1
|
Hernandez MH, Cohen JM, Skåra KH, Grindstad TK, Lee Y, Magnus P, Njølstad PR, Andreassen OA, Corfield EC, Havdahl A, Molden E, Furu K, Magnus MC, Hernaez A. Placental efflux transporters and antiseizure or antidepressant medication use impact birth weight in MoBa cohort. iScience 2024; 27:109285. [PMID: 38455980 PMCID: PMC10918264 DOI: 10.1016/j.isci.2024.109285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/29/2023] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Low birth weight raises neonatal risks and lifelong health issues and is linked to maternal medication use during pregnancy. We examined data from the Norwegian Mother, Father, and Child Cohort Study and the Medical Birth Registry of Norway, including 69,828 offspring with genotype data and 81,189 with maternal genotype data. We identified genetic risk variants in placental efflux transporters, calculated genetic scores based on alleles related to transporter activity, and assessed their interaction with prenatal use of antiseizure or antidepressant medication on offspring birth weight. Our study uncovered possible genetic variants in both offspring (rs3740066) and mothers (rs10248420; rs2235015) in placental efflux transporters (MRP2-ABCC2 and MDR1-ABCB1) that modulated the association between prenatal exposure to antiseizure medication and low birth weight in the offspring. Antidepressant exposure was associated with low birth weight, but there were no gene-drug interactions. The interplay between MRP2-ABCC2 and MDR1-ABCB1 variants and antiseizure medication may impact neonatal birth weight.
Collapse
Affiliation(s)
- Marta H. Hernandez
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Blanquerna School of Health Sciences, University Ramon Llull, Barcelona, Spain
| | - Jacqueline M. Cohen
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Chronic Diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Karoline H. Skåra
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Community Medicine and Global Health, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Thea K. Grindstad
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Yunsung Lee
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål R. Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research, NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elizabeth C. Corfield
- Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diakonale Hospital, Oslo, Norway
| | - Alexandra Havdahl
- Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diakonale Hospital, Oslo, Norway
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
- Section for Pharmacology and Pharmaceutical Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Kari Furu
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Chronic Diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Maria C. Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Alvaro Hernaez
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Blanquerna School of Health Sciences, University Ramon Llull, Barcelona, Spain
| |
Collapse
|
2
|
Liu Z, Zhai M, Zhang Q, Yang T, Wan Z, Li J, Liu X, Xu B, Du L, Chan RWS, Zhang L, Yeung WSB, Cheung KW, Chiu PCN, Wang WJ, Lee CL, Gao Y. Resolving the gene expression maps of human first-trimester chorionic villi with spatial transcriptome. Front Cell Dev Biol 2022; 10:1060298. [PMID: 36561369 PMCID: PMC9763897 DOI: 10.3389/fcell.2022.1060298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The placenta is important for fetal development in mammals, and spatial transcriptomic profiling of placenta helps to resolve its structure and function. In this study, we described the landscape of spatial transcriptome of human placental villi obtained from two pregnant women at the first trimester using the modified Stereo-seq method applied for paraformaldehyde (PFA) fixation samples. The PFA fixation of human placenta villi was better than fresh villi embedded in optimum cutting temperature (OCT) compound, since it greatly improved tissue morphology and the specificity of RNA signals. The main cell types in chorionic villi such as syncytiotrophoblasts (SCT), villous cytotrophoblasts (VCT), fibroblasts (FB), and extravillous trophoblasts (EVT) were identified with the spatial transcriptome data, whereas the minor cell types of Hofbauer cells (HB) and endothelial cells (Endo) were spatially located by deconvolution of scRNA-seq data. We demonstrated that the Stereo-seq data of human villi could be used for sophisticated analyses such as spatial cell-communication and regulatory activity. We found that the SCT and VCT exhibited the most ligand-receptor pairs that could increase differentiation of the SCT, and that the spatial localization of specific regulons in different cell types was associated with the pathways related to hormones transport and secretion, regulation of mitotic cell cycle, and nutrient transport pathway in SCT. In EVT, regulatory pathways such as the epithelial to mesenchyme transition, epithelial development and differentiation, and extracellular matrix organization were identified. Finally, viral receptors and drug transporters were identified in villi according to the pathway analysis, which could help to explain the vertical transmission of several infectious diseases and drug metabolism efficacy. Our study provides a valuable resource for further investigation of the placenta development, physiology and pathology in a spatial context.
