1
|
Abostait A, Abdelkarim M, Bao Z, Miyake Y, Tse WH, Di Ciano-Oliveir C, Buerki-Thurnherr T, Allen C, Keijzer R, Labouta HI. Optimizing lipid nanoparticles for fetal gene delivery in vitro, ex vivo, and aided with machine learning. J Control Release 2024; 376:678-700. [PMID: 39447842 DOI: 10.1016/j.jconrel.2024.10.047] [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: 07/30/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
There is a clinical need to develop lipid nanoparticles (LNPs) to deliver congenital therapies to the fetus during pregnancy. The aim of these therapies is to restore normal fetal development and prevent irreversible conditions after birth. As a first step, LNPs need to be optimized for transplacental transport, safety on the placental barrier and fetal organs and transfection efficiency. We developed and characterized a library of LNPs of varying compositions and used machine learning (ML) models to delineate the determinants of LNP size and zeta potential. Utilizing different in vitro placental models with the help of a Random Forest algorithm, we could identify the top features driving percentage LNP transport and kinetics at 24 h, out of a total of 18 input features represented by 41 LNP formulations and 48 different transport experiments. We further evaluated the LNPs for safety, placental cell uptake, transfection efficiency in placental trophoblasts and fetal lung fibroblasts. To ensure the integrity of the LNPs following transplacental transport, we screened LNPs for transport and transfection using a high-throughput integrated transport-transfection in vitro model. Finally, we assessed toxicity of the LNPs in a tracheal occlusion fetal lung explant model. LNPs showed little to no toxicity to fetal and placental cells. Immunoglobin G (IgG) orientation on the surface of LNPs, PEGylated lipids, and ionizable lipids had significant effects on placental transport. The Random Forest algorithm identified the top features driving LNPs placental transport percentage and kinetics. Zeta potential emerged in the top driving features. Building on the ML model results, we developed new LNP formulations to further optimize the transport leading to 622 % increase in transport at 24 h versus control LNP formulation. To induce preferential siRNA transfection of fetal lung, we further optimized cationic lipid percentage and the lipid-to-siRNA ratio. Studying LNPs in an integrated placental and fetal lung fibroblasts model showed a strong correlation between zeta potential and fetal lung transfection. Finally, we assessed the toxicity of LNPs in a tracheal occlusion lung explant model. The optimized formulations appeared to be safe on ex vivo fetal lungs as indicated by insignificant changes in apoptosis (Caspase-3) and proliferation (Ki67) markers. In conclusion, we have optimized an LNP formulation that is safe, with high transplacental transport and preferential transfection in fetal lung cells. Our research findings represent an important step toward establishing the safety and effectiveness of LNPs for gene delivery to the fetal organs.
Collapse
Affiliation(s)
- Amr Abostait
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Mahmoud Abdelkarim
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Zeqing Bao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Yuichiro Miyake
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo 113-8421, Japan
| | - Wai Hei Tse
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | | | - Tina Buerki-Thurnherr
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen 9014, Switzerland
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Richard Keijzer
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Hagar I Labouta
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada; Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto M5S 3G9, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada.
