. [PMID: 37932832 PMCID: PMC10629055 DOI: 10.1186/s40168-023-01656-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/23/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) has been associated with intrauterine growth restriction (IUGR), but the underlying mechanisms are unclear. RESULTS We found that the IUGR rat model induced by prenatal caffeine exposure (PCE) showed ASD-like symptoms, accompanied by altered gut microbiota and reduced production of indole 3-propionic acid (IPA), a microbiota-specific metabolite and a ligand of aryl hydrocarbon receptor (AHR). IUGR children also had a reduced serum IPA level consistent with the animal model. We demonstrated that the dysregulated IPA/AHR/NF-κB signaling caused by disturbed gut microbiota mediated the hippocampal microglia hyperactivation and neuronal synapse over-pruning in the PCE-induced IUGR rats. Moreover, postnatal IPA supplementation restored the ASD-like symptoms and the underlying hippocampal lesions in the IUGR rats. CONCLUSIONS This study suggests that the microbiota-IPA-brain axis regulates ASD susceptibility in PCE-induced IUGR offspring, and supplementation of microbiota-derived IPA might be a promising interventional strategy for ASD with a fetal origin. Video Abstract.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Beidi Chen
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Mingcui Luo
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lulu Xie
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengxi Lu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaoqian Lu
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Shuai Zhang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Liyi Wei
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xinli Zhou
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Dan Xu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
17
|
Skinner KT, Palkar AM, Hong AL. Genetics of ABCB1 in Cancer. Cancers (Basel) 2023; 15:4236. [PMID: 37686513 PMCID: PMC10487083 DOI: 10.3390/cancers15174236] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
ABCB1, also known as MDR1, is a gene that encodes P-glycoprotein (P-gp), a membrane-associated ATP-dependent transporter. P-gp is widely expressed in many healthy tissues-in the gastrointestinal tract, liver, kidney, and at the blood-brain barrier. P-gp works to pump xenobiotics such as toxins and drugs out of cells. P-gp is also commonly upregulated across multiple cancer types such as ovarian, breast, and lung. Overexpression of ABCB1 has been linked to the development of chemotherapy resistance across these cancers. In vitro work across a wide range of drug-sensitive and -resistant cancer cell lines has shown that upon treatment with chemotherapeutic agents such as doxorubicin, cisplatin, and paclitaxel, ABCB1 is upregulated. This upregulation is caused in part by a variety of genetic and epigenetic mechanisms. This includes single-nucleotide variants that lead to enhanced P-gp ATPase activity without increasing ABCB1 RNA and protein levels. In this review, we summarize current knowledge of genetic and epigenetic mechanisms leading to ABCB1 upregulation and P-gp-enhanced ATPase activity in the setting of chemotherapy resistance across a variety of cancers.
Collapse
Affiliation(s)
- Katie T. Skinner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (K.T.S.); (A.M.P.)
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Antara M. Palkar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (K.T.S.); (A.M.P.)
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Andrew L. Hong
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (K.T.S.); (A.M.P.)