Collapse
Affiliation(s)
| | | | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Tingyu Yang
- BGI-Shenzhen, Shenzhen, China,Shenzhen Engineering Laboratory for Birth Defects Screening, Shenzhen, China,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Jianlin Li
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaofeng Liu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Bo Xu
- Department of Obstetrics and Gynaecology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Libei Du
- Department of Obstetrics and Gynaecology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Rachel W. S. Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Li Zhang
- Department of Obstetrics and Gynaecology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ka Wang Cheung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wen-Jing Wang
- BGI-Shenzhen, Shenzhen, China,*Correspondence: Wen-Jing Wang, ; Cheuk-Lun Lee, ; Ya Gao,
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China,*Correspondence: Wen-Jing Wang, ; Cheuk-Lun Lee, ; Ya Gao,
| | - Ya Gao
- BGI-Shenzhen, Shenzhen, China,Shenzhen Engineering Laboratory for Birth Defects Screening, Shenzhen, China,*Correspondence: Wen-Jing Wang, ; Cheuk-Lun Lee, ; Ya Gao,
| |
Collapse
|
3
|
Goetzl L, Darbinian N, Merabova N, Devane LC, Ramamoorthy S. Gestational Age Variation in Human Placental Drug Transporters. Front Pharmacol 2022; 13:837694. [PMID: 35462922 PMCID: PMC9019509 DOI: 10.3389/fphar.2022.837694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/24/2022] [Indexed: 12/29/2022] Open
Abstract
Patient and providers’ fear of fetal exposure to medications may lead to discontinuation of treatment, disease relapse, and maternal morbidity. Placental drug transporters play a critical role in fetal exposure through active transport but the majority of data are limited to the 3rd trimester, when the majority of organogenesis has already occurred. Our objective was to define gestational age (GA) dependent changes in protein activity, expression and modifications of five major placental drug transporters: SERT, P-gp, NET, BCRP and MRP3. Apical brush border membrane fractions were prepared from fresh 1st, 2nd and 3rd trimester human placentas collected following elective pregnancy termination or planned cesarean delivery. A structured maternal questionnaire was used to identify maternal drug use and exclude exposed subjects. Changes in placental transporter activity and expression relative to housekeeping proteins were quantified. There was evidence for strong developmental regulation of SERT, NET, P-gp, BCRP and MRP3. P-gp and BCRP decreased with gestation (r = −0.72, p < 0.001 and r = −0.77, p < 0.001, respectively). Total SERT increased with gestation but this increase was due to a decrease in SERT cleavage products across trimesters. Uncleaved SERT increased with GA (r = 0.89, p < 0.001) while cleaved SERT decreased with GA (r = −0.94, p < 0.001). Apical membrane NET overall did not appear to be developmentally regulated (r = −0.08, p = 0.53). Two forms of MRP3 were identified; the 50 kD form did not change across GA; the 160 kD form was steady in the 1st and 2nd trimester and increased in the 3rd trimester (r = 0.24, p = 0.02). The 50 kD form was expressed at higher levels. The observed patterns of SERT, NET P-gp, BCRP and MRP3 expression and activity may be associated with transporter activity or decreased placental permeability in the 1st trimester to transporter specific substrates including commonly used psychoactive medications such as anti-depressants, anti-psychotics, and amphetamines, while transport of nutrients and serotonin is important in the 1st trimester. Overall these observations are consistent with a strong protective effect during organogenesis. 3rd trimester estimates of fetal exposure obtained from cord blood likely significantly overestimate early fetal exposure to these medications at any fixed maternal dose.
Collapse
|
4
|
Lopez Quiñones AJ, Vieira LS, Wang J. Clinical Applications and the Roles of Transporters in Disposition, Tumor Targeting, and Tissue Toxicity of meta-Iodobenzylguanidine (mIBG). Drug Metab Dispos 2022; 50:DMD-MR-2021-000707. [PMID: 35197314 PMCID: PMC9488973 DOI: 10.1124/dmd.121.000707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Transporters on the plasma membrane of tumor cells are promising molecular "Trojan horses" to deliver drugs and imaging agents into cancer cells. Radioiodine-labeled meta-iodobenzylguanidine (mIBG) is used as a diagnostic agent (123I-mIBG) and a targeted radiotherapy (131I-mIBG) for neuroendocrine cancers. mIBG enters cancer cells through the norepinephrine transporter (NET) where the radioactive decay of 131I causes DNA damage, cell death, and tumor necrosis. mIBG is predominantly eliminated unchanged by the kidney. Despite its selective uptake by neuroendocrine tumors, mIBG accumulates in several normal tissues and leads to tissue-specific radiation toxicities. Emerging evidences suggest that the polyspecific organic cation transporters play important roles in systemic disposition and tissue-specific uptake of mIBG. In particular, human organic cation transporter 2 (hOCT2) and toxin extrusion proteins 1 and 2-K (hMATE1/2-K) likely mediate renal secretion of mIBG whereas hOCT1 and hOCT3 may contribute to mIBG uptake into normal tissues such as the liver, salivary glands, and heart. This mini-review focuses on the clinical applications of mIBG in neuroendocrine cancers and the differential roles of NET, OCT and MATE transporters in mIBG disposition, response and toxicity. Understanding the molecular mechanisms governing mIBG transport in cancer and normal cells is a critical step for developing strategies to optimize the efficacy of 131I-mIBG while minimizing toxicity in normal tissues. Significance Statement Radiolabeled mIBG has been used as a diagnostic tool and as radiotherapy for neuroendocrine cancers and other diseases. NET, OCT and MATE transporters play differential roles in mIBG tumor targeting, systemic elimination, and accumulation in normal tissues. The clinical use of mIBG as a radiopharmaceutical in cancer diagnosis and treatment can be further improved by taking a holistic approach considering mIBG transporters in both cancer and normal tissues.