| |
Collapse
|
2
|
Fuenzalida B, Basler V, Koechli N, Yi N, Staud F, Albrecht C. Modelling the maternal-fetal interface: An in vitro approach to investigate nutrient and drug transport across the human placenta. J Cell Mol Med 2024; 28:e70151. [PMID: 39422159 PMCID: PMC11487339 DOI: 10.1111/jcmm.70151] [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: 05/21/2024] [Revised: 09/20/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
The placenta plays a critical role in maternal-fetal nutrient transport and fetal protection against drugs. Creating physiological in vitro models to study these processes is crucial, but technically challenging. This study introduces an efficient cell model that mimics the human placental barrier using co-cultures of primary trophoblasts and primary human umbilical vein endothelial cells (HUVEC) on a Transwell®-based system. Monolayer formation was examined over 7 days by determining transepithelial electrical resistance (TEER), permeability of Lucifer yellow (LY) and inulin, localization of transport proteins at the trophoblast membrane (immunofluorescence), and syncytialization markers (RT-qPCR/ELISA). We analysed diffusion-based (caffeine/antipyrine) and transport-based (leucine/Rhodamine-123) processes to study the transfer of physiologically relevant compounds. The latter relies on the adequate localization and function of the amino-acid transporter LAT1 and the drug transporter P-glycoprotein (P-gp) which were studied by immunofluorescence microscopy and application of respective inhibitors (2-Amino-2-norbornanecarboxylic acid (BCH) for LAT1; cyclosporine-A for P-gp). The formation of functional monolayer(s) was confirmed by increasing TEER values, low LY transfer rates, minimal inulin leakage, and appropriate expression/release of syncytialization markers. These results were supported by microscopic monitoring of monolayer formation. LAT1 was identified on the apical and basal sides of the trophoblast monolayer, while P-gp was apically localized. Transport assays confirmed the inhibition of LAT1 by BCH, reducing both intracellular leucine levels and leucine transport to the basal compartment. Inhibiting P-gp with cyclosporine-A increased intracellular Rhodamine-123 concentrations. Our in vitro model mimics key aspects of the human placental barrier. It represents a powerful tool to study nutrient and drug transport mechanisms across the placenta, assisting in evaluating safer pregnancy therapies.
Collapse
Affiliation(s)
- Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| | - Virginia Basler
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| | - Nadja Koechli
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| | - Nan Yi
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec KraloveCharles UniversityHradec KraloveCzech Republic
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| |
Collapse
|
3
|
Vachalova V, Kumnova F, Synova T, Anandam KY, Abad C, Karahoda R, Staud F. Metformin inhibits OCT3-mediated serotonin transport in the placenta. Biomed Pharmacother 2024; 179:117399. [PMID: 39243433 DOI: 10.1016/j.biopha.2024.117399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
Proper fetal development requires tight regulation of serotonin concentrations within the fetoplacental unit. This homeostasis is partly maintained by the placental transporter OCT3/SLC22A3, which takes up serotonin from the fetal circulation. Metformin, an antidiabetic drug commonly used to treat gestational diabetes mellitus, was shown to inhibit OCT3. We, therefore, hypothesized that its use during pregnancy could disrupt placental serotonin homeostasis. This hypothesis was tested using three experimental model systems: primary trophoblast cells isolated from the human term placenta, fresh villous human term placenta fragments, and rat term placenta perfusions. Inhibition of serotonin transport by metformin at three concentrations (1 μM, 10 μM, and 100 μM) was assessed in all three models. The OCT3 inhibitor decynium-22 (100 μM) and paroxetine (100 μM), a dual inhibitor of SERT and OCT3, were used as controls. In primary trophoblasts, paroxetine exhibited the strongest inhibition of serotonin uptake, followed by decynium-22. Metformin showed a concentration-dependent effect, reducing serotonin uptake by up to 57 % at the highest concentration. Its inhibitory effect was less pronounced in fresh villous fragments but remained statistically significant at all concentrations. In the perfused rat placenta, metformin demonstrated a concentration-dependent effect, reducing placental serotonin uptake by 44 % at the highest concentration tested. Our findings across all experimental models show inhibition of placental OCT3 by metformin, resulting in reduced serotonin uptake by the trophoblast. This sheds light on mechanisms that may underpin metformin-mediated effects on fetal development.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Fiona Kumnova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
4
|
Deng ZM, Dai FF, Wang RQ, Deng HB, Yin TL, Cheng YX, Chen GT. Organ-on-a-chip: future of female reproductive pathophysiological models. J Nanobiotechnology 2024; 22:455. [PMID: 39085921 PMCID: PMC11290169 DOI: 10.1186/s12951-024-02651-w] [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: 08/02/2023] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
The female reproductive system comprises the internal and external genitalia, which communicate through intricate endocrine pathways. Besides secreting hormones that maintain the female secondary sexual characteristics, it also produces follicles and offspring. However, the in vitro systems have been very limited in recapitulating the specific anatomy and pathophysiology of women. Organ-on-a-chip technology, based on microfluidics, can better simulate the cellular microenvironment in vivo, opening a new field for the basic and clinical research of female reproductive system diseases. This technology can not only reconstruct the organ structure but also emulate the organ function as much as possible. The precisely controlled fluidic microenvironment provided by microfluidics vividly mimics the complex endocrine hormone crosstalk among various organs of the female reproductive system, making it a powerful preclinical tool and the future of pathophysiological models of the female reproductive system. Here, we review the research on the application of organ-on-a-chip platforms in the female reproductive systems, focusing on the latest progress in developing models that reproduce the physiological functions or disease features of female reproductive organs and tissues, and highlighting the challenges and future directions in this field.
