1
|
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
|
2
|
Hourtovenko C, Sreetharan S, Tharmalingam S, Tai TC. Impact of Ionizing Radiation Exposure on Placental Function and Implications for Fetal Programming. Int J Mol Sci 2024; 25:9862. [PMID: 39337351 PMCID: PMC11432287 DOI: 10.3390/ijms25189862] [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: 07/22/2024] [Revised: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Accidental exposure to high-dose radiation while pregnant has shown significant negative effects on the developing fetus. One fetal organ which has been studied is the placenta. The placenta performs all essential functions for fetal development, including nutrition, respiration, waste excretion, endocrine communication, and immunological functions. Improper placental development can lead to complications during pregnancy, as well as the occurrence of intrauterine growth-restricted (IUGR) offspring. IUGR is one of the leading indicators of fetal programming, classified as an improper uterine environment leading to the predisposition of diseases within the offspring. With numerous studies examining fetal programming, there remains a significant gap in understanding the placenta's role in irradiation-induced fetal programming. This review aims to synthesize current knowledge on how irradiation affects placental function to guide future research directions. This review provides a comprehensive overview of placental biology, including its development, structure, and function, and summarizes the placenta's role in fetal programming, with a focus on the impact of radiation on placental biology. Taken together, this review demonstrates that fetal radiation exposure causes placental degradation and immune function dysregulation. Given the placenta's crucial role in fetal development, understanding its impact on irradiation-induced IUGR is essential.
Collapse
Affiliation(s)
- Cameron Hourtovenko
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- School of Natural Sciences, Laurentian University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
| | - Shayen Sreetharan
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- Department of Medical Imaging, London Health Sciences Centre, 339 Windermere Rd., London, ON N6A 5A5, Canada
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- School of Natural Sciences, Laurentian University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
| | - T C Tai
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- School of Natural Sciences, Laurentian University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
3
|
Radhakrishna U, Radhakrishnan R, Uppala LV, Muvvala SB, Prajapati J, Rawal RM, Bahado-Singh RO, Sadhasivam S. Prenatal opioid exposure significantly impacts placental protein kinase C (PKC) and drug transporters, leading to drug resistance and neonatal opioid withdrawal syndrome. Front Neurosci 2024; 18:1442915. [PMID: 39238930 PMCID: PMC11376091 DOI: 10.3389/fnins.2024.1442915] [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: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 09/07/2024] Open
Abstract
Background Neonatal Opioid Withdrawal Syndrome (NOWS) is a consequence of in-utero exposure to prenatal maternal opioids, resulting in the manifestation of symptoms like irritability, feeding problems, tremors, and withdrawal signs. Opioid use disorder (OUD) during pregnancy can profoundly impact both mother and fetus, disrupting fetal brain neurotransmission and potentially leading to long-term neurological, behavioral, and vision issues, and increased infant mortality. Drug resistance complicates OUD and NOWS treatment, with protein kinase regulation of drug transporters not fully understood. Methods DNA methylation levels of ATP-binding cassette (ABC) and solute carrier (SLC) drug transporters, along with protein kinase C (PKC) genes, were assessed in 96 placental samples using the Illumina Infinium MethylationEPIC array (850K). Samples were collected from three distinct groups: 32 mothers with infants prenatally exposed to opioids who needed pharmacological intervention for NOWS, 32 mothers with prenatally opioid-exposed infants who did not necessitate NOWS treatment, and 32 mothers who were not exposed to opioids during pregnancy. Results We identified 69 significantly differentially methylated SLCs, with 24 hypermethylated and 34 hypomethylated, and 11 exhibiting both types of methylation changes including SLC13A3, SLC15A2, SLC16A11, SLC16A3, SLC19A2, and SLC26A1. We identified methylation changes in 11 ABC drug transporters (ABCA1, ABCA12, ABCA2, ABCB10, ABCB5, ABCC12, ABCC2, ABCC9, ABCE1, ABCC7, ABCB3): 3 showed hypermethylation, 3 hypomethylation, and 5 exhibited both. Additionally, 7 PKC family genes (PRKCQ, PRKAA1, PRKCA, PRKCB, PRKCH, PRKCI, and PRKCZ) showed methylation changes. These genes are associated with 13 pathways involved in NOWS, including ABC transporters, bile secretion, pancreatic secretion, insulin resistance, glutamatergic synapse, and gastric acid secretion. Conclusion We report epigenetic changes in PKC-related regulation of drug transporters, which could improve our understanding of clinical outcomes like drug resistance, pharmacokinetics, drug-drug interactions, and drug toxicity, leading to maternal relapse and severe NOWS. Novel drugs targeting PKC pathways and transporters may improve treatment outcomes for OUD in pregnancy and NOWS.
Collapse
Affiliation(s)
- Uppala Radhakrishna
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, Royal Oak, MI, United States
| | - Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lavanya V Uppala
- College of Information Science & Technology, the University of Nebraska at Omaha, Peter Kiewit Institute, Omaha, NE, United States
| | - Srinivas B Muvvala
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Jignesh Prajapati
- Department of Biochemistry & Forensic Sciences, Gujarat University, Ahmedabad, India
| | - Rakesh M Rawal
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Ray O Bahado-Singh
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, Royal Oak, MI, United States
| | - Senthilkumar Sadhasivam
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Park S, Hunter ES. Modeling the human placenta: in vitro applications in developmental and reproductive toxicology. Crit Rev Toxicol 2024; 54:431-464. [PMID: 39016688 DOI: 10.1080/10408444.2023.2295349] [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: 07/31/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 07/18/2024]
Abstract
During its temporary tenure, the placenta has extensive and specialized functions that are critical for pre- and post-natal development. The consequences of chemical exposure in utero can have profound effects on the structure and function of pregnancy-associated tissues and the life-long health of the birthing person and their offspring. However, the toxicological importance and critical functions of the placenta to embryonic and fetal development and maturation have been understudied. This narrative will review early placental development in humans and highlight some in vitro models currently in use that are or can be applied to better understand placental processes underlying developmental toxicity due to in utero environmental exposures.
Collapse
Affiliation(s)
- Sarah Park
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| | - Edward Sidney Hunter
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| |
Collapse
|
5
|
Kotta-Loizou I, Pritsa A, Antasouras G, Vasilopoulos SN, Voulgaridou G, Papadopoulou SK, Coutts RHA, Lechouritis E, Giaginis C. Fetus Exposure to Drugs and Chemicals: A Holistic Overview on the Assessment of Their Transport and Metabolism across the Human Placental Barrier. Diseases 2024; 12:114. [PMID: 38920546 PMCID: PMC11202568 DOI: 10.3390/diseases12060114] [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: 04/12/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The placenta exerts a crucial role in fetus growth and development during gestation, protecting the fetus from maternal drugs and chemical exposure. However, diverse drugs and chemicals (xenobiotics) can penetrate the maternal placental barrier, leading to deleterious, adverse effects concerning fetus health. Moreover, placental enzymes can metabolize drugs and chemicals into more toxic compounds for the fetus. Thus, evaluating the molecular mechanisms through which drugs and chemicals transfer and undergo metabolism across the placental barrier is of vital importance. In this aspect, this comprehensive literature review aims to provide a holistic approach by critically summarizing and scrutinizing the potential molecular processes and mechanisms governing drugs and chemical transfer and metabolism across the placental barrier, which may lead to fetotoxicity effects, as well as analyzing the currently available experimental methodologies used to assess xenobiotics placental transfer and metabolism. METHODS A comprehensive and in-depth literature review was conducted in the most accurate scientific databases such as PubMed, Scopus, and Web of Science by using relevant and effective keywords related to xenobiotic placental transfer and metabolism, retrieving 8830 published articles until 5 February 2024. After applying several strict exclusion and inclusion criteria, a final number of 148 relevant published articles were included. RESULTS During pregnancy, several drugs and chemicals can be transferred from the mother to the fetus across the placental barrier by either passive diffusion or through placental transporters, resulting in fetus exposure and potential fetotoxicity effects. Some drugs and chemicals also appear to be metabolized across the placental barrier, leading to more toxic products for both the mother and the fetus. At present, there is increasing research development of diverse experimental methodologies to determine the potential molecular processes and mechanisms of drug and chemical placental transfer and metabolism. All the currently available methodologies have specific strengths and limitations, highlighting the strong demand to utilize an efficient combination of them to obtain reliable evidence concerning drug and chemical transfer and metabolism across the placental barrier. To derive the most consistent and safe evidence, in vitro studies, ex vivo perfusion methods, and in vivo animal and human studies can be applied together with the final aim to minimize potential fetotoxicity effects. CONCLUSIONS Research is being increasingly carried out to obtain an accurate and safe evaluation of drug and chemical transport and metabolism across the placental barrier, applying a combination of advanced techniques to avoid potential fetotoxic effects. The improvement of the currently available techniques and the development of novel experimental protocols and methodologies are of major importance to protect both the mother and the fetus from xenobiotic exposure, as well as to minimize potential fetotoxicity effects.
Collapse
Affiliation(s)
- Ioly Kotta-Loizou
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK;
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Georgios Antasouras
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| | - Spyridon N. Vasilopoulos
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece;
| | - Gavriela Voulgaridou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Sousana K. Papadopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Robert H. A. Coutts
- Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK;
| | - Eleftherios Lechouritis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| |
Collapse
|
6
|
Khorami-Sarvestani S, Vanaki N, Shojaeian S, Zarnani K, Stensballe A, Jeddi-Tehrani M, Zarnani AH. Placenta: an old organ with new functions. Front Immunol 2024; 15:1385762. [PMID: 38707901 PMCID: PMC11066266 DOI: 10.3389/fimmu.2024.1385762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
The transition from oviparity to viviparity and the establishment of feto-maternal communications introduced the placenta as the major anatomical site to provide nutrients, gases, and hormones to the developing fetus. The placenta has endocrine functions, orchestrates maternal adaptations to pregnancy at different periods of pregnancy, and acts as a selective barrier to minimize exposure of developing fetus to xenobiotics, pathogens, and parasites. Despite the fact that this ancient organ is central for establishment of a normal pregnancy in eutherians, the placenta remains one of the least studied organs. The first step of pregnancy, embryo implantation, is finely regulated by the trophoectoderm, the precursor of all trophoblast cells. There is a bidirectional communication between placenta and endometrium leading to decidualization, a critical step for maintenance of pregnancy. There are three-direction interactions between the placenta, maternal immune cells, and the endometrium for adaptation of endometrial immune system to the allogeneic fetus. While 65% of all systemically expressed human proteins have been found in the placenta tissues, it expresses numerous placenta-specific proteins, whose expression are dramatically changed in gestational diseases and could serve as biomarkers for early detection of gestational diseases. Surprisingly, placentation and carcinogenesis exhibit numerous shared features in metabolism and cell behavior, proteins and molecular signatures, signaling pathways, and tissue microenvironment, which proposes the concept of "cancer as ectopic trophoblastic cells". By extensive researches in this novel field, a handful of cancer biomarkers has been discovered. This review paper, which has been inspired in part by our extensive experiences during the past couple of years, highlights new aspects of placental functions with emphasis on its immunomodulatory role in establishment of a successful pregnancy and on a potential link between placentation and carcinogenesis.