Collapse
Affiliation(s)
| | | | - Joanne Wang
- Dept. of Pharmaceutics, University of Washington, United States
| |
Collapse
|
5
|
Zhang X, Cheng X, Yu Y, Lei B, Yu Y. Insight into the transplacental transport mechanism of methoxylated polybrominated diphenyl ethers using a BeWo cell monolayer model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114836. [PMID: 32454380 DOI: 10.1016/j.envpol.2020.114836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/02/2020] [Accepted: 05/16/2020] [Indexed: 06/11/2023]
Abstract
Methoxylated polybrominated diphenyl ethers (MeO-PBDEs), a type of emerging environmental contaminants, can accumulate through the food chain and eventually enter the human body. For pregnant women, these chemicals may be transplacentally transported to their fetuses, causing early intrauterine exposure. This study was designed to explore the transport process and characteristics of MeO-PBDEs using a BeWo cell monolayer model to simulate the placental barrier effect. Concentration-dependent transplacental transport indicates that the transport of MeO-PBDEs may be dominated by passive diffusion within the studied concentration range. According to the apparent permeability coefficients, MeO-BDE congeners investigated can be classified as poorly transported compounds, with the exception of MeO-BDE28. Time-dependent transplacental transport was observed (R2 = 0.97-0.99), which showed that the intracellular accumulation of MeO-PBDEs followed pseudo-first-order kinetics process. The transport process of MeO-PBDEs in the BeWo cell assay was not found to be sensitive to the pH of 6.5-7.4. An efflux transporter, breast cancer resistance protein, may be involved in the transport process of some MeO-PBDE congeners, and influx transporters, including organic anion transporters and organic cation transporters, may also be involved in the transport process. Although the present results indicated the possible transplacental transport mechanism, more molecular biological studies should be conducted to advance the understanding of the transport mechanisms.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Xiaomeng Cheng
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Yuling Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Bingli Lei
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Yingxin Yu
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, PR China; Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
6
|
Arumugasaamy N, Gudelsky A, Hurley-Novatny A, Kim PC, Fisher JP. Model Placental Barrier Phenotypic Response to Fluoxetine and Sertraline: A Comparative Study. Adv Healthc Mater 2019; 8:e1900476. [PMID: 31407872 PMCID: PMC6752965 DOI: 10.1002/adhm.201900476] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/10/2019] [Indexed: 12/20/2022]
Abstract
Medications taken during pregnancy may significantly impact fetal development, yet there are few studies that rigorously assess medication safety due to ethical concerns. Selective serotonin reuptake inhibitors (SSRIs) are a class of drug increasingly being prescribed for depression, yet multiple studies have shown that taking SSRIs during pregnancy can lead to preterm birth and potential health concerns for the baby. Therefore, a biomimetic placental barrier model is utilized herein to assess transport profiles and phenotypic effects resulting from SSRI exposure, comparing fluoxetine and sertraline. Results show that the placental barrier quickly uptakes drug from the maternal side, but slowly releases on the fetal side. Phenotypically, there is a dose-dependent change in cell adhesion molecule (CAM) and transforming growth factor beta (TGFβ) secretions, markers of cell adhesion and angiogenesis. Both drugs impact CAM secretions, whereas sertraline alone impacts TGFβ secretions. When evaluating cell type, it becomes clear that endothelial cells, not trophoblast, are the main cell type involved in these phenotypic changes. Overall, these findings further the understanding of SSRI transplacental transport and drug-induced effects on the placental barrier.
Collapse
Affiliation(s)
- Navein Arumugasaamy
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
| | - Alana Gudelsky
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
| | - Amelia Hurley-Novatny
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
| | - Peter C.W. Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
- Department of Surgery, The George Washington University, Washington, D.C. 20037
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
| |
Collapse
|
7
|
Kott J, Brummelte S. Trick or treat? Evaluating contributing factors and sex-differences for developmental effects of maternal depression and its treatment. Horm Behav 2019; 111:31-45. [PMID: 30658054 DOI: 10.1016/j.yhbeh.2019.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/14/2018] [Accepted: 01/11/2019] [Indexed: 11/29/2022]
Abstract
Maternal depression and treatment with selective serotonin reuptake inhibitors (SSRIs), the most common form of pharmaceutical intervention, can both have an impact on infant development. As such, it is difficult for healthcare providers to recommend a course of treatment to expectant mothers suffering from depression, or to women on antidepressant medication prior to pregnancy. This review will discuss the existing research on the developmental impacts of maternal depression and its treatment with SSRIs, with a particular focus on contributing factors that complicate our attempt to disentangle the consequences of maternal depression and its treatment such as the timing or severity of the depression. We will explore avenues for translational animal models to help address the question of "Trick or Treat", i.e.: which is worse for offspring development: exposure to maternal depression, or the SSRI treatment? Further, we will explore sex-dependent outcomes for the offspring in human and animal studies as male and female offspring may react differently to the presence of maternal depression or antidepressant treatment. Without more clinical and preclinical data, it remains difficult for women to make an informed decision regarding their depression treatment before, during, and after their pregnancy.
Collapse
Affiliation(s)
- Jennifer Kott
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | | |
Collapse
|
8
|
Developmental outcomes after gestational antidepressant treatment with sertraline and its discontinuation in an animal model of maternal depression. Behav Brain Res 2019; 366:1-12. [PMID: 30836156 DOI: 10.1016/j.bbr.2019.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/04/2019] [Accepted: 03/01/2019] [Indexed: 01/21/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are commonly prescribed to women before or during pregnancy to manage their depressive symptoms. However, there is still little knowledge regarding the long-term development effects of SSRI exposure for the fetus or the effects of discontinuing SSRI treatment during pregnancy. This study utilized a translational animal model of maternal depression (based on giving high levels of corticosterone (CORT, 40 mg/kg, s.c.) or vehicle (Oil) for 21 days prior to conception) to investigate the effects of sertraline (a frequently prescribed SSRI; 20 mg/kg p.o., treatment started ∼7 days prior to conception) and its discontinuation during pregnancy (on gestational day 16) compared to vehicle (water) treatment on the development of the offspring. Our results revealed that both corticosterone exposure prior to pregnancy and sertraline administration and its discontinuation during gestation had sex-specific effects on behavior in the adult offspring. In particular, pre-conceptional maternal corticosterone treatment impacted the stress response, anxiety-like behavior and cognitive performance in adult female offspring, while gestational SSRI exposure and its discontinuation compared to full-term exposure affected impulsivity in females, and exploratory behavior in males. More research is needed on the effects of exposure to antidepressant medication and its discontinuation compared to depression during pregnancy and how each impacts development to better help women make informed decisions about their medication use during pregnancy.
Collapse
|
9
|
Al-Enazy S, Ali S, Albekairi N, El-Tawil M, Rytting E. Placental control of drug delivery. Adv Drug Deliv Rev 2017; 116:63-72. [PMID: 27527665 DOI: 10.1016/j.addr.2016.08.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/25/2016] [Accepted: 08/04/2016] [Indexed: 01/04/2023]
Abstract
The placenta serves as the interface between the maternal and fetal circulations and regulates the transfer of oxygen, nutrients, and waste products. When exogenous substances are present in the maternal bloodstream-whether from environmental contact, occupational exposure, medication, or drug abuse-the extent to which this exposure affects the fetus is determined by transport and biotransformation processes in the placental barrier. Advances in drug delivery strategies are expected to improve the treatment of maternal and fetal diseases encountered during pregnancy.