Collapse
Affiliation(s)
- Zhi-Min Deng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Fang-Fang Dai
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui-Qi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Hong-Bing Deng
- Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan, Hubei, 430060, China
| | - Tai-Lang Yin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Gan-Tao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
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
|
Hori T, Okae H, Shibata S, Kobayashi N, Kobayashi EH, Oike A, Sekiya A, Arima T, Kaji H. Trophoblast stem cell-based organoid models of the human placental barrier. Nat Commun 2024; 15:962. [PMID: 38332125 PMCID: PMC10853531 DOI: 10.1038/s41467-024-45279-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Human placental villi have essential roles in producing hormones, mediating nutrient and waste exchange, and protecting the fetus from exposure to xenobiotics. Human trophoblast organoids that recapitulate the structure of villi could provide an important in vitro tool to understand placental development and the transplacental passage of xenobiotics. However, such organoids do not currently exist. Here we describe the generation of trophoblast organoids using human trophoblast stem (TS) cells. Following treatment with three kinds of culture medium, TS cells form spherical organoids with a single outer layer of syncytiotrophoblast (ST) cells that display a barrier function. Furthermore, we develop a column-type ST barrier model based on the culture condition of the trophoblast organoids. The bottom membrane of the column is almost entirely covered with syndecan 1-positive ST cells. The barrier integrity and maturation levels of the model are confirmed by measuring transepithelial/transendothelial electrical resistance (TEER) and the amount of human chorionic gonadotropin. Further analysis reveals that the model can be used to derive the apparent permeability coefficients of model compounds. In addition to providing a suite of tools for the study of placental development, our trophoblast models allow the evaluation of compound transfer and toxicity, which will facilitate drug development.
Collapse
Affiliation(s)
- Takeshi Hori
- Department of Diagnostic and Therapeutic Systems Engineering, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Hiroaki Okae
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Shun Shibata
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Norio Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eri H Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Akira Oike
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Asato Sekiya
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Hirokazu Kaji
- Department of Diagnostic and Therapeutic Systems Engineering, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan.
| |
Collapse
|
7
|
Fuenzalida B, Albrecht C. Assessing Cholesterol Efflux on Primary Human Trophoblast Cells. Methods Mol Biol 2024; 2728:123-129. [PMID: 38019396 DOI: 10.1007/978-1-0716-3495-0_10] [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] [Indexed: 11/30/2023]
Abstract
Cholesterol transport across the placenta must be tightly regulated to avoid a deficiency or an oversupply of cholesterol which is transferred from the mother to the fetus. In trophoblasts, the transport of cholesterol across the cell membrane is mainly mediated by the ATP-binding transporters, ABCA1 and ABCG1. The localization of the transporters at the apical and basal sides of syncytiotrophoblasts has been described. A frequently used method to quantify the amount of cholesterol that cells are capable of exporting is the cholesterol efflux assay. The principle of this assay is that when exogenous [3H]-labeled cholesterol is provided to cultured cells, the efflux of the radioactive cholesterol toward different acceptors in the culture medium is evaluated. Then, the percentage of cholesterol efflux from the cells to the acceptors is calculated. The present work gives an overview on the principle of this assay and a detailed protocol of this technique performed in primary trophoblasts isolated from human term placentas.