Collapse
Affiliation(s)
- Sara Khorami-Sarvestani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Negar Vanaki
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Shojaeian
- Department of Biochemistry, School of Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Kayhan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
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
|
8
|
Chen W, Li Z, Zhong R, Sun W, Chu M. Expression profiles of oviductal mRNAs and lncRNAs in the follicular phase and luteal phase of sheep (Ovis aries) with 2 fecundity gene (FecB) genotypes. G3 (BETHESDA, MD.) 2023; 14:jkad270. [PMID: 38051961 PMCID: PMC10755197 DOI: 10.1093/g3journal/jkad270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023]
Abstract
FecB (also known as BMPR1B) is a crucial gene in sheep reproduction, which has a mutation (A746G) that was found to increase the ovulation rate and litter size. The FecB mutation is associated with reproductive endocrinology, such mutation can control external estrous characteristics and affect follicle-stimulating hormone during the estrous cycle. Previous researches showed that the FecB mutation can regulate the transcriptomic profiles in the reproductive-related tissues including hypothalamus, pituitary, and ovary during the estrous cycle of small-tailed Han (STH) sheep. However, little research has been reported on the correlation between FecB mutation and the estrous cycle in STH sheep oviduct. To investigate the coding and noncoding transcriptomic profiles involved in the estrous cycle and FecB in the sheep oviduct, RNA sequencing was performed to analyze the transcriptomic profiles of mRNAs and long noncoding RNAs (lncRNAs) in the oviduct during the estrous cycle of STH sheep with mutant (FecBBB) and wild-type (FecB++) genotypes. In total, 21,863 lncRNAs and 43,674 mRNAs were screened, the results showed that mRNAs had significantly higher expression levels than the lncRNAs, and the expression levels of these screened transcripts were lower in the follicular phase than they were in the luteal phase. Among them, the oviductal glycoprotein gene (OVGP1) had the highest expression level. In the comparison between the follicular and luteal phases, 57 differentially expressed (DE) lncRNAs and 637 DE mRNAs were detected, including FSTL5 mRNA and LNC_016628 lncRNA. In the comparison between the FecBBB and FecB++ genotypes, 26 DE lncRNAs and 421 DE mRNAs were detected, including EEF1D mRNA and LNC_006270 lncRNA. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis indicated that the DE mRNAs were enriched mainly in terms related to reproduction such as the tight junction, SAGA complex, ATP-binding cassette, nestin, and Hippo signaling pathway. The interaction network between DE lncRNAs and DE mRNAs indicated that LNC_018420 may be the key regulator in sheep oviduct. Together, our results can provide novel insights into the oviductal transcriptomic function against a FecB mutation background in sheep reproduction.
Collapse
Affiliation(s)
- Weihao Chen
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Zhifeng Li
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Rongzhen Zhong
- Jilin Provincial Key Laboratory of Grassland Farming, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun 130102, China
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
9
|
Gong C, Bertagnolli LN, Boulton DW, Coppola P. A Literature Review of Changes in Phase II Drug-Metabolizing Enzyme and Drug Transporter Expression during Pregnancy. Pharmaceutics 2023; 15:2624. [PMID: 38004602 PMCID: PMC10674389 DOI: 10.3390/pharmaceutics15112624] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
The purpose of this literature review is to comprehensively summarize changes in the expression of phase II drug-metabolizing enzymes and drug transporters in both the pregnant woman and the placenta. Using PubMed®, a systematic search was conducted to identify literature relevant to drug metabolism and transport in pregnancy. PubMed was searched with pre-specified terms during the period of 26 May 2023 to 10 July 2023. The final dataset of 142 manuscripts was evaluated for evidence regarding the effect of gestational age and hormonal regulation on the expression of phase II enzymes (n = 16) and drug transporters (n = 38) in the pregnant woman and in the placenta. This comprehensive review exposes gaps in current knowledge of phase II enzyme and drug transporter localization, expression, and regulation during pregnancy, which emphasizes the need for further research. Moreover, the information collected in this review regarding phase II drug-metabolizing enzyme and drug transporter changes will aid in optimizing pregnancy physiologically based pharmacokinetic (PBPK) models to inform dose selection in the pregnant population.
Collapse
Affiliation(s)
- Christine Gong
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lynn N. Bertagnolli
- AstraZeneca LP, Biopharmaceuticals R&D, Clinical Pharmacology & Safety Sciences, Clinical Pharmacology & Quantitative Pharmacology, Gaithersburg, MD 20878, USA
| | - David W. Boulton
- AstraZeneca LP, Biopharmaceuticals R&D, Clinical Pharmacology & Safety Sciences, Clinical Pharmacology & Quantitative Pharmacology, Gaithersburg, MD 20878, USA
| | - Paola Coppola
- AstraZeneca LP, Biopharmaceuticals R&D, Clinical Pharmacology & Safety Sciences, Clinical Pharmacology & Quantitative Pharmacology, Cambridge CB2 0AA, UK
| |
Collapse
|
10
|
Conformational space exploration of cryo-EM structures by variability refinement. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184133. [PMID: 36738875 DOI: 10.1016/j.bbamem.2023.184133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Cryo-EM observation of biological samples enables visualization of sample heterogeneity, in the form of discrete states that are separable, or continuous heterogeneity as a result of local protein motion before flash freezing. Variability analysis of this continuous heterogeneity describes the variance between a particle stack and a volume, and results in a map series describing the various steps undertaken by the sample in the particle stack. While this observation is absolutely stunning, it is very hard to pinpoint structural details to elements of the maps. In order to bridge the gap between observation and explanation, we designed a tool that refines an ensemble of structures into all the maps from variability analysis. Using this bundle of structures, it is easy to spot variable parts of the structure, as well as the parts that are not moving. Comparison with molecular dynamics simulations highlights the fact that the movements follow the same directions, albeit with different amplitudes. Ligand can also be investigated using this method. Variability refinement is available in the Phenix software suite, accessible under the program name phenix.varref.
Collapse
|
11
|
Justesen S, Bilde K, Olesen RH, Pedersen LH, Ernst E, Larsen A. ABCB1 expression is increased in human first trimester placenta from pregnant women classified as overweight or obese. Sci Rep 2023; 13:5175. [PMID: 36997557 PMCID: PMC10063677 DOI: 10.1038/s41598-023-31598-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/14/2023] [Indexed: 04/01/2023] Open
Abstract
Obesity has become a global health challenge also affecting reproductive health. In pregnant women, obesity increases the risk of complications such as preterm birth, macrosomia, gestational diabetes, and preeclampsia. Moreover, obesity is associated with long-term adverse effects for the offspring, including increased risk of cardiovascular and metabolic diseases and neurodevelopmental difficulties. The underlying mechanisms are far from understood, but placental function is essential for pregnancy outcome. Transporter proteins P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) are important for trans-placental transport of endogenous substances like lipids and cortisol, a key hormone in tissue maturation. They also hold a protective function protecting the fetus from xenobiotics (e.g. pharmaceuticals). Animal studies suggest that maternal nutritional status can affect expression of placental transporters, but little is known about the effect on the human placenta, especially in early pregnancy. Here, we investigated if overweight and obesity in pregnant women altered mRNA expression of ABCB1 encoding P-gp or ABCG2 encoding BCRP in first trimester human placenta. With informed consent, 75 first trimester placental samples were obtained from women voluntarily seeking surgical abortion (< gestational week 12) (approval no.: 20060063). Villous samples (average gestational age 9.35 weeks) were used for qPCR analysis. For a subset (n = 38), additional villi were snap-frozen for protein analysis. Maternal BMI was defined at the time of termination of pregnancy. Compared to women with BMI 18.5-24.9 kg/m2 (n = 34), ABCB1 mRNA expression was significantly increased in placenta samples from women classified as overweight (BMI 25-29.9 kg/m2, n = 18) (p = 0.040) and women classified as obese (BMI ≥ 30 kg/m2, n = 23) (p = 0.003). Albeit P-gp expression did not show statistically significant difference between groups, the effect of increasing BMI was the same in male and female pregnancies. To investigate if the P-gp increase was compensated, we determined the expression of ABCG2 which was unaffected by maternal obesity (p = 0.291). Maternal BMI affects ABCB1 but not ABCG2 mRNA expression in first trimester human placenta. Further studies of early placental function are needed to understand how the expression of placental transport proteins is regulated by maternal factors such as nutritional status and determine the potential consequences for placental-fetal interaction.
Collapse
Affiliation(s)
- Signe Justesen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Katrine Bilde
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Rasmus H Olesen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
- Department of Obstetrics and Gynecology, Randers Regional Hospital, 8930, Randers, Denmark
| | - Lars H Pedersen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark
- Department of Obstetrics and Gynecology, Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Erik Ernst
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
- Department of Obstetrics and Gynecology, Horsens Regional Hospital, 8700, Horsens, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark.
| |
Collapse
|
12
|
Mineiro R, Santos C, Gonçalves I, Lemos M, Cavaco JEB, Quintela T. Regulation of ABC transporters by sex steroids may explain differences in drug resistance between sexes. J Physiol Biochem 2023:10.1007/s13105-023-00957-1. [PMID: 36995571 DOI: 10.1007/s13105-023-00957-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/09/2023] [Indexed: 03/31/2023]
Abstract
Drug efficacy is dependent on the pharmacokinetics and pharmacodynamics of therapeutic agents. Tight junctions, detoxification enzymes, and drug transporters, due to their localization on epithelial barriers, modulate the absorption, distribution, and the elimination of a drug. The epithelial barriers which control the pharmacokinetic processes are sex steroid hormone targets, and in this way, sex hormones may also control the drug transport across these barriers. Thus, sex steroids contribute to sex differences in drug resistance and have a relevant impact on the sex-related efficacy of many therapeutic drugs. As a consequence, for the further development and optimization of therapeutic strategies, the sex of the individuals must be taken into consideration. Here, we gather and discuss the evidence about the regulation of ATP-binding cassette transporters by sex steroids, and we also describe the signaling pathways by which sex steroids modulate ATP-binding cassette transporters expression, with a focus in the most important ATP-binding cassette transporters involved in multidrug resistance.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Cecília Santos
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Manuel Lemos
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - José Eduardo B Cavaco
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal.
- UDI-IPG-Unidade de Investigação Para o Desenvolvimento Do Interior, Instituto Politécnico da Guarda, Guarda, Portugal.
| |
Collapse
|
13
|
Lintao RCV, Kammala AK, Vora N, Yaklic JL, Menon R. Fetal membranes exhibit similar nutrient transporter expression profiles to the placenta. Placenta 2023; 135:33-42. [PMID: 36913807 DOI: 10.1016/j.placenta.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
INTRODUCTION During pregnancy, the growth of the fetus is supported by the exchange of nutrients, waste, and other molecules between maternal and fetal circulations in the utero-placental unit. Nutrient transfer, in particular, is mediated by solute transporters such as solute carrier (SLC) and adenosine triphosphate-binding cassette (ABC) proteins. While nutrient transport has been extensively studied in the placenta, the role of human fetal membranes (FM), which was recently reported to have a role in drug transport, in nutrient uptake remains unknown. OBJECTIVES This study determined nutrient transport expression in human FM and FM cells and compared expression with placental tissues and BeWo cells. METHODS RNA sequencing (RNA-Seq) of placental and FM tissues and cells was done. Genes of major solute transporter groups, such as SLC and ABC, were identified. Proteomic analysis of cell lysates was performed via nano-liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) to confirm expression at a protein level. RESULTS We determined that FM tissues and cells derived from the fetal membrane tissues express nutrient transporter genes, and their expression is similar to that seen in the placenta or BeWo cells. In particular, transporters involved in macronutrient and micronutrient transfer were identified in both placental and FM cells. Consistent with RNA-Seq findings, carbohydrate transporters (3), vitamin transport-related proteins (8), amino acid transporters (21), fatty acid transport-related proteins (9), cholesterol transport-related proteins (6) and nucleoside transporters (3) were identified in BeWo and FM cells, with both groups sharing similar nutrient transporter expression. CONCLUSION This study determined the expression of nutrient transporters in human FMs. This knowledge is the first step in improving our understanding of nutrient uptake kinetics during pregnancy. Functional studies are required to determine the properties of nutrient transporters in human FMs.
Collapse
Affiliation(s)
- Ryan C V Lintao
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA; College of Medicine, University of the Philippines Manila, 547 Pedro Gil St., Manila, 1000, Philippines
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| | - Natasha Vora
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA; John Sealy School of Medicine, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
| | - Jerome L Yaklic
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| |
Collapse
|
14
|
McColl ER, Kwok J, Benowitz NL, Patten CA, Hughes CA, Koller KR, Flanagan CA, Thomas TK, Hiratsuka VY, Tyndale RF, Piquette-Miller M. The Effect of Tobacco Use on the Expression of Placental Transporters in Alaska Native Women. Clin Pharmacol Ther 2023; 113:634-642. [PMID: 36053152 PMCID: PMC10234256 DOI: 10.1002/cpt.2737] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022]
Abstract
Prenatal tobacco use among Alaska Native (AN) women has decreased substantially over the past two decades. Previous research suggests that providing AN women with feedback regarding fetal exposure to tobacco may further promote cessation. Transporters in the placenta regulate fetal exposure to nutrients and xenobiotics, including compounds associated with tobacco use. We examined whether prenatal tobacco use impacts transporter expression in the placenta, and whether this is influenced by fetal sex, degree of tobacco exposure, or transporter genotype. At delivery, we obtained placental samples from AN research participants who smoked cigarettes, used commercial chew or iqmik (oral tobacco), or did not use tobacco during pregnancy. Transporter expression was evaluated using qRT-PCR and Western blotting and tested for correlations between transcript levels and urinary biomarkers of tobacco use. The impact of BCRP/ABCG2 and OATP2B1/SLCO2B1 genotypes on protein expression was also examined. Oral tobacco use was associated with decreased P-gp and increased MRP1, MRP3, LAT1, and PMAT mRNA expression. Transcript levels of multiple transporters significantly correlated with tobacco biomarkers in maternal and fetal urine. In women carrying male fetuses, both smoking and oral tobacco were associated with decreased P-gp. Oral tobacco was also associated with decreased LAT1 in women carrying female fetuses. BCRP and OATP2B1 genotypes did not appear to impact protein expression. In conclusion, prenatal tobacco use is associated with altered expression of multiple placental transporters which differs by fetal sex. As transcript levels of multiple transporters were significantly correlated with tobacco use biomarkers, eliminating prenatal tobacco use should alleviate these changes.