Collapse
|
10
|
Bank AM, Stowe ZN, Newport DJ, Ritchie JC, Pennell PB. Placental passage of antiepileptic drugs at delivery and neonatal outcomes. Epilepsia 2017; 58:e82-e86. [PMID: 28387929 DOI: 10.1111/epi.13733] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2017] [Indexed: 11/29/2022]
Abstract
Children of women treated with antiepileptic drugs (AEDs) are at increased risk of adverse outcomes detectable in the neonatal period, which may be associated with the amount of AEDs in the fetal circulation. Placental passage of AEDs can be measured by calculating the ratio of umbilical cord to maternal AED concentrations collected at delivery. The aims of this study were to determine the umbilical cord concentrations and umbilical-to-maternal ratios for AEDs, and whether higher cord concentrations are associated with increased risk of neonatal complications. AED cord and maternal blood concentrations from 70 mother-newborn dyads and neonatal complications were recorded. Logistic regressions were performed to determine the association between AED concentrations and complications. Mean umbilical-to-maternal ratios for total concentrations ranged from 0.79 for carbamazepine to 1.20 for valproic acid, and mean umbilical-to-maternal ratios for free concentrations ranged from 0.86 for valproic acid to 1.42 for carbamazepine, indicating complete placental passage. Neither umbilical cord concentrations nor umbilical-to-maternal ratios were associated with adverse neonatal outcomes. Additional investigations are warranted to delineate the relationship between quantified fetal AED exposure and neonatal complications.
Collapse
Affiliation(s)
- Anna M Bank
- Brigham and Women's Hospital, Boston, Massachusetts, U.S.A.,Massachusetts General Hospital, Boston, Massachusetts, U.S.A.,Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Zachary N Stowe
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, U.S.A.,Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, U.S.A
| | - D Jeffrey Newport
- University of Miami Miller School of Medicine, Miami, Florida, U.S.A
| | - James C Ritchie
- Emory University School of Medicine, Atlanta, Georgia, U.S.A
| | - Page B Pennell
- Brigham and Women's Hospital, Boston, Massachusetts, U.S.A.,Harvard Medical School, Boston, Massachusetts, U.S.A
| |
Collapse
|
11
|
Placental transfer of antidepressant medications: implications for postnatal adaptation syndrome. Clin Pharmacokinet 2015; 54:359-70. [PMID: 25711391 DOI: 10.1007/s40262-014-0233-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Seven to thirteen percent of women are either prescribed or taking (depending on the study) an antidepressant during pregnancy. Because antidepressants freely cross into the intrauterine environment, we aim to summarize the current findings on placental transfer of antidepressants. Although generally low risk, antidepressants have been associated with postnatal adaptation syndrome (PNAS). Specifically, we explore whether the antidepressants most closely associated with PNAS (paroxetine, fluoxetine, venlafaxine) cross the placenta to a greater extent than other antidepressants. We review research on antidepressants in the context of placental anatomy, placental transport mechanisms, placental metabolism, pharmacokinetics, as well as non-placental maternal and fetal factors. This provides insight into the complexity involved in understanding how placental transfer of antidepressants may relate to adverse perinatal outcomes. Ultimately, from this data there is no pattern in which PNAS is related to placental transfer of antidepressant medications. In general, there is large interindividual variability for each type of antidepressant. To make the most clinically informed decisions about the use of antidepressants in pregnancy, studies that link maternal, placental and fetal genetic polymorphisms, placental transfer rates and infant outcomes are needed.
Collapse
|
12
|
Nigam SK, Bush KT, Martovetsky G, Ahn SY, Liu HC, Richard E, Bhatnagar V, Wu W. The organic anion transporter (OAT) family: a systems biology perspective. Physiol Rev 2015; 95:83-123. [PMID: 25540139 PMCID: PMC4281586 DOI: 10.1152/physrev.00025.2013] [Citation(s) in RCA: 315] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The organic anion transporter (OAT) subfamily, which constitutes roughly half of the SLC22 (solute carrier 22) transporter family, has received a great deal of attention because of its role in handling of common drugs (antibiotics, antivirals, diuretics, nonsteroidal anti-inflammatory drugs), toxins (mercury, aristolochic acid), and nutrients (vitamins, flavonoids). Oats are expressed in many tissues, including kidney, liver, choroid plexus, olfactory mucosa, brain, retina, and placenta. Recent metabolomics and microarray data from Oat1 [Slc22a6, originally identified as NKT (novel kidney transporter)] and Oat3 (Slc22a8) knockouts, as well as systems biology studies, indicate that this pathway plays a central role in the metabolism and handling of gut microbiome metabolites as well as putative uremic toxins of kidney disease. Nuclear receptors and other transcription factors, such as Hnf4α and Hnf1α, appear to regulate the expression of certain Oats in conjunction with phase I and phase II drug metabolizing enzymes. Some Oats have a strong selectivity for particular signaling molecules, including cyclic nucleotides, conjugated sex steroids, odorants, uric acid, and prostaglandins and/or their metabolites. According to the "Remote Sensing and Signaling Hypothesis," which is elaborated in detail here, Oats may function in remote interorgan communication by regulating levels of signaling molecules and key metabolites in tissues and body fluids. Oats may also play a major role in interorganismal communication (via movement of small molecules across the intestine, placental barrier, into breast milk, and volatile odorants into the urine). The role of various Oat isoforms in systems physiology appears quite complex, and their ramifications are discussed in the context of remote sensing and signaling.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Kevin T Bush
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Gleb Martovetsky
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Sun-Young Ahn
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Henry C Liu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Erin Richard
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Vibha Bhatnagar
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Wei Wu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
13
|
Julius JM, Tindall A, Moise KJ, Refuerzo JS, Berens PD, Smith JA. Evaluation of the maternal-fetal transfer of granisetron in an ex vivo placenta perfusion model. Reprod Toxicol 2014; 49:43-7. [PMID: 25019977 DOI: 10.1016/j.reprotox.2014.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 11/26/2022]
Abstract
The objective of this study was to estimate maternal-fetal transplacental passage of granisetron in an ex vivo placental perfusion model. Term human placentas (N=8) were collected immediately after delivery. A single cotyledon from each placenta was perfused granisetron concentration to mimic systemic maternal peak plasma concentrations following either IV (50ng/mL) or transdermal administration (5ng/mL). To assess drug transfer and accumulation, samples were collected from maternal and fetal compartments. In the 50ng/mL open model, the mean transport fraction was 0.21±0.08 with clearance index of 0.53±0.66. Fetal peak concentrations achieved was 5.6±6.6ng/mL with mean accumulation of 5.35±6.4ng/mL. No drug was detected in the fetal compartment with the 5ng/mL models. Transplacental passage of granisetron was inconsistent at the 50ng/mL concentration that achieved with IV dosing. However, there consistently was no detectable passage in all the placentas evaluated of the granisetron at 5ng/mL concentration that would be achieved after transdermal patch administration.