Collapse
Affiliation(s)
- Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Zaugg J, Albrecht C. Assessment of Placental Sodium-Independent Leucine Uptake and Transfer in Trophoblast Cells. Methods Mol Biol 2024; 2728:105-121. [PMID: 38019395 DOI: 10.1007/978-1-0716-3495-0_9] [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] [Indexed: 11/30/2023]
Abstract
The placenta maintains the balance between nutrition and growth control of the fetus through selective and regulated supply of macronutrients such as carbohydrates, proteins, lipids, and critical micronutrients. Perturbations in the balanced supply of nutrients as found in gestational diseases and altered fetal development have been associated with changes in amino acid transport proteins, such as the System L amino acid heterodimeric exchangers LAT1/SLC7A5 and LAT2/SLC7A8. Syncytiotrophoblasts (STB) form the crucial cell layer at the placental barrier coordinating the transfer of essential amino acids such as leucine from the maternal to the fetal circulation. The System L-mediated leucine transport across the placental barrier is a Na+-independent process against a counter-directed gradient, maintained by a tightly regulated interplay between accumulative transporters, exchangers, and facilitators.The two methods described here allow to standardize and characterize the uptake kinetics of leucine in conventionally cultured BeWo cells and the transfer of leucine across the placental cell barrier using a BeWo monolayer in the Transwell® system.
Collapse
Affiliation(s)
- Jonas Zaugg
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland.
| |
Collapse
|
9
|
Zhang L, Huang S, Ma K, Chen Y, Wei T, Ye H, Wu J, Liu L, Deng J, Luo H, Tan C. Retinoic Acid-PPARα Mediates β-Carotene Resistance to Placental Dysfunction Induced by Deoxynivalenol. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18696-18708. [PMID: 38012857 DOI: 10.1021/acs.jafc.3c06647] [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: 11/29/2023]
Abstract
Deoxynivalenol (DON), one of the most polluted mycotoxins in the environment and food, has been proven to have strong embryonic and reproductive toxicities. However, the effects of DON on placental impairment and effective interventions are still unclear. This study investigated the effect of β-carotene on placental functional impairment and its underlying molecular mechanism under DON exposure. Adverse pregnancy outcomes were caused by intraperitoneal injection of DON from 13.5 to 15.5 days of gestation in mice, resulting in higher enrichment of DON in placenta than in other tissue samples. Interestingly, 0.1% β-carotene dietary supplementation could significantly alleviate DON-induced pregnancy outcomes. Additionally, in vivo and in vitro placental barrier models demonstrated the association of DON-induced placental function impairment with placental permeability barrier disruption, angiogenesis impairment, and oxidative stress induction. Moreover, β-carotene regulated DON-induced placental toxicity by activating the expressions of claudin 1, zonula occludens-1, and vascular endothelial growth factor-A through retinoic acid-peroxisome proliferator-activated receptor α signaling.