Collapse
Affiliation(s)
- Eliza R. McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Jacinda Kwok
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Neal L. Benowitz
- Department of Medicine, Division of Cardiology and Center for Tobacco Control Research and Education, University of California, San Francisco, San Francisco, CA, USA
| | - Christi A. Patten
- Department of Psychiatry and Psychology and Behavioral Health Research Program, Mayo Clinic, Rochester, MN, USA
| | - Christine A. Hughes
- Department of Psychiatry and Psychology and Behavioral Health Research Program, Mayo Clinic, Rochester, MN, USA
| | - Kathryn R. Koller
- Clinical and Research Services, Division of Community Health Services, Alaska Native Tribal Health Consortium (ANTHC), Anchorage, AK, USA
| | - Christie A. Flanagan
- Clinical and Research Services, Division of Community Health Services, Alaska Native Tribal Health Consortium (ANTHC), Anchorage, AK, USA
| | - Timothy K Thomas
- Clinical and Research Services, Division of Community Health Services, Alaska Native Tribal Health Consortium (ANTHC), Anchorage, AK, USA
| | | | - Rachel F. Tyndale
- Departments of Pharmacology and Toxicology, and Psychiatry, Temerty Faculty of Medicine, University of Toronto, and Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Torres-Vergara P, Rivera R, Escudero C, Penny J. Maternal and Fetal Expression of ATP-Binding Cassette and Solute Carrier Transporters Involved in the Brain Disposition of Drugs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:149-177. [PMID: 37466773 DOI: 10.1007/978-3-031-32554-0_7] [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: 07/20/2023]
Abstract
Evidence from preclinical and clinical studies demonstrate that pregnancy is a physiological state capable of modifying drug disposition. Factors including increased hepatic metabolism and renal excretion are responsible for impacting disposition, and the role of membrane transporters expressed in biological barriers, including the placental- and blood-brain barriers, has received considerable attention. In this regard, the brain disposition of drugs in the mother and fetus has been the subject of studies attempting to characterize the mechanisms by which pregnancy could alter the expression of ATP-binding cassette (ABC) and solute carrier (SLC) transporters. This chapter will summarize findings of the influence of pregnancy on the maternal and fetal expression of ABC and SLC transporters in the brain and the consequences of such changes on the disposition of therapeutic drugs.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.
- Grupo de Investigación Vascular (GRIVAS), Universidad del Bio-Bio, Chillán, Chile.
| | - Robin Rivera
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Grupo de Investigación Vascular (GRIVAS), Universidad del Bio-Bio, Chillán, Chile
- Laboratorio de Fisiología Vascular, Facultad de Ciencias Básicas, Universidad del Bio Bio, Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| |
Collapse
|
16
|
Tang C, Deng Y, Shao S, Guo Y, Yang L, Yan Y, Zhang Y, Qiu D, Zhou K, Hua Y, Wang C. Long noncoding RNA UCA1 promotes the expression and function of P-glycoprotein by sponging miR-16-5p in human placental BeWo cells. FASEB J 2023; 37:e22657. [PMID: 36459147 DOI: 10.1096/fj.202201051r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/24/2022] [Accepted: 11/03/2022] [Indexed: 12/04/2022]
Abstract
Investigations on placental P-glycoprotein (P-gp) regulation could provide more therapeutic targets for individualized and safe pharmacotherapy during pregnancy. The role of long noncoding RNA (lncRNA) on placental P-gp regulation is lacking. The present study was carried out to investigate the regulation and underlying mechanisms of lncRNA urothelial carcinoma associated 1 (UCA1) on P-gp in Bewo cells. lncRNA UCA1 inhibition or overexpression could decrease or increase ABCB1 mRNA expression, P-gp expression and its cellular efflux function, respectively. RNA-FISH revealed that lncRNA UCA1 was mainly located in the cytoplasm of Bewo cells. MicroRNA array was applied and 10 significant miRNAs was identified after lncRNA UCA1 inhibition. Databases of LncTarD, LncRNA2Target, and miRcode were further used to search potential target miRNAs of lncRNA UCA1 and miR-16-5p was screened out. Thereafter, we confirmed that miR-16-5p expression was significantly upregulated or reduced after lncRNA UCA1 knockdown or overexpression, respectively. Furthermore, we also proved that ABCB1 mRNA expression, P-gp expression and its cellular efflux function was enhanced or reduced after miR-16-5p inhibition or overexpression, respectively. The rescue experiment further indicated that miR-16-5p was involved in the positive regulation of lncRNA UCA1 on the expression and function of P-gp. Lastly, dual-luciferase reporter system, RNA-binding protein immunoprecipitation and RNA pull-down assays were performed to explore the relationships among lncRNA UCA1, miR-16-5p, and ABCB1. It was found that lncRNA UCA1(1103-1125) could directly interact with miR-16-5p and miR-16-5p could directly target ABCB1 coding DNA sequence region (882-907). In conclusion, LncRNA UCA1 could promote the expression and function of P-gp by sponging miR-16-5p in BeWo cells.
Collapse
Affiliation(s)
- Changqing Tang
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, China.,Department of Pediatric Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Yuxin Deng
- The major of MSc Cancer, Cancer Institute of University College London, London, UK
| | - Shuran Shao
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, China.,West China Medical School of Sichuan University, Chengdu, China
| | - Yafei Guo
- The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lixia Yang
- The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yu Yan
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, China.,West China Medical School of Sichuan University, Chengdu, China
| | - Yi Zhang
- The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,Key Laboratory of Development and Diseases of Women and Children of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dajian Qiu
- The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Kaiyu Zhou
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, China.,The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,Key Laboratory of Development and Diseases of Women and Children of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yimin Hua
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, China.,The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,Key Laboratory of Development and Diseases of Women and Children of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chuan Wang
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, China.,The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Minoia JM, Filia MF, Roma MI, De Fino FT, Copello GJ, Peroni RN. Selective modulation of placental and fetal MDR transporters by chronic in utero exposure to NRTIs in Sprague-Dawley rats: Importance for fetoprotection. Toxicol Appl Pharmacol 2022; 450:116170. [PMID: 35843342 DOI: 10.1016/j.taap.2022.116170] [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: 04/16/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
Multidrug resistance (MDR) transporters present in placenta and fetal tissues reduce intracellular accumulation of their substrates. Consequently, induction of protein expression may further reduce toxic effects of specific xenobiotics. This work aimed to study whether sustained drug treatments in utero could modulate MDR transporters P-gp, BCRP, and MRP2 and thus impact their fetoprotective action. Pregnant Sprague-Dawley rats were daily treated by gavage with zidovudine (AZT, 60 mg/kg) or lamivudine (3TC, 30 mg/kg) from gestation day (GD) 11 to 20. On GD 21, DNA damage and MDR protein abundance were assessed by comet assay and western blotting, respectively. Moreover, a single IV dose of AZT or 3TC was administered on GD 21 and drug concentrations were measured in maternal blood and fetal liver by HPLC-UV. Chronic exposure to 3TC caused significantly higher DNA damage than AZT in fetal liver cells, whereas no differences were observed in maternal blood cells. Increased levels of BCRP protein were found in the placenta and fetal liver after AZT, but not 3TC, chronic in utero exposure. Contrarily, no modifications in the protein abundance of P-gp or MRP2 were found after sustained exposure to these drugs. The area under the curve of AZT in fetal liver was significantly lower in the AZT-pretreated rats than in the VEH or 3TC groups. Moreover, pre-administration of the BCRP inhibitor gefitinib (20 mg/kg, IP) increased AZT levels to the values observed in the VEH-treated group in this tissue. On the other hand, the disposition of 3TC in maternal blood or fetal liver was not modified after chronic treatment in either group. In conclusion, chronic exposure to AZT selectively induces BCRP expression in the placenta and fetal liver decreasing its own accumulation which may account for the lower DNA damage observed for AZT compared to 3TC in fetal liver cells.
Collapse
Affiliation(s)
- Juan Mauricio Minoia
- Instituto de Investigaciones Farmacológicas (ININFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Cátedra de Farmacología, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
| | - María Fernanda Filia
- Instituto de Investigaciones Farmacológicas (ININFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Martín Ignacio Roma
- Instituto de Investigaciones Farmacológicas (ININFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Cátedra de Química Analítica Instrumental e Instituto de Química y Metabolismo del Fármaco (IQUIMEFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernanda Teresa De Fino
- Instituto de Investigaciones Farmacológicas (ININFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Guillermo Javier Copello
- Cátedra de Química Analítica Instrumental e Instituto de Química y Metabolismo del Fármaco (IQUIMEFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Roxana Noemí Peroni
- Instituto de Investigaciones Farmacológicas (ININFA UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Cátedra de Farmacología, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
18
|
Balhara A, Kumar AR, Unadkat JD. Predicting Human Fetal Drug Exposure Through Maternal-Fetal PBPK Modeling and In Vitro or Ex Vivo Studies. J Clin Pharmacol 2022; 62 Suppl 1:S94-S114. [PMID: 36106781 PMCID: PMC9494623 DOI: 10.1002/jcph.2117] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/20/2022] [Indexed: 11/06/2022]
Abstract
Medication (drug) use in human pregnancy is prevalent. Determining fetal safety and efficacy of drugs is logistically challenging. However, predicting (not measuring) fetal drug exposure (systemic and tissue) throughout pregnancy is possible through maternal-fetal physiologically based pharmacokinetic (PBPK) modeling and simulation. Such prediction can inform fetal drug safety and efficacy. Fetal drug exposure can be quantified in 2 complementary ways. First, the ratio of the steady-state unbound plasma concentration in the fetal plasma (or area under the plasma concentration-time curve) to the corresponding maternal plasma concentration (ie, Kp,uu ). Second, the maximum unbound peak (Cu,max,ss,f ) and trough (Cu,min,ss,f ) fetal steady-state plasma concentrations. We (and others) have developed a maternal-fetal PBPK model that can successfully predict maternal drug exposure. To predict fetal drug exposure, the model needs to be populated with drug specific parameters, of which transplacental clearances (active and/or passive) and placental/fetal metabolism of the drug are critical. Herein, we describe in vitro studies in cells/tissue fractions or the perfused human placenta that can be used to determine these drug-specific parameters. In addition, we provide examples whereby this approach has successfully predicted systemic fetal exposure to drugs that passively or actively cross the placenta. Apart from maternal-fetal PBPK models, animal studies also have the potential to estimate fetal drug exposure by allometric scaling. Whether such scaling will be successful is yet to be determined. Here, we review the above approaches to predict fetal drug exposure, outline gaps in our knowledge to make such predictions and map out future research directions that could fill these gaps.
Collapse
Affiliation(s)
- Ankit Balhara
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Aditya R Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
19
|
Ashar Y, Teng Q, Wurpel JND, Chen ZS, Reznik SE. Palmitic Acid Impedes Extravillous Trophoblast Activity by Increasing MRP1 Expression and Function. Biomolecules 2022; 12:1162. [PMID: 36009056 PMCID: PMC9406058 DOI: 10.3390/biom12081162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 12/05/2022] Open
Abstract
Normal function of placental extravillous trophoblasts (EVTs), which are responsible for uteroplacental vascular remodeling, is critical for adequate delivery of oxygen and nutrients to the developing fetus and normal fetal programming. Proliferation and invasion of spiral arteries by EVTs depends upon adequate levels of folate. Multidrug resistance-associated protein 1 (MRP1), which is an efflux transporter, is known to remove folate from these cells. We hypothesized that palmitic acid increases MRP1-mediated folate removal from EVTs, thereby interfering with EVTs' role in early placental vascular remodeling. HTR-8/SVneo and Swan-71 cells, first trimester human EVTs, were grown in the absence or presence of 0.5 mM and 0.7 mM palmitic acid, respectively, for 72 h. Palmitic acid increased ABCC1 gene expression and MRP1 protein expression in both cell lines. The rate of folate efflux from the cells into the media increased with a decrease in migration and invasion functions in the cultured cells. Treatment with N-acetylcysteine (NAC) prevented the palmitic acid-mediated upregulation of MRP1 and restored invasion and migration in the EVTs. Finally, in an ABCC1 knockout subline of Swan-71 cells, there was a significant increase in invasion and migration functions. The novel finding in this study that palmitic acid increases MRP1-mediated folate efflux provides a missing link that helps to explain how maternal consumption of saturated fatty acids compromises the in utero environment.