Collapse
Affiliation(s)
- Justin M Julius
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew Tindall
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kenneth J Moise
- Department of Gynecology, Obstetrics & Reproductive Sciences, The University of Texas Health Sciences Center at Houston Medical School, Houston, TX, United States
| | - Jerrie S Refuerzo
- Department of Gynecology, Obstetrics & Reproductive Sciences, The University of Texas Health Sciences Center at Houston Medical School, Houston, TX, United States
| | - Pamela D Berens
- Department of Gynecology, Obstetrics & Reproductive Sciences, The University of Texas Health Sciences Center at Houston Medical School, Houston, TX, United States
| | - Judith A Smith
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Gynecology, Obstetrics & Reproductive Sciences, The University of Texas Health Sciences Center at Houston Medical School, Houston, TX, United States.
| |
Collapse
|
14
|
Zheng S, Easterling TR, Hays K, Umans JG, Miodovnik M, Clark S, Calamia JC, Thummel KE, Shen DD, Davis CL, Hebert MF. Tacrolimus placental transfer at delivery and neonatal exposure through breast milk. Br J Clin Pharmacol 2014; 76:988-96. [PMID: 23528073 DOI: 10.1111/bcp.12122] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/10/2013] [Indexed: 01/16/2023] Open
Abstract
AIM(S) The current investigation aims to provide new insights into fetal exposure to tacrolimus in utero by evaluating maternal and umbilical cord blood (venous and arterial), plasma and unbound concentrations at delivery. This study also presents a case report of tacrolimus excretion via breast milk. METHODS Maternal and umbilical cord (venous and arterial) samples were obtained at delivery from eight solid organ allograft recipients to measure tacrolimus and metabolite bound and unbound concentrations in blood and plasma. Tacrolimus pharmacokinetics in breast milk were assessed in one subject. RESULTS Mean (±SD) tacrolimus concentrations at the time of delivery in umbilical cord venous blood (6.6 ± 1.8 ng ml(-1)) were 71 ± 18% (range 45-99%) of maternal concentrations (9.0 ± 3.4 ng ml(-1)). The mean umbilical cord venous plasma (0.09 ± 0.04 ng ml(-1)) and unbound drug concentrations (0.003 ± 0.001 ng ml(-1)) were approximately one fifth of the respective maternal concentrations. Arterial umbilical cord blood concentrations of tacrolimus were 100 ± 12% of umbilical venous concentrations. In addition, infant exposure to tacrolimus through the breast milk was less than 0.3% of the mother's weight-adjusted dose. CONCLUSIONS Differences between maternal and umbilical cord tacrolimus concentrations may be explained in part by placental P-gp function, greater red blood cell partitioning and higher haematocrit levels in venous cord blood. The neonatal drug exposure to tacrolimus via breast milk is very low and likely does not represent a health risk to the breastfeeding infant.
Collapse
Affiliation(s)
- Songmao Zheng
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Pharmacotherapy for mood disorders in pregnancy: a review of pharmacokinetic changes and clinical recommendations for therapeutic drug monitoring. J Clin Psychopharmacol 2014; 34:244-55. [PMID: 24525634 PMCID: PMC4105343 DOI: 10.1097/jcp.0000000000000087] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pharmacotherapy for mood disorders during pregnancy is often complicated by pregnancy-related pharmacokinetic changes and the need for dose adjustments. The objectives of this review are to summarize the evidence for change in perinatal pharmacokinetics of commonly used pharmacotherapies for mood disorders, discuss the implications for clinical and therapeutic drug monitoring (TDM), and make clinical recommendations. METHODS The English-language literature indexed on MEDLINE/PubMed was searched for original observational studies (controlled and uncontrolled, prospective and retrospective), case reports, and case series that evaluated or described pharmacokinetic changes or TDM during pregnancy or the postpartum period. RESULTS Pregnancy-associated changes in absorption, distribution, metabolism, and elimination may result in lowered psychotropic drug levels and possible treatment effects, particularly in late pregnancy. Mechanisms include changes in both phase 1 hepatic cytochrome P450 and phase 2 uridine diphosphate glucuronosyltransferase enzyme activities, changes in hepatic and renal blood flow, and glomerular filtration rate. Therapeutic drug monitoring, in combination with clinical monitoring, is indicated for tricyclic antidepressants and mood stabilizers during the perinatal period. CONCLUSIONS Substantial pharmacokinetic changes can occur during pregnancy in a number of commonly used antidepressants and mood stabilizers. Dose increases may be indicated for antidepressants including citalopram, clomipramine, imipramine, fluoxetine, fluvoxamine, nortriptyline, paroxetine, and sertraline, especially late in pregnancy. Antenatal dose increases may also be needed for lithium, lamotrigine, and valproic acid because of perinatal changes in metabolism. Close clinical monitoring of perinatal mood disorders and TDM of tricyclic antidepressants and mood stabilizers are recommended.