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Chen K, Lu X, Xu D, Guo Y, Ao Y, Wang H. Prenatal exposure to corn oil, CMC-Na or DMSO affects physical development and multi-organ functions in fetal mice. Reprod Toxicol 2023; 118:108366. [PMID: 36958465 DOI: 10.1016/j.reprotox.2023.108366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023]
Abstract
Corn oil, sodium carboxymethyl cellulose (CMC-Na), and dimethyl sulfoxide (DMSO) are widely used as solvents or suspensions in animal experiments, but the effects of prenatal exposure to them on fetal development have not been reported. In this study, Kunming mice were given a conventional dose of corn oil (9.2g/kg·d), CMC-Na (0.05g/kg·d) or DMSO (0.088g/kg·d) during gestation days 10-18, and the pregnancy outcome, fetal physical development, serum phenotype, and multi-organ function changes were observed. The results showed that corn oil decreased serum triglyceride level in males but increased their serum testosterone and CORT levels, and affected female placenta and female/male multi-organ functions (mainly bone, liver, kidney). CMC-Na increased female/male body lengths and tail lengths, decreased serum glucose and total cholesterol levels in males as well as increased their serum LDL-C/HDL-C ratio and testosterone level, decreased female serum bile acid level, and affected male/female placenta and multi-organ functions (mainly bone, liver, hippocampus). DMSO decreased male body weight and serum glucose level, decreased male/female serum bile acid levels, and affected male/female placenta and multi-organs functions (mainly bone, hippocampus, adrenal gland). In conclusion, prenatal exposure to a conventional dose of corn oil, CMC-Na or DMSO could affect fetal physical development and multi-organ functions, and has the characteristics of "multi-pathway, multi-organ and multi-target". This study provides the experimental basis for the rational selection of solvents or suspensions in pharmacology and toxicology studies. DATA AVAILABILITY: Data will be made available on request.
Collapse
Affiliation(s)
- Kaiqi Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Xiaoqian Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Dan Xu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Ying Ao
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
19
|
Lin S, Ye MY, Fu QY, Pan CL, Liu YJ, Zheng LM, Hong Q, Chen YH. Cholic acid exposure during late pregnancy causes placental dysfunction and fetal growth restriction by reactive oxygen species-mediated activation of placental GCN2/eIF2α pathway. FASEB J 2023; 37:e22820. [PMID: 36801982 DOI: 10.1096/fj.202202126r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/19/2023] [Accepted: 01/30/2023] [Indexed: 02/23/2023]
Abstract
Epidemiological studies suggest that fetal growth restriction (FGR) caused by gestational cholestasis is associated with elevated serum cholic acid (CA). Here, we explore the mechanism by which CA induces FGR. Pregnant mice except controls were orally administered with CA daily from gestational day 13 (GD13) to GD17. Results found that CA exposure decreased fetal weight and crown-rump length, and increased the incidence of FGR in a dose-dependent manner. Furthermore, CA caused placental glucocorticoid (GC) barrier dysfunction via down-regulating the protein but not the mRNA level of placental 11β-Hydroxysteroid dehydrogenase-2 (11β-HSD2). Additionally, CA activated placental GCN2/eIF2α pathway. GCN2iB, an inhibitor of GCN2, significantly inhibited CA-induced down-regulation of 11β-HSD2 protein. We further found that CA caused excessive reactive oxygen species (ROS) production and oxidative stress in mouse placentas and human trophoblasts. NAC significantly rescued CA-induced placental barrier dysfunction by inhibiting activation of GCN2/eIF2α pathway and subsequent down-regulation of 11β-HSD2 protein in placental trophoblasts. Importantly, NAC rescued CA-induced FGR in mice. Overall, our results suggest that CA exposure during late pregnancy induces placental GC barrier dysfunction and subsequent FGR may be via ROS-mediated placental GCN2/eIF2α activation. This study provides valuable insight for understanding the mechanism of cholestasis-induced placental dysfunction and subsequent FGR.