Collapse
Affiliation(s)
- Yunali Ashar
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY 11439, USA
| | - Qiuxu Teng
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY 11439, USA
| | - John N. D. Wurpel
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY 11439, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY 11439, USA
| | - Sandra E. Reznik
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY 11439, USA
- Departments of Pathology and Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
20
|
Expression of ABCA1 Transporter and LXRA/LXRB Receptors in Placenta of Women with Late Onset Preeclampsia. J Clin Med 2022; 11:jcm11164809. [PMID: 36013052 PMCID: PMC9410380 DOI: 10.3390/jcm11164809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Appropriate levels of cholesterol are necessary for the mother and developing fetus, but theirexcess may cause preeclampsia. The ABCA1 transporter mediates the secretion of cholesterol and is highly regulated at the transcriptional level via the nuclear liver X receptors (LXRs). Methods: Sixteen preeclamptic and 39 normotensives healthy women with uncomplicated pregnancies were involved in the case-control study. The placental levels of ABCA1, LXRA and LXRB mRNA were quantified by real-time quantitative PCR. The concentrations of ABCA1, LXRA and LXRB proteins from the placenta were determined using an enzyme-linked immunosorbent assay Results: We found in the logistic regression model significantly lower placental expression of LXRB mRNA (crude OR = 0.26, 95% CI: 0.07–0.94, p = 0.040) and LXRA protein level (crude OR = 0.19, 95% CI: 0.05–0.69, p = 0.012) in late-onset preeclamptic women compared to healthy pregnant women. The values remained statistically significant after adjustment for possible confounders. Conclusions: Our results suggest that high placenta LXRA mRNA and LXRA protein expression levels decrease the risk of late-onset preeclampsia. These nuclear receptors could play a role in the development of preeclampsia through disturbances of lipid metabolism.
Collapse
|
21
|
Love TM, Wahlberg K, Pineda D, Watson GE, Zareba G, Thurston SW, Davidson PW, Shamlaye CF, Myers GJ, Rand M, van Wijngaarden E, Broberg K. Contribution of child ABC-transporter genetics to prenatal MeHg exposure and neurodevelopment. Neurotoxicology 2022; 91:228-233. [PMID: 35654246 PMCID: PMC9723801 DOI: 10.1016/j.neuro.2022.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/01/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND There is emerging evidence that exposure to prenatal methylmercury (MeHg) from maternal fish consumption during pregnancy can differ between individuals due to genetic variation. In previous studies, we have reported that maternal polymorphisms in ABC-transporter genes were associated with maternal hair MeHg concentrations, and with children's early neurodevelopmental tests. In this study, we add to these findings by evaluating the contribution of genetic variation in children's ABC-transporter genes to prenatal MeHg exposure and early child neurodevelopmental tests. METHODS We genotyped six polymorphisms (rs2032582, rs10276499 and rs1202169 in ABCB1; rs11075290 and rs215088 in ABCC1; rs717620 in ABCC2) in DNA from cord blood and maternal blood of the Seychelles Child Development Study Nutrition Cohort 2. We determined prenatal MeHg exposure by measuring total mercury (Hg) in cord blood by atomic fluorescence spectrometry. We assessed neurodevelopment in children at approximately 20 months using the Bayley Scales of Infant Development (BSID-II). We used linear regression models to analyze covariate-adjusted associations of child genotype with cord MeHg and BSID-II outcomes (Mental Developmental and Psychomotor Developmental Indexes). We also evaluated interactions between genotypes, cord MeHg, and neurodevelopmental outcomes. All models were run with and without adjustment for maternal genotype. RESULTS Of the six evaluated polymorphisms, only ABCC1 rs11075290 was associated with cord blood MeHg; children homozygous for the T-allele had on average 29.99 µg/L MeHg in cord blood while those homozygous for the C-allele had on average 38.06 µg/L MeHg in cord blood (p < 0.001). No polymorphisms in the children were associated with either subscale of the BSID. However, the association between cord MeHg and the Mental Developmental Index (MDI) of the BSID differed significantly across the three genotypes of ABCB1 rs10276499 (2df F-test, p = 0.045). With increasing cord MeHg, the MDI decreased (slope=-0.091, p = 0.014) among children homozygous for the rare C-allele. CONCLUSIONS These findings support the possibility that child ABC genetics might influence prenatal MeHg exposure.
Collapse
Affiliation(s)
- Tanzy M Love
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Karin Wahlberg
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, 22185 Lund, Sweden
| | - Daniela Pineda
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, 22185 Lund, Sweden
| | - Gene E Watson
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Grazyna Zareba
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Sally W Thurston
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Philip W Davidson
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Conrad F Shamlaye
- The Child Development Centre, Ministry of Health, Mahé, Republic of Seychelles
| | - Gary J Myers
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Matthew Rand
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Edwin van Wijngaarden
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Karin Broberg
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, 22185 Lund, Sweden; Institute of Environmental Medicine, Metals and Health, Box 210, 171 77 Stockholm, Sweden.
| |
Collapse
|
22
|
Santos JX, Rasga C, Marques AR, Martiniano H, Asif M, Vilela J, Oliveira G, Sousa L, Nunes A, Vicente AM. A Role for Gene-Environment Interactions in Autism Spectrum Disorder Is Supported by Variants in Genes Regulating the Effects of Exposure to Xenobiotics. Front Neurosci 2022; 16:862315. [PMID: 35663546 PMCID: PMC9161282 DOI: 10.3389/fnins.2022.862315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
Heritability estimates support the contribution of genetics and the environment to the etiology of Autism Spectrum Disorder (ASD), but a role for gene-environment interactions is insufficiently explored. Genes involved in detoxification pathways and physiological permeability barriers (e.g., blood-brain barrier, placenta and respiratory airways), which regulate the effects of exposure to xenobiotics during early stages of neurodevelopment when the immature brain is extremely vulnerable, may be particularly relevant in this context. Our objective was to identify genes involved in the regulation of xenobiotic detoxification or the function of physiological barriers (the XenoReg genes) presenting predicted damaging variants in subjects with ASD, and to understand their interaction patterns with ubiquitous xenobiotics previously implicated in this disorder. We defined a panel of 519 XenoReg genes through literature review and database queries. Large ASD datasets were inspected for in silico predicted damaging Single Nucleotide Variants (SNVs) (N = 2,674 subjects) or Copy Number Variants (CNVs) (N = 3,570 subjects) in XenoReg genes. We queried the Comparative Toxicogenomics Database (CTD) to identify interaction pairs between XenoReg genes and xenobiotics. The interrogation of ASD datasets for variants in the XenoReg gene panel identified 77 genes with high evidence for a role in ASD, according to pre-specified prioritization criteria. These include 47 genes encoding detoxification enzymes and 30 genes encoding proteins involved in physiological barrier function, among which 15 are previous reported candidates for ASD. The CTD query revealed 397 gene-environment interaction pairs between these XenoReg genes and 80% (48/60) of the analyzed xenobiotics. The top interacting genes and xenobiotics were, respectively, CYP1A2, ABCB1, ABCG2, GSTM1, and CYP2D6 and benzo-(a)-pyrene, valproic acid, bisphenol A, particulate matter, methylmercury, and perfluorinated compounds. Individuals carrying predicted damaging variants in high evidence XenoReg genes are likely to have less efficient detoxification systems or impaired physiological barriers. They can therefore be particularly susceptible to early life exposure to ubiquitous xenobiotics, which elicit neuropathological mechanisms in the immature brain, such as epigenetic changes, oxidative stress, neuroinflammation, hypoxic damage, and endocrine disruption. As exposure to environmental factors may be mitigated for individuals with risk variants, this work provides new perspectives to personalized prevention and health management policies for ASD.
Collapse
Affiliation(s)
- João Xavier Santos
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Célia Rasga
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Ana Rita Marques
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Hugo Martiniano
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Muhammad Asif
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Joana Vilela
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Guiomar Oliveira
- Unidade de Neurodesenvolvimento e Autismo, Serviço do Centro de Desenvolvimento da Criança, Centro de Investigação e Formação Clínica, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculty of Medicine, University Clinic of Pediatrics and Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Lisete Sousa
- Departamento de Estatística e Investigação Operacional e Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Nunes
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
- Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Astrid M. Vicente
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
- BioISI–Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
- *Correspondence: Astrid M. Vicente,
| |
Collapse
|
23
|
Human Placental Intracellular Cholesterol Transport: A Focus on Lysosomal and Mitochondrial Dysfunction and Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11030500. [PMID: 35326150 PMCID: PMC8944475 DOI: 10.3390/antiox11030500] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
The placenta participates in cholesterol biosynthesis and metabolism and regulates exchange between the maternal and fetal compartments. The fetus has high cholesterol requirements, and it is taken up and synthesized at elevated rates during pregnancy. In placental cells, the major source of cholesterol is the internalization of lipoprotein particles from maternal circulation by mechanisms that are not fully understood. As in hepatocytes, syncytiotrophoblast uptake of lipoprotein cholesterol involves lipoprotein receptors such as low-density lipoprotein receptor (LDLR) and scavenger receptor class B type I (SR-BI). Efflux outside the cells requires proteins such as the ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. However, mechanisms associated with intracellular traffic of cholesterol in syncytiotrophoblasts are mostly unknown. In hepatocytes, uptaken cholesterol is transported to acidic late endosomes (LE) and lysosomes (LY). Proteins such as Niemann–Pick type C 1 (NPC1), NPC2, and StAR related lipid transfer domain containing 3 (STARD3) are required for cholesterol exit from the LE/LY. These proteins transfer cholesterol from the lumen of the LE/LY into the LE/LY-limiting membrane and then export it to the endoplasmic reticulum, mitochondria, or plasma membrane. Although the production, metabolism, and transport of cholesterol in placental cells are well explored, there is little information on the role of proteins related to intracellular cholesterol traffic in placental cells during physiological or pathological pregnancies. Such studies would be relevant for understanding fetal and placental cholesterol management. Oxidative stress, induced by generating excess reactive oxygen species (ROS), plays a critical role in regulating various cellular and biological functions and has emerged as a critical common mechanism after lysosomal and mitochondrial dysfunction. This review discusses the role of cholesterol, lysosomal and mitochondrial dysfunction, and ROS in the development and progression of hypercholesterolemic pregnancies.
Collapse
|
24
|
Sethuraman V, Pu Y, Gingrich J, Jing J, Long R, Olomu IN, Veiga-Lopez A. Expression of ABC transporters during syncytialization in preeclampsia. Pregnancy Hypertens 2022; 27:181-188. [PMID: 35124425 PMCID: PMC9017055 DOI: 10.1016/j.preghy.2022.01.006] [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: 11/24/2020] [Revised: 12/29/2021] [Accepted: 01/23/2022] [Indexed: 01/12/2023]
Abstract
Preeclampsia complicates 2-8% of pregnancies and is associated with prematurity and intrauterine growth restriction. Cholesterol and sterol transport is a key function of the placenta and it is elicited through ATP binding cassette (ABC) transporters. ABCA1 expression changes during trophoblast cell fusion, a process required to form the placental syncytium that enables maternal-fetal nutrient transfer. ABCA1 expression is dysregulated in preeclamptic placentas. But whether ABC transporters expression during trophoblast fusion is disrupted in preeclampsia remains unknown. We investigated if cholesterol and sterol ABC transporters are altered in term and preterm preeclampsia placentas and during human cytotrophoblast syncytialization. Human placental biopsies were collected from healthy term (≥37 weeks; n = 11) and term preeclamptic (≥36 6/7 weeks; n = 8) and pre-term preeclamptic (28-35 weeks; n = 8) pregnancies. Both, protein and mRNA expression for ABCA1, ABCG1, ABCG5, and ABCG8 were evaluated. Primary cytotrophoblasts isolated from a subset of placentas were induced to syncytialize for 96 h and ABCA1, ABCG1 and ABCG8 mRNA expression evaluated at 0 h and 96 h. Protein and gene expression of ABC transporters were not altered in preeclamptic placentas. In the healthy Term group, ABCA1 expression was similar before and after syncytialization. After 96 h of syncytialization, mRNA expression of ABCA1 and ABCG1 increased significantly, while ABCG8 decreased significantly in term-preeclampsia, but not pre-term preeclampsia. While placental expression of ABCA1 and ABCG1 remained unaltered in term preeclampsia, the disruption in their dynamic expression pattern during cytotrophoblast syncytialization suggests that cholesterol transport may contribute to the pathophysiologic role of the placenta in preeclampsia.