Collapse
|
16
|
Bourke CH, Stowe ZN, Owens MJ. Prenatal antidepressant exposure: clinical and preclinical findings. Pharmacol Rev 2014; 66:435-65. [PMID: 24567054 DOI: 10.1124/pr.111.005207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pharmacological treatment of any maternal illness during pregnancy warrants consideration of the consequences of the illness and/or medication for both the mother and unborn child. In the case of major depressive disorder, which affects up to 10-20% of pregnant women, the deleterious effects of untreated depression on the offspring can be profound and long lasting. Progress has been made in our understanding of the mechanism(s) of action of antidepressants, fetal exposure to these medications, and serotonin's role in development. New technologies and careful study designs have enabled the accurate sampling of maternal serum, breast milk, umbilical cord serum, and infant serum psychotropic medication concentrations to characterize the magnitude of placental transfer and exposure through human breast milk. Despite this progress, the extant clinical literature is largely composed of case series, population-based patient registry data that are reliant on nonobjective means and retrospective recall to determine both medication and maternal depression exposure, and limited inclusion of suitable control groups for maternal depression. Conclusions drawn from such studies often fail to incorporate embryology/neurotransmitter ontogeny, appropriate gestational windows, or a critical discussion of statistically versus clinically significant. Similarly, preclinical studies have predominantly relied on dosing models, leading to exposures that may not be clinically relevant. The elucidation of a defined teratological effect or mechanism, if any, has yet to be conclusively demonstrated. The extant literature indicates that, in many cases, the benefits of antidepressant use during pregnancy for a depressed pregnant woman may outweigh potential risks.
Collapse
|
17
|
Huuskonen P, Myllynen P, Storvik M, Pasanen M. The effects of aflatoxin B1 on transporters and steroid metabolizing enzymes in JEG-3 cells. Toxicol Lett 2013; 218:200-6. [PMID: 23402939 DOI: 10.1016/j.toxlet.2013.01.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 01/17/2013] [Accepted: 01/20/2013] [Indexed: 01/03/2023]
Abstract
Effects of 96 h aflatoxin B1 (AFB1) exposure at concentrations from 0.2 μM to 6 μM on the mRNA and protein expression levels of the following transporters ABCB1/B4, ABCC1, ABCC2, ABCG2, OAT4 and the mRNA expression of steroid-metabolizing enzymes CYP1A1, CYP19A1, HSD3B1 and HSD17B1, and conjugating enzyme family UGT1A were evaluated in trophoblastic JEG-3 cells. Statistically significant dose-dependent five-fold increases in the expression levels with ABCC2 and OAT4 were recorded at 2 and 6μM AFB1. Protein expression of ABCG2 was decreased dose-dependently with 0.2-6 μM AFB1. With the other transporters, only a trend of increased expression was observed. Analogously, a three-fold increase in the expressions of CYP19A1, HSD3B1, HSD17B1 and UGT1A-family were observed at 0.3 μM AFB1. When an inhibitor of CYP19A1, finrozole, was dosed simultaneously with AFB1, no increases in the transcripts of transporters or steroid hydroxylases or CYP19A1 were observed. This delayed increase in the expression levels - only after 96h incubations - may indicate that the response is due to a secondary metabolite of AFB1 or other secondary controlling cascades rather than the parent compound itself. In conclusion, AFB1 affected the placental steroid synthesizing, metabolizing and conjugating enzymes as well as the expression levels of several transporter proteins in JEG-3 cells. These alterations may lead to anomalies in the foetoplacental hormonal homeostasis.
Collapse
Affiliation(s)
- Pasi Huuskonen
- University of Eastern Finland, Faculty of Health Sciences, School of Pharmacy, FI-70211 Kuopio, Finland.
| | | | | | | |
Collapse
|
18
|
Desai G, Babu GN, Chandra PS. Unplanned pregnancies leading to psychotropic exposure in women with mental illness - Findings from a perinatal psychiatry clinic. Indian J Psychiatry 2012; 54:59-63. [PMID: 22556440 PMCID: PMC3339222 DOI: 10.4103/0019-5545.94649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE The aims of this study were (a) to describe the sociodemographic and clinical profile of women with unplanned pregnancies and consequent exposure to psychotropic drugs, (b) to describe the nature and timing of psychotropic exposure during pregnancy among these women, and (c) to examine the outcome of decisions related to pregnancy following consultation at a perinatal psychiatric service. MATERIALS AND METHODS WOMEN ATTENDING THE PERINATAL PSYCHIATRY SERVICES REFERRED FOR ACCIDENTAL EXPOSURE TO PSYCHOTROPICS WERE ASSESSED BY STRUCTURED INTERVIEWS FOR THE FOLLOWING DETAILS: sociodemographic details, clinical details, psychotropic drug use, advice given in the clinic, and outcome related to this advice. RESULTS Fifty-three women were referred for counseling related to unplanned pregnancies and consequential psychotropic exposure. Forty-two women (79%) sought consultation in the first trimester. More than a third of the women, 19 (36%), were taking more than one psychotropic medication during the first consultation. Only 11 (20%) women had received any form of prepregnancy counseling prior to becoming pregnant. Of the 37 women who came for follow-up in the clinic, 35 (94%) of them continued the pregnancy. CONCLUSIONS Unplanned pregnancies in women with mental illness are common and result in exposure to multiple psychotropic medications during the first trimester. Majority of women did not report of having prepregnancy counseling and which needs to be an integral part of treatment and education.