Collapse
Affiliation(s)
- Shuai Lin
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Meng-Ying Ye
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qian-Yun Fu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Chao-Lin Pan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ya-Jie Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li-Ming Zheng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qiang Hong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuan-Hua Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Yu P, Zhou J, Ge C, Fang M, Zhang Y, Wang H. Differential expression of placental 11β-HSD2 induced by high maternal glucocorticoid exposure mediates sex differences in placental and fetal development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 827:154396. [PMID: 35259391 DOI: 10.1016/j.scitotenv.2022.154396] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/20/2022] [Accepted: 03/04/2022] [Indexed: 06/14/2023]
Abstract
A variety of adverse environmental factors during pregnancy cause maternal chronic stress. Caffeine is a common stressor, and its consumption during pregnancy is widespread. Our previous study showed that prenatal caffeine exposure (PCE) increased maternal blood glucocorticoid levels and caused abnormal development of offspring. However, the placental mechanism for fetal development inhibition caused by PCE-induced high maternal glucocorticoid has not been reported. This study investigated the effects of PCE-induced high maternal glucocorticoid level on placental and fetal development by regulating placental 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) expression and its underlying mechanism. First, human placenta and umbilical cord blood samples were collected from women without prenatal use of synthetic glucocorticoids. We found that placental 11β-HSD2 expression was significantly correlated with umbilical cord blood cortisol level and birth weight in male newborns but not in females. Furthermore, we established a rat model of high maternal glucocorticoids induced by PCE (caffeine, 60 mg/kg·d, ig), and found that the expression of 11β-HSD2 in male PCE placenta was decreased and negatively correlated with the maternal/fetal/placental corticosterone levels. Meanwhile, we found abnormal placental structure and nutrient transporter expression. In vitro, BeWo cells were used and confirm that 11β-HSD2 mediated inhibition of placental nutrient transporter expression induced by high levels of glucocorticoid. Finally, combined with the animal and cell experiments, we further confirmed that high maternal glucocorticoid could activate the GR-C/EBPα-Egr1 signaling pathway, leading to decreased expression of 11β-HSD2 in males. However, there was no significant inhibition of placental 11β-HSD2 expression, placental and fetal development in females. In summary, we confirmed that high maternal glucocorticoids could regulate placental 11β-HSD2 expression in a sex-specific manner, leading to differences in placental and fetal development. This study provides the theoretical and experimental basis for analyzing the inhibition of fetoplacental development and its sex difference caused by maternal stress.
Collapse
Affiliation(s)
- Pengxia Yu
- Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Jin Zhou
- Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Caiyun Ge
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Man Fang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, China; Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
21
|
Karthika C, Sureshkumar R, Zehravi M, Akter R, Ali F, Ramproshad S, Mondal B, Tagde P, Ahmed Z, Khan FS, Rahman MH, Cavalu S. Multidrug Resistance of Cancer Cells and the Vital Role of P-Glycoprotein. Life (Basel) 2022; 12:897. [PMID: 35743927 PMCID: PMC9227591 DOI: 10.3390/life12060897] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
P-glycoprotein (P-gp) is a major factor in the multidrug resistance phenotype in cancer cells. P-gp is a protein that regulates the ATP-dependent efflux of a wide range of anticancer medicines and confers resistance. Due to its wide specificity, several attempts have been made to block the action of P-gp to restore the efficacy of anticancer drugs. The major goal has been to create molecules that either compete with anticancer medicines for transport or function as a direct P-gp inhibitor. Despite significant in vitro success, there are presently no drugs available in the clinic that can "block" P-gp-mediated resistance. Toxicity, unfavourable pharmacological interactions, and a variety of pharmacokinetic difficulties might all be the reason for the failure. On the other hand, P-gp has a significant effect in the body. It protects the vital organs from the entry of foreign bodies and other toxic chemicals. Hence, the inhibitors of P-gp should not hinder its action in the normal cells. To develop an effective inhibitor of P-gp, thorough background knowledge is needed in this field. The main aim of this review article was to set forth the merits and demerits of the action of P-gp on cancer cells as well as on normal cells. The influence of P-gp on cancer drug delivery and the contribution of P-gp to activating drug resistance were also mentioned.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Raman Sureshkumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University Alkharj, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Faraat Ali
- Department of Licensing and Enforcement, Laboratory Services, Botswana Medicines Regulatory Authority (BoMRA), Gaborone 999106, Botswana;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Priti Tagde
- Amity Institute of Pharmacy, Amity University, Noida 201303, Uttar Pradesh, India;
| | - Zubair Ahmed
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia;
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Mahala Campus, Community College, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Abha 61413, Saudi Arabia;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|