Collapse
Affiliation(s)
- Visalakshi Sethuraman
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago
| | - Jeremy Gingrich
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA
| | - Jiongjie Jing
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA
| | - Robert Long
- Department of Obstetrics and Gynecology, Sparrow Health System, East Lansing, Michigan, USA
| | - Isoken Nicholas Olomu
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, USA; Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
25
|
Fragki S, Hoogenveen R, van Oostrom C, Schwillens P, Piersma AH, Zeilmaker MJ. Integrating in vitro chemical transplacental passage into a generic PBK model: A QIVIVE approach. Toxicology 2022; 465:153060. [PMID: 34871708 DOI: 10.1016/j.tox.2021.153060] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 11/19/2021] [Accepted: 12/01/2021] [Indexed: 11/26/2022]
Abstract
With the increasing application of cell culture models as primary tools for predicting chemical safety, the quantitative extrapolation of the effective dose from in vitro to in vivo (QIVIVE) is of increasing importance. For developmental toxicity this requires scaling the in vitro observed dose-response characteristics to in vivo fetal exposure, while integrating maternal in vivo kinetics during pregnancy, in particular transplacental transfer. Here the transfer of substances across the placental barrier, has been studied using the in vitro BeWo cell assay and six embryotoxic compounds of different kinetic complexity. The BeWo assay results were incorporated in an existing generic Physiologically Based Kinetic (PBK) model which for this purpose was extended with rat pregnancy. Finally, as a "proof of principle", the BeWo PBK model was used to perform a QIVIVE based on developmental toxicity as observed in various different in vitro toxicity assays. The BeWo results illustrated different transport profiles of the chemicals across the BeWo monolayer, allocating the substances into two distinct groups: the 'quickly-transported' and the 'slowly-transported'. BeWo PBK exposure simulations during gestation were compared to experimentally measured maternal blood and fetal concentrations and a reverse dosimetry approach was applied to translate in vitro observed embryotoxicity into equivalent in vivo dose-response curves. This approach allowed for a direct comparison of the in vitro dose-response characteristics as observed in the Whole Embryo Culture (WEC), and the Embryonic Stem Cell test (cardiac:ESTc and neural:ESTn) with in vivo rat developmental toxicity data. Overall, the in vitro to in vivo comparisons suggest a promising future for the application of such QIVIVE methodologies for screening and prioritization purposes of developmental toxicants. Nevertheless, the clear need for further improvements is acknowledged for a wider application of the approach in chemical safety assessment.
Collapse
Affiliation(s)
- Styliani Fragki
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Rudolf Hoogenveen
- Centre for Statistics, Informatics and Modelling, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Conny van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Paul Schwillens
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80178, 3508 TD, Utrecht, the Netherlands
| | - Marco J Zeilmaker
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
26
|
Weyrich A, Frericks M, Eichenlaub M, Schneider S, Hofmann T, Van Cruchten S, van Ravenzwaay B. Ontogeny of renal, hepatic, and placental expression of ATP-binding cassette and solute carrier transporters in the rat and the rabbit. Reprod Toxicol 2022; 107:1-9. [PMID: 34757165 DOI: 10.1016/j.reprotox.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 02/08/2023]
Abstract
Species differences in developmental toxicity can be due to varying expression of xenobiotic transporters. Hence, knowledge on the ontogeny of these transporters, especially in human, rat and rabbit, is pivotal. Two superfamilies of transporters, the ATP-binding cassette (ABC) and the solute carrier (SLC) transporters, are well known for their role in the absorption, distribution and/or elimination of xenobiotics and endogenous substances. The aim of this study was to compare the expression levels of these xenobiotic transporters in liver, kidney and placenta of man, Wistar rat and New Zealand White rabbit during pre- and postnatal development. For this purpose, qPCR experiments were performed for rat and rabbit tissues and the gene expression profiles were compared with literature data from man, rat and rabbit. Data analysis showed large differences in transporter expression in development and between species. These results can be used to better understand developmental toxicity findings in non-clinical species and their relevance for man.
Collapse
Affiliation(s)
- Anastasia Weyrich
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany.
| | - Markus Frericks
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Michael Eichenlaub
- Bioscience Research, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Steffen Schneider
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Thomas Hofmann
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Steven Van Cruchten
- Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Bennard van Ravenzwaay
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| |
Collapse
|
27
|
Yamashita M, Markert UR. Overview of Drug Transporters in Human Placenta. Int J Mol Sci 2021; 22:ijms222313149. [PMID: 34884954 PMCID: PMC8658420 DOI: 10.3390/ijms222313149] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 01/10/2023] Open
Abstract
The transport of drugs across the placenta is a point of great importance in pharmacotherapy during pregnancy. However, the knowledge of drug transport in pregnancy is mostly based on experimental clinical data, and the underlying biological mechanisms are not fully understood. In this review, we summarize the current knowledge of drug transporters in the human placenta. We only refer to human data since the placenta demonstrates great diversity among species. In addition, we describe the experimental models that have been used in human placental transport studies and discuss their availability. A better understanding of placental drug transporters will be beneficial for the health of pregnant women who need drug treatment and their fetuses.
Collapse
Affiliation(s)
- Michiko Yamashita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Osaka 5650871, Japan
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany;
- Correspondence:
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany;
| |
Collapse
|
28
|
Thunbo MØ, Vendelbo JH, Volqvartz T, Witte DR, Larsen A, Pedersen LH. Polypharmacy in polymorbid pregnancies and the risk of congenital malformations-A systematic review. Basic Clin Pharmacol Toxicol 2021; 130:394-414. [PMID: 34841667 DOI: 10.1111/bcpt.13695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022]
Abstract
With an increased prevalence of concurrent morbidities during pregnancy, polypharmacy has become increasingly common in pregnant women. The risks associated with polypharmacy may exceed those of individual medication because of drug-drug interactions. This systematic review aims to evaluate the risk of congenital malformations in polymorbid pregnancies exposed to first-trimester polypharmacy. PubMed, Embase and Scopus were searched to identify original human studies with first- trimester polypharmacy due to polymorbidity as the exposure and congenital malformations as the outcome. After screening of 4034 identified records, seven studies fulfilled the inclusion criteria. Four of the seven studies reported an increased risk of congenital malformations compared with unexposed or monotherapy, odds ratios ranging from 1.1 to >10.0. Particularly, short-term anti-infective treatment combined with other drugs and P-glycoprotein substrates were associated with increased malformation risks. In conclusion, knowledge is limited on risks associated with first-trimester polypharmacy due to polymorbidity with the underlying evidence of low quantity and quality. Therefore, an increased focus on pharmacovigilance to enable safe drug use in early pregnancy is needed. Large-scale register-based studies and better knowledge of placental biology are needed to support the clinical management of polymorbid pregnancies that require polypharmacy.
Collapse
Affiliation(s)
| | | | - Tabia Volqvartz
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lars Henning Pedersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Obstetrics and Gynaecology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
29
|
Wang C, Ma D, Hua Y, Duan H. Modulation of Placental Breast Cancer Resistance Protein by HDAC1 in Mice: Implications for Optimization of Pharmacotherapy During Pregnancy. Reprod Sci 2021; 28:3540-3546. [PMID: 34668144 PMCID: PMC8580892 DOI: 10.1007/s43032-021-00773-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/09/2021] [Indexed: 11/28/2022]
Abstract
Breast cancer resistance protein (BCRP/ABCG2) is a critical drug efflux transporters by limiting drugs’ transplacental transfer rates. More investigations on the regulation of placental BCRP offer great promise for enabling pronounced progress in individualized and safe pharmacotherapy during pregnancy. Histone deacetylases (HDACs) play an important role in epigenetic regulation of placental genes. It was reported recently by us that HDAC1 was involved in placental BCRP regulation in vitro. The aim of this study was to further explore the effect of HDAC1 on placental BCRP expression and functionality in animals. Randomly assigned C57BL pregnant dams received intraperitoneal injections of a negative control siRNA or Hdac1 siRNA from embryonic day 7.5 (E7.5) to E15.5, respectively. At E16.5, glyburide (GLB), a probe for evaluating placental BCRP efflux functionality, was injected via the tail vein. Animals were sacrificed through cervical dislocation at various times (5–180 min) after drug administration. The maternal blood, placentas, and fetal-units were collected. GLB concentrations were determined by a validated high-performance liquid chromatography/mass spectrometry (HPLC-MS) assay. Real-time quantitative PCR (qRT-PCR), Western blot, and immunohistochemical (IHC) analysis were employed to identify mRNA/protein levels and localization of gene expressions, respectively. It was noted that Hdac1 inhibition significantly decreased placental Bcrp expression, with markedly increases of GLB concentrations and area under the concentration-time curve (AUC) in fetal-units. Particularly, the ratios of fetal-unit/maternal plasma GLB concentrations were also significantly elevated following Hdac1 repression. Taken together, these findings suggested that HDAC1 was involved in positive regulation of placental BCRP expression and functionality in vivo.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, RenminNanLu Road, Chengdu, 610041, Sichuan, China
- Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Ma
- Department of Pediatric Rehabilitation, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yimin Hua
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, RenminNanLu Road, Chengdu, 610041, Sichuan, China
- Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- Key Laboratory of Development and Diseases of Women and Children of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongyu Duan
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, RenminNanLu Road, Chengdu, 610041, Sichuan, China.
- Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Shoshan-Barmatz V, Anand U, Nahon-Crystal E, Di Carlo M, Shteinfer-Kuzmine A. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target? Front Physiol 2021; 12:730048. [PMID: 34671273 PMCID: PMC8521008 DOI: 10.3389/fphys.2021.730048] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used for treating diabetes mellitus since the late 1950s. In addition to its antihyperglycemic activity, it was shown to be a potential drug candidate for treating a range of other diseases that include various cancers, cardiovascular diseases, diabetic kidney disease, neurodegenerative diseases, renal diseases, obesity, inflammation, COVID-19 in diabetic patients, and aging. In this review, we focus on the important aspects of mitochondrial dysfunction in energy metabolism and cell death with their gatekeeper VDAC1 (voltage-dependent anion channel 1) as a possible metformin target, and summarize metformin's effects in several diseases and gut microbiota. We question how the same drug can act on diseases with opposite characteristics, such as increasing apoptotic cell death in cancer, while inhibiting it in neurodegenerative diseases. Interestingly, metformin's adverse effects in many diseases all show VDAC1 involvement, suggesting that it is a common factor in metformin-affecting diseases. The findings that metformin has an opposite effect on various diseases are consistent with the fact that VDAC1 controls cell life and death, supporting the idea that it is a target for metformin.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | | | - Marta Di Carlo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
31
|
Anoshchenko O, Storelli F, Unadkat JD. Successful Prediction of Human Fetal Exposure to P-Glycoprotein Substrate Drugs Using the Proteomics-Informed Relative Expression Factor Approach and PBPK Modeling and Simulation. Drug Metab Dispos 2021; 49:919-928. [PMID: 34426410 PMCID: PMC8626637 DOI: 10.1124/dmd.121.000538] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Many women take drugs during their pregnancy to treat a variety of clinical conditions. To optimize drug efficacy and reduce fetal toxicity, it is important to determine or predict fetal drug exposure throughout pregnancy. Previously, we developed and verified a maternal-fetal physiologically based pharmacokinetic (m-f PBPK) model to predict fetal Kp,uu (unbound fetal plasma AUC/unbound maternal plasma AUC) of drugs that passively cross the placenta. Here, we used in vitro transport studies in Transwell, in combination with our m-f PBPK model, to predict fetal Kp,uu of drugs that are effluxed by placental P-glycoprotein (P-gp)-namely, dexamethasone, betamethasone, darunavir, and lopinavir. Using Transwell, we determined the efflux ratio of these drugs in hMDR1-MDCKcP-gpKO cells, in which human P-gp was overexpressed and the endogenous P-gp was knocked out. Then, using the proteomics-informed efflux ratio-relative expressive factor approach, we predicted the fetal Kp,uu of these drugs at term. Finally, to verify our predictions, we compared them with the observed in vivo fetal Kp,uu at term. The latter was estimated using our m-f PBPK model and published fetal [umbilical vein (UV)]/maternal plasma drug concentrations obtained at term (UV/maternal plasma). Fetal Kp,uu predictions for dexamethasone (0.63), betamethasone (0.59), darunavir (0.17), and lopinavir (0.08) were successful, as they fell within the 90% confidence interval of the corresponding in vivo fetal Kp,uu (0.30-0.66, 0.29-0.71, 0.11-0.22, 0.04-0.19, respectively). This is the first demonstration of successful prediction of fetal Kp,uu of P-gp drug substrates from in vitro studies. SIGNIFICANCE STATEMENT: For the first time, using in vitro studies in cells, this study successfully predicted human fetal Kp,uu of P-gp substrate drugs. This success confirms that the m-f PBPK model, combined with the ER-REF approach, can successfully predict fetal drug exposure to P-gp substrates. This success provides increased confidence in the use of the ER-REF approach, combined with the m-f PBPK model, to predict fetal Kp,uu of drugs (transported by P-gp or other transporters), both at term and at earlier gestational ages.