Collapse
Affiliation(s)
- Geetha Desai
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India.
| | | | | |
Collapse
|
19
|
Clinical therapeutics in pregnancy. J Biomed Biotechnol 2011; 2011:783528. [PMID: 21785566 PMCID: PMC3139199 DOI: 10.1155/2011/783528] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 05/03/2011] [Indexed: 12/14/2022] Open
Abstract
Most drugs are not tested for use during pregnancy, consequently, labeling, which may include information about fetal safety, includes nothing about dosing, efficacy, or maternal safety. Yet these are concerns of health care providers considering treatment of disease during pregnancy. Therefore, the practitioner treats the pregnant woman with the same dose recommended for use in adults (typically men) or may decide not to treat the disease at all. However, is the choice of not treating a woman during pregnancy better than dealing with the challenges which accompany treatment? This paper, which summarizes metabolic and physiologic changes induced by pregnancy, illustrates that standard adult dosing is likely to be incorrect during pregnancy.
Collapse
|
20
|
Sit D, Perel JM, Wisniewski SR, Helsel JC, Luther JF, Wisner KL. Mother-infant antidepressant concentrations, maternal depression, and perinatal events. J Clin Psychiatry 2011; 72:994-1001. [PMID: 21824458 PMCID: PMC3158589 DOI: 10.4088/jcp.10m06461] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 10/26/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The authors explored the relationship of cord-maternal antidepressant concentration ratios and maternal depression with perinatal events and preterm birth. METHOD The investigators examined 21 mother-infant pairs that had antidepressant exposure during pregnancy. The antidepressants included serotonin reuptake inhibitors (SRIs) and nortriptyline (a norepinephrine inhibitor and mild SRI). The mothers were evaluated with the Structured Clinical Interview for DSM-IV. Depression ratings were repeated at 20, 30, and 36 weeks' pregnancy. At delivery, investigators assessed cord and maternal antidepressant concentrations, neonatal outcomes on the Peripartum Events Scale (PES), and gestational weeks at birth. The investigators performed this study at the Women's Behavioral HealthCARE Program, Western Psychiatric Institute and Clinic, University of Pittsburgh Medical Center, Pennsylvania, from April 2003 until September 2006. RESULTS Mean ± SD cord-to-maternal concentration ratios were 0.52 ± 0.35 (range, 0.00-1.64) for the parent drug and 0.54 ± 0.17 (range, 0.28-0.79) for the metabolite. Nine of 21 mothers (43%) had a major depressive episode. From examining the maximum depression ratings, the mean ± SD Structured Interview Guide for the Hamilton Depression Rating Scale, Atypical Depression Symptoms Version score was 16.0 ± 7.6. One third (7/21) of infants had at least 1 perinatal event (PES ≥ 1). The frequency of deliveries complicated by any perinatal event was similar in depressed and nondepressed mothers. There was no significant association between perinatal events and cord-to-maternal antidepressant concentration ratios or maternal depression levels. Exposure to short half-life antidepressants compared to fluoxetine resulted in more perinatal events (7/16 = 44% vs 0/5 = 0%; P = .06). Fourteen percent (3/21) of infants were preterm. Preterm birth was not associated with cord-to-maternal metabolite concentration ratios, depression levels, or exposure to fluoxetine. CONCLUSIONS Antidepressant-exposed infants experienced a limited number of transient perinatal events. No association between cord-maternal concentration ratios or maternal depression and perinatal events could be identified. Contrary to other reports, we detected no increased risk for perinatal events with fluoxetine therapy compared to the short half-life antidepressants. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00279370.
Collapse
Affiliation(s)
- Dorothy Sit
- Women's Behavioral HealthCARE, Western Psychiatric Institute and Clinic, University of Pittsburgh, 3811 O'Hara St, Oxford 410, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Pregnancy neither protects women from susceptibility to illness nor generally decreases drug dosage requirements of preexisting pharmacotherapy. Thus, a major concern of drug use during pregnancy is maintenance of maternal health while minimizing fetal drug exposure. Important assumptions in reproductive medicine are that fetal exposure to any medication can present risks in pregnancy--because not all risks are known, as little exposure as possible is desirable.
Collapse
|
22
|
McCracken JT, Aman MG, McDougle CJ, Tierney E, Shiraga S, Whelan F, Arnold LE, Posey D, Ritz L, Vitiello B, Scahill L. Possible influence of variant of the P-glycoprotein gene (MDR1/ABCB1) on clinical response to guanfacine in children with pervasive developmental disorders and hyperactivity. J Child Adolesc Psychopharmacol 2010; 20:1-5. [PMID: 20166790 PMCID: PMC2835389 DOI: 10.1089/cap.2009.0059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Guanfacine has been shown to reduce hyperactive behaviors in children with attention-deficit/hyperactivity disorder (ADHD) and possibly in children with pervasive developmental disorder (PDD) and hyperactivity. The aim of this exploratory study was to examine whether gene variants encoding the multidrug resistance protein (MDR1 or ABCB1) , a drug transporter at the blood-brain barrier, are associated with variability in the efficacy of guanfacine in children with PDD and hyperactivity. METHODS Children with PDD who participated in an 8-week open-label trial of guanfacine were genotyped for the C3435T single-nucleotide polymorphism (SNP) variant of the MDR1 gene, a variant reported to alter function of the transporter. The decrease from baseline to 8 weeks in parent-rated Aberrant Behavior Checklist (ABC) hyperactivity and Swanson, Nolan, and Pelham (SNAP) scores were analyzed by MDR1 genotype. Response was compared between subjects homozygous for the minor allele T of the C34535T MDR1 variant (T/T) versus other genotypes (C/T and C/C). RESULTS Disruptive behavior decreased during guanfacine treatment as assessed by several end points in the 25 enrolled children (23 boys and 2 girls). Genotype data were available from 22 children. Subjects with either C/T or C/C (n = 16) genotypes showed a three-fold greater improvement than T/T MDR1 C3435T genotype (n = 6) (mean decrease of 15.1 +/- 12.6, or 50.7% from baseline, versus 4.5 +/- 5.1, or 15.6% from baseline) in parent-rated ABC Hyperactivity scores over 8 weeks (p = 0.03). Parent-rated ADHD SNAP scores also differed by genotype (p = 0.05). CONCLUSIONS Gene variants in MDR1 may influence guanfacine response on hyperactive-impulsive behaviors via altered membrane transport. If replicated in larger samples, additional studies would be important to clarify the mechanisms underlying this effect and to determine its clinical significance. 2.