Collapse
Affiliation(s)
- Olena Anoshchenko
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Flavia Storelli
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
32
|
Gély CA, Lacroix MZ, Morin M, Vayssière C, Gayrard V, Picard-Hagen N. Comparison of the materno-fetal transfer of fifteen structurally related bisphenol analogues using an ex vivo human placental perfusion model. CHEMOSPHERE 2021; 276:130213. [PMID: 34088095 DOI: 10.1016/j.chemosphere.2021.130213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/14/2021] [Accepted: 03/04/2021] [Indexed: 06/12/2023]
Abstract
Regulatory measures and public concerns regarding bisphenol A (BPA) have led to its replacement by a variety of alternatives in consumer products. Due to their structural similarity to BPA, these alternatives are under surveillance, however, for potential endocrine disruption. Understanding the materno-fetal transfer of these BPA-related alternatives across the placenta is therefore crucial to assess prenatal exposure risks. The objective of the study was to assess and compare the placental transfer of a set of 15 selected bisphenols (BPs) (BP 4-4, BPA, BPAF, BPAP, 3-3 BPA, BPB, BPBP, BPC, BPE, BPF, BPFL, BPM, BPP, BPS and BPZ) using the ex vivo human placental perfusion model. The UHPLC-MS/MS method for simultaneous quantification of these BPs in perfusion media, within a concentration range of 0.003-5 μM, was able to measure placenta transfer rates as low as 0.6%-4%. Despite their structural similarities, these BPs differed greatly in placental transport efficiency. The placental transfer rates of BP4-4, BPAP, BPE, BPF, 3-3BPA, BPB, BPA were similar to that of antipyrine, indicating that their main transport mechanism was passive diffusion. By contrast, the placental transfer rates of BPFL and BPS were very limited, and intermediate for BPBP, BPZ, BPC, BPM, BPP and BPAF, suggesting weak diffusional permeability and/or that their passage might involve efflux transport. These placental transfer data will be particularly useful for predicting the fetal exposure of this important class of emerging contaminants.
Collapse
Affiliation(s)
- Clémence A Gély
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France; INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France.
| | | | - Mathieu Morin
- Department of Obstetrics and Gynecology, Paule de Viguier Hospital, CHU Toulouse, Toulouse, France.
| | - Christophe Vayssière
- Department of Obstetrics and Gynecology, Paule de Viguier Hospital, CHU Toulouse, Toulouse, France; UMR 1027 INSERM, Team SPHERE, Université de Toulouse, France.
| | - Véronique Gayrard
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France.
| | - Nicole Picard-Hagen
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
33
|
Pastuschek J, Bär C, Göhner C, Budde U, Leidenmuehler P, Groten T, Schleußner E, Markert UR. Ex vivo human placental transfer study on recombinant Von Willebrand factor (rVWF). Placenta 2021; 111:69-75. [PMID: 34171523 DOI: 10.1016/j.placenta.2021.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/29/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Deficiency or mutation of von Willebrand factor (VWF) leads to a coagulation disorder (von Willebrand disease; VWD) which requires a lifelong therapy. For avoiding maternal complications treatment may be necessary also in pregnancy, but placental transfer to the fetus might impact its coagulation system and evoke undesired side effects. As VWF is a very large molecule it may be assumed that it does not pass the placental barrier. To prove this hypothesis the materno-fetal transfer of recombinant VWF (rVWF) has been analyzed ex vivo in a total of 21 valid dual side placenta perfusions. Three groups of five placentas each have been perfused with physiological and up to ten-fold increased concentrations of rVWF for 2 h. Six placentas have been used for control perfusions. A series of different control parameters has been assessed for documentation of intactness and functionality of the placenta and the perfusion system. In not a single analysis, independent of time and concentration, rVWF was detected in the fetal circuit. In the maternal circuit VWF concentration decreased slightly during perfusion. These results demonstrate that recombinant VWF does not pass the human placenta.
Collapse
Affiliation(s)
- J Pastuschek
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - C Bär
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - C Göhner
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - U Budde
- Medilys Laborgesellschaft MbH, Paul-Ehrlich-Str. 1, 22763, Hamburg, Germany
| | | | - T Groten
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - E Schleußner
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - U R Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany.
| |
Collapse
|
34
|
Drug Exposure in Newborns: Effect of Selected Drugs Prescribed to Mothers During Pregnancy and Lactation. Ther Drug Monit 2021; 42:255-263. [PMID: 32068668 DOI: 10.1097/ftd.0000000000000747] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The number of newborns exposed to therapeutic drugs during pregnancy is growing because of the increased use of drugs during pregnancy. In recent years, advances in our understanding of drug placental transfer have augmented the likelihood of a healthy baby in mothers with chronic diseases needing drug therapy. Globally, for example, more than 1.4 million pregnancies in 2015 have been burdened with antiretroviral drugs due to an increasing number of HIV-positive women treated with these drugs, particularly in low- and middle-income countries. In most cases, the fetus is exposed to much higher drug doses in utero than the newborn nursed by the mother. Drug transfer through the placenta takes place by passive diffusion, active transport, or facilitated transport, and drug concentrations in the fetal circulation may be comparable to that in the mother's blood concentration. The excretion of drugs into breastmilk predominantly occurs by passive diffusion, allowing only the non-protein-bound fraction of the blood drug concentration to penetrate. Drug agencies in the United States and Europe highly recommend performing clinical trials in pregnant or breastfeeding women. However, only a few drugs have reported statistically sound data in these patient groups. Most available results concerning pregnancy are obtained from observational studies after birth, assessing outcomes in the newborn or by measuring drug concentrations in the mother and umbilical cord blood. In the case of the lactation period, some studies have evaluated drug concentrations in breastmilk and blood of the mother and/or infant. In this review, exposure to antiretrovirals, immunosuppressants used after solid organ transplantation, and antiepileptics during pregnancy and lactation has been discussed in detail.
Collapse
|
35
|
Ren Z, Gao Y, Gao Y, Liang G, Chen Q, Jiang S, Yang X, Fan C, Wang H, Wang J, Shi YW, Xiao C, Zhong M, Yang X. Distinct placental molecular processes associated with early-onset and late-onset preeclampsia. Am J Cancer Res 2021; 11:5028-5044. [PMID: 33754042 PMCID: PMC7978310 DOI: 10.7150/thno.56141] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Patients with preeclampsia display a spectrum of onset time and severity of clinical presentation, yet the underlying molecular bases for the early-onset and late-onset clinical subtypes are not known. Although several transcriptome studies have been done on placentae from PE patients, only a small number of differentially expressed genes have been identified due to very small sample sizes and no distinguishing of clinical subtypes. Methods: We carried out RNA-seq on 65 high-quality placenta samples, including 33 from 30 patients and 32 from 30 control subjects, to search for dysregulated genes and the molecular network and pathways they are involved in. Results: We identified two functionally distinct sets of dysregulated genes in the two major subtypes: 2,977 differentially expressed genes in early-onset severe preeclampsia, which are enriched with metabolism-related pathways, notably transporter functions; and 375 differentially expressed genes in late-onset severe preeclampsia, which are enriched with immune-related pathways. We also identified some key transcription factors, which may drive the widespread gene dysregulation in both early-onset and late-onset patients. Conclusion: These results suggest that early-onset and late-onset severe preeclampsia have different molecular mechanisms, whereas the late-onset mild preeclampsia may have no placenta-specific causal factors. A few regulators may be the key drivers of the dysregulated molecular pathways.
Collapse
|
36
|
Wang X, Chen P, Zhao L, Zhu L, Wu F. Transplacental Behaviors of Organophosphate Tri- and Diesters Based on Paired Human Maternal and Cord Whole Blood: Efficiencies and Impact Factors. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:3091-3100. [PMID: 33397100 DOI: 10.1021/acs.est.0c06095] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Organophosphate tri- and diesters (tri-OPEs and di-OPEs) were quantified in 63 paired maternal and cord whole blood samples collected in Hubei, China, in which tri-o-cresyl phosphate (ToCP) was predominant. The transplacental transfer efficiencies (expressed as cord blood to maternal blood (C:M) concentration ratios) of aryl-tri-OPEs, such as ToCP (1.61) and triphenyl phosphate (TPHP) (1.06), were higher than those of alkyl-tri-OPEs (0.66-0.76). For the target tri-OPEs and some traditional organic compounds, the C:M ratios first increased with log Kow in the range of 1.63-5.23 and then decreased, showing a parabolic relationship. However, ToCP, with a log Kow of 6.34, deviated from this relationship and displayed the highest C:M ratio (1.61). Molecular docking indicated a very strong binding affinity between ToCP and transthyretin, suggesting that ToCP might be actively transported by transthyretin in the placenta. The di-OPE levels in the blood samples were significantly lower than the corresponding tri-OPE levels, and those in the cord blood were influenced not only by their transplacental behaviors but also by their low excretion rates and the metabolic characteristics of their parent compounds in the fetus. This study provides useful information for accurately assessing the health risks posed by tri-OPEs to pregnant women and fetuses.
Collapse
Affiliation(s)
- Xiaolei Wang
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, P.R. China
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, P. R. China
| | - Pengyu Chen
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, P.R. China
| | - Lixia Zhao
- Agro-Environmental Protection Institute, Ministry of Agriculture, Tianjin 300191, P. R. China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, P.R. China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, P. R. China
| |
Collapse
|
37
|
Ochiai W, Kitaoka S, Kawamura T, Hatogai J, Harada S, Iizuka M, Ariumi M, Takano S, Nagai T, Sasatsu M, Sugiyama K. Maternal and Fetal Pharmacokinetic Analysis of Cannabidiol during Pregnancy in Mice. Drug Metab Dispos 2021; 49:337-343. [PMID: 33531413 DOI: 10.1124/dmd.120.000270] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/25/2021] [Indexed: 11/22/2022] Open
Abstract
Cannabidiol (CBD), a major component of cannabis, has various effects, such as antiemetic and anxiolytic activities, and has recently been marketed as a supplement. The number of people using CBD during pregnancy is increasing, and there are concerns about its effects on the fetus. In addition, the scientific evidence supporting the fetal safety of CBD use during pregnancy is insufficient. To investigate CBD transfer from the mother to the fetus, a single intravenous dose of CBD was administered to pregnant mice in this study, and fetal pharmacokinetics (distribution and elimination) was analyzed. The transfer of CBD from the maternal blood to the fetus was rapid, and the compound accumulated in the fetal brain, liver, and gastrointestinal tract. Conversely, little CBD was transferred from the mother to the amniotic fluid. We analyzed the pharmacokinetics of CBD using a two-compartment model and found that the maternal and fetal half-lives of CBD were approximately 5 and 2 hours, respectively. Furthermore, we performed a moment analysis of the pharmacokinetics of CBD, observing a mean residence time of less than 2 hours in both the mother and fetus. These results suggest that once-daily CBD intake during pregnancy is unlikely to result in CBD accumulation in the mother or fetus. SIGNIFICANCE STATEMENT: CBD is currently marketed as a supplement, and despite its increasing use during pregnancy, little information concerning its fetal effects has been reported. In the present study, CBD was administered to pregnant mice, and the pharmacokinetics in the fetus was investigated using a two-compartment model and moment analysis. The results of these analyses provide important information for estimating the risk to the fetus if CBD is mistakenly consumed during pregnancy.
Collapse
Affiliation(s)
- Wataru Ochiai
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Satoshi Kitaoka
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Taisuke Kawamura
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Jo Hatogai
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Shohei Harada
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Misa Iizuka
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Mashu Ariumi
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Seiya Takano
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Tomomi Nagai
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Masanaho Sasatsu
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Kiyoshi Sugiyama
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
38
|
Nicklisch SC, Hamdoun A. Disruption of small molecule transporter systems by Transporter-Interfering Chemicals (TICs). FEBS Lett 2020; 594:4158-4185. [PMID: 33222203 PMCID: PMC8112642 DOI: 10.1002/1873-3468.14005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/25/2022]
Abstract
Small molecule transporters (SMTs) in the ABC and SLC families are important players in disposition of diverse endo- and xenobiotics. Interactions of environmental chemicals with these transporters were first postulated in the 1990s, and since validated in numerous in vitro and in vivo scenarios. Recent results on the co-crystal structure of ABCB1 with the flame-retardant BDE-100 demonstrate that a diverse range of man-made and natural toxic molecules, hereafter termed transporter-interfering chemicals (TICs), can directly bind to SMTs and interfere with their function. TIC-binding modes mimic those of substrates, inhibitors, modulators, inducers, and possibly stimulants through direct and allosteric mechanisms. Similarly, the effects could directly or indirectly agonize, antagonize or perhaps even prime the SMT system to alter transport function. Importantly, TICs are distinguished from drugs and pharmaceuticals that interact with transporters in that exposure is unintended and inherently variant. Here, we review the molecular mechanisms of environmental chemical interaction with SMTs, the methodological considerations for their evaluation, and the future directions for TIC discovery.
Collapse
Affiliation(s)
- Sascha C.T. Nicklisch
- Department of Environmental Toxicology, University of California, Davis, Davis, CA 95616
| | - Amro Hamdoun
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093-0202
| |
Collapse
|
39
|
Blake BE, Fenton SE. Early life exposure to per- and polyfluoroalkyl substances (PFAS) and latent health outcomes: A review including the placenta as a target tissue and possible driver of peri- and postnatal effects. Toxicology 2020; 443:152565. [PMID: 32861749 PMCID: PMC7530144 DOI: 10.1016/j.tox.2020.152565] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/14/2020] [Accepted: 08/22/2020] [Indexed: 01/09/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are ubiquitous drinking water contaminants of concern due to mounting evidence implicating adverse health outcomes associated with exposure, including reduced kidney function, metabolic syndrome, thyroid disruption, and adverse pregnancy outcomes. PFAS have been produced in the U.S. since the 1940s and now encompass a growing chemical family comprised of diverse chemical moieties, yet the toxicological effects have been studied for relatively few compounds. Critically, exposures to some PFAS in utero are associated with adverse outcomes for both mother and offspring, such as hypertensive disorders of pregnancy (HDP), including preeclampsia, and low birth weight. Given the relationship between HDP, placental dysfunction, adverse health outcomes, and increased risk for chronic diseases in adulthood, the role of both developmental and lifelong exposure to PFAS likely contributes to disease risk in complex ways. Here, evidence for the role of some PFAS in disrupted thyroid function, kidney disease, and metabolic syndrome is synthesized with an emphasis on the placenta as a critical yet understudied target of PFAS and programming agent of adult disease. Future research efforts must continue to fill the knowledge gap between placental susceptibility to environmental exposures like PFAS, subsequent perinatal health risks for both mother and child, and latent health effects in adult offspring.
Collapse
Affiliation(s)
- Bevin E Blake
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of the National Toxicology Program (DNTP), NTP Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institute of Health (NIH), Research Triangle Park, NC, USA.
| | - Suzanne E Fenton
- Division of the National Toxicology Program (DNTP), NTP Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institute of Health (NIH), Research Triangle Park, NC, USA
| |
Collapse
|
40
|
Arumugasaamy N, Rock KD, Kuo CY, Bale TL, Fisher JP. Microphysiological systems of the placental barrier. Adv Drug Deliv Rev 2020; 161-162:161-175. [PMID: 32858104 DOI: 10.1016/j.addr.2020.08.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022]
Abstract
Methods to evaluate maternal-fetal transport across the placental barrier have generally involved clinical observations after-the-fact, ex vivo perfused placenta studies, or in vitro Transwell assays. Given the ethical and technical limitations in these approaches, and the drive to understand fetal development through the lens of transport-induced injury, such as with the examples of thalidomide and Zika Virus, efforts to develop novel approaches to study these phenomena have expanded in recent years. Notably, within the past 10 years, placental barrier models have been developed using hydrogel, bioreactor, organ-on-a-chip, and bioprinting approaches. In this review, we discuss the biology of the placental barrier and endeavors to recapitulate this barrier in vitro using these approaches. We also provide analysis of current limitations to drug discovery in this context, and end with a future outlook.
Collapse
|
41
|
Kazma JM, van den Anker J, Allegaert K, Dallmann A, Ahmadzia HK. Anatomical and physiological alterations of pregnancy. J Pharmacokinet Pharmacodyn 2020; 47:271-285. [PMID: 32026239 PMCID: PMC7416543 DOI: 10.1007/s10928-020-09677-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
The extensive metabolic demands of pregnancy require specific physiological and anatomical changes. These changes affect almost all organ systems, including the cardiovascular, respiratory, renal, gastrointestinal, and hematologic system. The placenta adds another layer of complexity. These changes make it challenging for clinicians to understand presenting signs and symptoms, or to interpret laboratory and radiological tests. Furthermore, these physiological alterations can affect the pharmacokinetics and pharmacodynamics of drugs. Drug safety in lactation is only supported by limited evidence. In addition, the teratogenic effects of medications are often extrapolated from animals, which further adds uncertainties. Unfortunately, pregnant women are only rarely included in clinical drug trials, while doses, regimens, and side effects are often extrapolated from studies conducted in non-pregnant populations. In this comprehensive review, we present the changes occurring in each system with its effects on the pharmacokinetic variables. Understanding these physiological changes throughout normal pregnancy helps clinicians to optimize the health of pregnant women and their fetuses. Furthermore, the information on pregnancy-related physiology is also critical to guide study design in this vulnerable 'orphan' population, and provides a framework to explore pregnancy-related pathophysiology such as pre-eclampsia.
Collapse
Affiliation(s)
- Jamil M Kazma
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Karel Allegaert
- Department of Development and Regeneration, and Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- Department of Clinical Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - André Dallmann
- Clinical Pharmacometrics, Research & Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| | - Homa K Ahmadzia
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
42
|
Anoshchenko O, Prasad B, Neradugomma NK, Wang J, Mao Q, Unadkat JD. Gestational Age-Dependent Abundance of Human Placental Transporters as Determined by Quantitative Targeted Proteomics. Drug Metab Dispos 2020; 48:735-741. [PMID: 32591415 PMCID: PMC7469251 DOI: 10.1124/dmd.120.000067] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/11/2020] [Indexed: 11/24/2022] Open
Abstract
Some women take medication during pregnancy to address a variety of clinical conditions. Because of ethical and logistical concerns, it is impossible to determine fetal drug exposure, and therefore fetal risk, during pregnancy. Hence, alternative approaches need to be developed to predict maternal-fetal drug exposure throughout pregnancy. To do so, we previously developed and verified a maternal-fetal physiologically based pharmacokinetic model, which can predict fetal exposure to drugs that passively cross the placenta. However, many drugs are actively transported by the placenta (e.g., human immunodeficiency virus protease inhibitors). To extend our maternal-fetal physiologically based pharmacokinetic model to these actively transported drugs, we determined the gestational age–dependent changes in the protein abundance of placental transporters. Total cellular membrane fractions from first trimester (T1; n = 15), second trimester (T2; n = 19), and term (n = 15) human placentae obtained from uncomplicated pregnancies were isolated by ultracentrifugation. Transporter protein abundance was determined by targeted quantitative proteomics using liquid chromatography tandem mass specrometry. We observed that breast cancer resistance protein and P-glycoprotein abundance significantly decreased from T1 to term by 55% and 69%, respectively (per gram of tissue). Organic anion–transporting polypeptide (OATP) 2B1 abundance significantly decreased from T1 to T2 by 32%. In contrast, organic cation transporter (OCT) 3 and organic anion transporter 4 abundance significantly increased with gestational age (2-fold from T1 to term, 1.6-fold from T2 to term). Serotonin transporter and norepinephrine transporter did not change with gestational age. The abundance of bile salt export pump, multidrug resistance-associated protein 1-5, Na+-taurocholate cotransporting polypeptide, OATP1B1, OATP1B3, OCTN1-2, concentrative nucleoside transporter 1-3, equilibrative nucleoside transporter 2, and multidrug and toxin extrusion 1 could not be quantified. These data can be incorporated into our maternal-fetal physiologically based pharmacokinetic model to predict fetal exposure to drugs that are actively transported across the placenta.
Collapse
Affiliation(s)
- Olena Anoshchenko
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Qingcheng Mao
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
43
|
Chiral Discrimination of P-glycoprotein in Parturient Women: Effect of Fluoxetine on Maternal-Fetal Fexofenadine Pharmacokinetics. Pharm Res 2020; 37:131. [PMID: 32557079 DOI: 10.1007/s11095-020-02854-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/09/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Fluoxetine, antidepressant widely-used during pregnancy, is a selective inhibitor for P-glycoprotein (P-gp). Fexofenadine, an in vivo P-gp probe, is an antihistamine drug for seasonal allergic rhinitis and chronic urticaria treatment during pregnancy and it is available as a racemic mixture. This study evaluated the chiral discrimination of P-gp investigating the effect of fluoxetine on maternal-fetal pharmacokinetics of fexofenadine. METHODS Healthy parturient women received either a single oral dose of 60 mg racemic fexofenadine (Control group; n = 8) or a single oral dose of 40 mg racemic fluoxetine 3 h before a single oral dose of 60 mg racemic fexofenadine (Interaction group; n = 8). Maternal blood and urine samples were collected up to 48 h after fexofenadine administration. At delivery, maternal-placental-fetal blood samples were collected. RESULTS The maternal pharmacokinetics of fexofenadine was enantioselective (AUC0-∞R-(+)/S-(-) ~ 1.5) in both control and interaction groups. Fluoxetine increased AUC0-∞ (267.7 vs 376.1 ng.h/mL) and decreased oral total clearance (105.1 vs 74.4 L/h) only of S-(-)-fexofenadine, whereas the renal clearance were reduced for both enantiomers, suggesting that the intestinal P-gp-mediated transport of S-(-)-fexofenadine is influenced by fluoxetine to a greater extent that the R-(+)-fexofenadine. However, the transplacental transfer of fexofenadine is low (~16%), non-enantioselective and non-influenced by fluoxetine. CONCLUSIONS A single oral dose of 40 mg fluoxetine inhibited the intestinal P-gp mediated transport of S-(-)-fexofenadine to a greater extent than R-(+)-fexofenadine in parturient women. However, the placental P-gp did not discriminate fexofenadine enantiomers and was not inhibited by fluoxetine.