Collapse
Affiliation(s)
- James T McCracken
- Research Units on Pediatric Psychopharmacology Autism Network, University of California at Los Angeles, Los Angeles, CA 90024-1759, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Partanen HA, El-Nezami HS, Leppänen JM, Myllynen PK, Woodhouse HJ, Vähäkangas KH. Aflatoxin B1 Transfer and Metabolism in Human Placenta. Toxicol Sci 2009; 113:216-25. [DOI: 10.1093/toxsci/kfp257] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
24
|
Vähäkangas K, Myllynen P. Drug transporters in the human blood-placental barrier. Br J Pharmacol 2009; 158:665-78. [PMID: 19788499 DOI: 10.1111/j.1476-5381.2009.00336.x] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Studies on the increasing number of transporters found in the placental barrier are gaining momentum, because of their tissue-specific expression, significance in physiology and disease, and the possible utilization of the emerging knowledge in pharmacology. In the placenta, both syncytiotrophoblast and fetal capillary endothelium express transporters. Fetal exposure is determined by the net effect of combination of transporters, their nature and localization in relation to placental cells and their substrate specificity. Although the significance of placental transporters on human fetal drug exposure is almost an unstudied field so far, their potential use to design drugs that do not cross the placenta is already being pursued. It is thus of interest to review the existing knowledge of human placental transporters. Transporters in all groups which take part in drug transport are found in human placenta. Especially, ATP-binding cassette transporters ABCG2/breast cancer resistance protein, ABCB1/P-glycoprotein and ABCC2/MRP2 are all expressed at the apical surface of syncytiotrophoblast facing maternal blood and are putatively important protective proteins both for placental tissue and the fetus, because they are efflux transporters and their substrates include many drugs and also environmental chemicals. Such protective effect has been shown in animals, but these results cannot be directly extrapolated to humans due to interspecies differences in placental structure and function. Experimental models utilizing human placental tissue, especially human placental perfusion, offer valuable possibilities, which have been insufficiently studied so far.
Collapse
Affiliation(s)
- Kirsi Vähäkangas
- Department of Pharmacology and Toxicology, University of Kuopio, Kuopio, Finland.
| | | |
Collapse
|
25
|
Rollason V, Samer C, Piguet V, Dayer P, Desmeules J. Pharmacogenetics of analgesics: toward the individualization of prescription. Pharmacogenomics 2008; 9:905-33. [DOI: 10.2217/14622416.9.7.905] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The use of analgesics is based on the empiric administration of a given drug with clinical monitoring for efficacy and toxicity. However, individual responses to drugs are influenced by a combination of pharmacokinetic and pharmacodynamic factors that can sometimes be regulated by genetic factors. Whereas polymorphic drug-metabolizing enzymes and drug transporters may affect the pharmacokinetics of drugs, polymorphic drug targets and disease-related pathways may influence the pharmacodynamic action of drugs. After a usual dose, variations in drug toxicity and inefficacy can be observed depending on the polymorphism, the analgesic considered and the presence or absence of active metabolites. For opioids, the most studied being morphine, mutations in the ABCB1 gene, coding for P-glycoprotein (P-gp), and in the µ-opioid receptor reduce morphine potency. Cytochrome P450 (CYP) 2D6 mutations influence the analgesic effect of codeine and tramadol, and polymorphism of CYP2C9 is potentially linked to an increase in nonsteroidal anti-inflammatory drug-induced adverse events. Furthermore, drug interactions can mimic genetic deficiency and contribute to the variability in response to analgesics. This review summarizes the available data on the pharmacokinetic and pharmacodynamic consequences of known polymorphisms of drug-metabolizing enzymes, drug transporters, drug targets and other nonopioid biological systems on central and peripheral analgesics.
Collapse
Affiliation(s)
- Victoria Rollason
- Geneva University Hospital, University of Geneva, Clinical Pharmacology & Toxicology, Micheli-du-Crest Street 24, 1211 Geneva 14, Switzerland
| | - Caroline Samer
- Geneva University Hospital, University of Geneva, Clinical Pharmacology & Toxicology, Micheli-du-Crest Street 24, 1211 Geneva 14, Switzerland
| | - Valerie Piguet
- Geneva University Hospital, University of Geneva, Clinical Pharmacology & Toxicology, Micheli-du-Crest Street 24, 1211 Geneva 14, Switzerland
| | - Pierre Dayer
- Geneva University Hospital, University of Geneva, Clinical Pharmacology & Toxicology, Micheli-du-Crest Street 24, 1211 Geneva 14, Switzerland
| | - Jules Desmeules
- Geneva University Hospital, University of Geneva, Clinical Pharmacology & Toxicology, Micheli-du-Crest Street 24, 1211 Geneva 14, Switzerland
| |
Collapse
|