Collapse
|
44
|
Wang X, Song Y, Wu D, Li Q, Lu S, Li X, Huang H, Zhang J. Inter-individual variations and modulators of MDR1 transport activity in human placenta. Placenta 2020; 97:46-50. [PMID: 32792062 DOI: 10.1016/j.placenta.2020.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/17/2020] [Accepted: 06/01/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION ATP binding cassette (ABC) membrane transporter multidrug resistance 1 (MDR1) is one of the most important efflux transporters in the human placenta protecting the fetus from exposure to xenobiotic toxicity. Recent studies have focused on placental MDR1 expression, but few studies have analyzed placental MDR1 transport activity. The purpose of this study was to investigate placental MDR1 transport activity using a relatively large sample size of human placentas. Furthermore, the effect of ABCB1 gene polymorphisms were investigated along with physiological factors including maternal age, times of pregnancy, BMI, delivery mode or pregnancy complications on placental MDR1 transport activity. METHODS A total of 252 human placentas were obtained after delivery. MDR1 transport activity was detected by N-methyl quinidine uptake in placental microvillus membrane vesicles (MVMVs). Nine common single nucleotide polymorphisms (SNPs) in ABCB1 genes were determined by Snapshot. The association between ABCB1 gene polymorphisms, maternal age, times of pregnancy, BMI, delivery mode or pregnancy complications, and transporter activity was investigated. RESULTS Inter-individual variations of MDR1 transport activity were observed among 252 subjects. The per unit protein activity was ranged from 0.05 to 0.15/mg. Nine SNPs in ABCB1 gene didn't exhibit significant association with transporter activity of MDR1. Likewise, neither age, times of pregnancy, delivery mode nor pregnancy complications showed any significant effect of placental MDR1 transport activity. But placental MDR1 transport activity in obese pregnant women was lower than those in non-obese pregnant women. CONCLUSION Inter-individual variations of MDR1 transport activity existed in human placentas. This may contribute to variations in drug exposure to the fetus affecting clinical outcomes. Maternal age, times of pregnancy, delivery mode nor pregnancy complications included in this study maybe not significantly impact placental MDR1 transport activity, but maternal obese could inhibit placental MDR1 activity.
Collapse
Affiliation(s)
- Xi Wang
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanqin Song
- Second Affiliated Hospital of Yunnan University of Chinese Medicine, China
| | - Di Wu
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qian Li
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shanshan Lu
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaosu Li
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hua Huang
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Zhang
- The Department of Clinical Pharmacy, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
45
|
Indomethacin-grafted and pH-sensitive dextran micelles for overcoming inflammation-mediated multidrug resistance in breast cancer. Carbohydr Polym 2020; 237:116139. [DOI: 10.1016/j.carbpol.2020.116139] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
|
46
|
Jinno N, Furugen A, Kurosawa Y, Kanno Y, Narumi K, Kobayashi M, Iseki K. Effects of single and repetitive valproic acid administration on the gene expression of placental transporters in pregnant rats: An analysis by gestational period. Reprod Toxicol 2020; 96:47-56. [PMID: 32437819 DOI: 10.1016/j.reprotox.2020.04.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
The use of valproic acid (VPA), an antiepileptic drug, during pregnancy, is known to increase various fetal risks. Since VPA has been known to inhibit histone deacetylases (HDACs); its administration could alter gene transcription levels. However, in vivo effects of VPA administration on placental transporters have not been fully elucidated. The purpose of the present study was to comprehensively evaluate the effects of single and repetitive VPA administration on the expression of placental transporters and analyze them by gestational day. We investigated 18 transporters (8 ATP-binding cassette (ABC) and 10 solute carrier (SLC) transporters) in the placentas of pregnant rats that were orally administered 400 mg/kg/day VPA for one or four days, during mid- or late gestation. In the control rats, 4 ABC transporter genes (Abcb1a, 1b, Abcc2, Abcc4) were upregulated, 3 (Abcc3, Abcc5, Abcg2) downregulated through gestation, whereas 1 (Abcc1) was not changed. Regarding SLC transporters, 6 genes (Slc7a5, Slc16a3, Slc22a3, Slc22a4, Slco2b1, Slco4a1) were increased, 1 (Slc29a1) decreased through gestation, whereas 3 (Slc7a8, Slc22a5, Slco2a1) showed no significant change. Single VPA administration altered the expression of 9 transporters and repetitive administration, 13 transporters. In particular, VPA remarkably decreased Abcc4 and Slc22a4 in late gestation and increased Abcc5 during mid-gestation. Our findings indicated that VPA administration changed transporter expression levels in rat placenta, and suggested that sensitivity to VPA differs across gestational stages.
Collapse
Affiliation(s)
- Naoko Jinno
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ayako Furugen
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Yuko Kurosawa
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Yuki Kanno
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan.
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
47
|
Del Gobbo A, Scarfone G, Peccatori FA, Villa A, Ossola W, Ercoli G, Bosari S, Ferrero S, Boggio FL, Grossi E, Cribiù FM. Chemotherapy for breast cancer during pregnancy induces vascular alterations and impaired development of placental villi: A preliminary histopathological study. Eur J Obstet Gynecol Reprod Biol 2020; 250:155-161. [PMID: 32442841 DOI: 10.1016/j.ejogrb.2020.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To evaluate histological alterations in placentas of women affected by breast cancer and treated with chemotherapy during pregnancy. STUDY DESIGN We retrospectively reviewed histological slides of 23 placentas of patients affected by breast cancer and treated with chemotherapy during pregnancy and 23 control placentas of women without breast cancer and with physiological pregnancies of the same gestational age. RESULTS All the patients had breast ductal infiltrating carcinoma, 19 of 23 cases had a G3 cancer. All patients were treated with 2-6 cycles of chemotherapy starting after 16 weeks of gestation, with different protocols. No hypertensive complications and no pre-eclampsia episodes were observed; birth weight was consistent with gestational age in all babies in both group with no uneventful outcomes and no perinatal mortality or fetal malformations. Twenty out of 23 cases (86 %) showed hypoxia-induced villous alterations, including increased syncytial knotting (Tenney-Parker changes), perivillar fibrin deposits, distal villous hypoplasia or accelerated maturation and focal villous chorangiosis. These alterations were found in 19 out of 23 controls (83 %), with no statistically significant difference between the two groups. CONCLUSIONS These results shows that chemotherapy in the second and third trimester of pregnancy may lead to non-specific alterations in placental vasculature and morphology.
Collapse
Affiliation(s)
- Alessandro Del Gobbo
- Division of Pathology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy.
| | - Giovanna Scarfone
- Department of Obstetrics, Gynecology and Neonatology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Fedro Alessandro Peccatori
- Fertility and Procreation Unit, Division of Gynecologic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Antonella Villa
- Department of Obstetrics, Gynecology and Neonatology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Wally Ossola
- Department of Obstetrics, Gynecology and Neonatology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Ercoli
- Division of Pathology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvano Bosari
- Division of Pathology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Francesca Laura Boggio
- Division of Pathology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Grossi
- Fertility and Procreation Unit, Division of Gynecologic Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Clinical and Community Sciences, University of Milan, Milan, Italy
| | - Fulvia Milena Cribiù
- Division of Pathology, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
48
|
Cotteret C, Pham YV, Marcais A, Driessen M, Cisternino S, Schlatter J. Maternal ABVD chemotherapy for Hodgkin lymphoma in a dichorionic diamniotic pregnancy: a case report. BMC Pregnancy Childbirth 2020; 20:231. [PMID: 32306909 PMCID: PMC7168968 DOI: 10.1186/s12884-020-02928-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 04/07/2020] [Indexed: 12/20/2022] Open
Abstract
Background Hodgkin lymphoma (HL) is the most common hematological malignancy during pregnancy. The first-line treatment for HL in pregnancy is the standard ABVD regimen without any drug and/or dose adjustment. However, data on chemotherapy during twin pregnancies are sparse, and a better understanding of the mechanisms involved in exposure to and the toxic effects of anticancer drugs in the fetuses is needed. Case presentation A 41-year-old dichorionic diamniotic pregnant patient was given ABVD treatment for HL at a gestational age of 28 weeks and 3 days. The patient received 2 cycles of chemotherapy with a 15-day therapeutic window including an actual 25 mg/m2 dose of doxorubicin per cycle. Unlike the female twin, the male twin presented four days after birth a left cardiac dysfunction. Doxorubicin cardiotoxicity in the male newborn was also supported by high blood levels of troponin. At one month of age, echocardiography findings were normal. We investigated literature data on physiological aspects of pregnancy that may influence doxorubicin pharmacokinetics, and pharmacodynamic and pharmacokinetic data on the use of doxorubicin in pregnancy. We detailed the role of the transporters in doxorubicin placenta distribution, and tried to understand why only one fetus was affected. Conclusions Fetal safety depends at least on maternal doxorubicin pharmacokinetics.Because of drug interactions (i.e. drug metabolism and drug transport), care should always be taken to avoid maternal pharmacokinetic variability. The toxic effects were discrepant between the dizygotic twins, suggesting additional fetus-specific pharmacokinetic/pharmacodynamic factors in doxorubicin toxicity.
Collapse
Affiliation(s)
- Camille Cotteret
- Hôpital universitaire Necker - Enfants malades, Pharmacie, F-75015, Paris, France
| | - Yen-Vi Pham
- Hôpital universitaire Necker - Enfants malades, Pharmacie, F-75015, Paris, France
| | - Ambroise Marcais
- Hôpital universitaire Necker - Enfants malades, Hématologie adultes, F-75015, Paris, France
| | - Marine Driessen
- Hôpital universitaire Necker - Enfants malades, gynécologie-obstétrique, F-75015, Paris, France
| | - Salvatore Cisternino
- Hôpital universitaire Necker - Enfants malades, Pharmacie, F-75015, Paris, France.,Inserm UMR-S 1144, Team "Blood-brain barrier in brain pathophysiology and therapy", Université Paris Descartes, F-75006, Paris, France
| | - Joël Schlatter
- Hôpital universitaire Necker - Enfants malades, Pharmacie, F-75015, Paris, France.
| |
Collapse
|
49
|
Eke AC, Brooks KM, Gebreyohannes RD, Sheffield JS, Dooley KE, Mirochnick M. Tenofovir alafenamide use in pregnant and lactating women living with HIV. Expert Opin Drug Metab Toxicol 2020; 16:333-342. [PMID: 32125906 PMCID: PMC9214649 DOI: 10.1080/17425255.2020.1738384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/02/2020] [Indexed: 12/23/2022]
Abstract
Introduction: Tenofovir alafenamide (TAF)-containing fixed-dose drug combinations (FDCs) are increasingly being used in managing pregnant women living with HIV. However, TAF is not currently recommended during pregnancy due to limited pharmacokinetic and safety data. TAF, a newer nucleotide phosphonamidate prodrug of tenofovir (TFV), achieves high levels of tenofovir-diphosphate in lymphoid cells and hepatocytes, and 90% lower systemic concentrations of TFV compared to tenofovir disoproxil fumarate (TDF), thereby maximizing TAF's antiviral efficacy, potency and clinical safety.Areas covered: This review discusses the currently available information on the pharmacology of TAF in pregnant women living with HIV. Pharmacokinetic studies with TAF during pregnancy have yielded varying results compared to postpartum, but TAF exposures during pregnancy have been within the range of those typically observed in non-pregnant adults. The efficacy and safety of TAF in treatment-naïve pregnant women living with HIV is currently being evaluated in the VESTED study, a phase-III NIH randomized clinical trial.Expert opinion: Initial pregnancy data suggest that TAF-based FDCs have high efficacy and low risk of adverse effects during pregnancy. TAF is likely to become part of first-line regimens for use in pregnant women living with HIV once additional pregnancy data from phase III trials are available.
Collapse
Affiliation(s)
- Ahizechukwu C. Eke
- Division of Maternal Fetal Medicine & Clinical Pharmacology, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Doctoral Training Program (Phd), Graduate Training Program in Clinical Investigation (GTPCI), Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Kristina M. Brooks
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rahel D. Gebreyohannes
- Department of Obstetrics & Gynecology, Addis Ababa University College of Health Science, Addis Ababa, Ethiopia
| | - Jeanne S. Sheffield
- Division of Maternal Fetal Medicine & Clinical Pharmacology, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly E. Dooley
- Division of Clinical Pharmacology & Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark Mirochnick
- Division of Neonatology, Department of Pediatrics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
50
|
Abstract
Pharmacologic interventions play a major role in obstetrical care throughout pregnancy, labor and delivery and the postpartum. Traditionally, obstetrical providers have utilized standard dosing regimens developed for non-obstetrical indications based on pharmacokinetic knowledge from studies in men or non-pregnant women. With the recognition of pregnancy as a special pharmacokinetic population in the late 1990s, investigators have begun to study drug disposition in this unique patient dyad. Many of the basic physiologic changes that occur during pregnancy have significant impact on drug absorption, distribution and clearance. Activity of Phase I and Phase II drug metabolizing enzymes are differentially altered by pregnancy, resulting in drug concentrations sufficiently different for some medications that efficacy or toxicity is affected. Placental transporters play a major dynamic role in determining fetal drug exposure. In the past two decades, we have begun to expand our understanding of obstetrical pharmacology; however, to truly optimize pharmacologic care of our pregnant patients and their developing fetus, additional research is critically needed.
Collapse
|