Collapse
Affiliation(s)
- Longmiao Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Shuangbo Huang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Kaidi Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yiling Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Tanghong Wei
- Dekon Food and Agriculture Group, Chengdu, Sichuan 610225, China
| | - Hongxuan Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Junyi Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Liudan Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Hefeng Luo
- Dekon Food and Agriculture Group, Chengdu, Sichuan 610225, China
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| |
Collapse
|
10
|
Zhou Z, Luo D, Li M, Lao G, Zhou Z, Dinnyés A, Xu W, Sun Q. A Novel Multicellular Placental Barrier Model to Investigate the Effect of Maternal Aflatoxin B 1 Exposure on Fetal-Side Neural Stem Cells. Toxins (Basel) 2023; 15:toxins15050312. [PMID: 37235346 DOI: 10.3390/toxins15050312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Ingestion of food toxins such as aflatoxin B1 (AFB1) during pregnancy may impair fetal neurodevelopment. However, animal model results may not be accurate due to the species' differences, and testing on humans is ethically impermissible. Here, we developed an in vitro human maternal-fetal multicellular model composed of a human hepatic compartment, a bilayer placental barrier, and a human fetal central nervous system compartment using neural stem cells (NSCs) to investigate the effect of AFB1 on fetal-side NSCs. AFB1 passed through the HepG2 hepatocellular carcinoma cells to mimic the maternal metabolic effects. Importantly, even at the limited concentration (0.0641 ± 0.0046 μM) of AFB1, close to the national safety level standard of China (GB-2761-2011), the mixture of AFB1 crossing the placental barrier induced NSC apoptosis. The level of reactive oxygen species in NSCs was significantly elevated and the cell membrane was damaged, causing the release of intracellular lactate dehydrogenase (p < 0.05). The comet experiment and γ-H2AX immunofluorescence assay showed that AFB1 caused significant DNA damage to NSCs (p < 0.05). This study provided a new model for the toxicological evaluation of the effect of food mycotoxin exposure during pregnancy on fetal neurodevelopment.
Collapse
Affiliation(s)
- Zhiwei Zhou
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Dongmei Luo
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Mengxue Li
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Guangjie Lao
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Zhiqiang Zhou
- Department of Food Engineering, Sichuan University, Chengdu 610064, China
| | - András Dinnyés
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Godollo, Hungary
- Department of Cell Biology and Molecular Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Wenming Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610064, China
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Qun Sun
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
11
|
Dusza HM, van Boxel J, van Duursen MBM, Forsberg MM, Legler J, Vähäkangas KH. Experimental human placental models for studying uptake, transport and toxicity of micro- and nanoplastics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 860:160403. [PMID: 36417947 DOI: 10.1016/j.scitotenv.2022.160403] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Micro- and nanoplastics (MNPs) are ubiquitous in the environment and have recently been found in human lungs, blood and placenta. However, data on the possible effects of MNPs on human health is extremely scarce. The potential toxicity of MNPs during pregnancy, a period of increased susceptibility to environmental insults, is of particular concern. The placenta provides a unique interface between maternal and fetal circulation which is essential for in utero survival and healthy pregnancy. Placental toxicokinetics and toxicity of MNPs are still largely unexplored and the limited studies performed up to now focus mainly on polystyrene particles. Practical and ethical considerations limit research options in humans, and extrapolation from animal studies is challenging due to marked differences between species. Nevertheless, diverse in vitro and ex vivo human placental models exist e.g., plasma membrane vesicles, mono-culture and co-culture of placental cells, placenta-on-a-chip, villous tissue explants, and placental perfusion that can be used to advance this research area. The objective of this concise review is to recapitulate different human placental models, summarize the current understanding of placental uptake, transport and toxicity of MNPs and define knowledge gaps. Moreover, we provide perspectives for future research urgently needed to assess the potential hazards and risks of MNP exposure to maternal and fetal health.
Collapse
Affiliation(s)
- Hanna M Dusza
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Jeske van Boxel
- Amsterdam Institute for Life and Environment, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Majorie B M van Duursen
- Amsterdam Institute for Life and Environment, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Markus M Forsberg
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Juliette Legler
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Kirsi H Vähäkangas
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
12
|
Krausová M, Braun D, Buerki-Thurnherr T, Gundacker C, Schernhammer E, Wisgrill L, Warth B. Understanding the Chemical Exposome During Fetal Development and Early Childhood: A Review. Annu Rev Pharmacol Toxicol 2023; 63:517-540. [PMID: 36202091 DOI: 10.1146/annurev-pharmtox-051922-113350] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Early human life is considered a critical window of susceptibility to external exposures. Infants are exposed to a multitude of environmental factors, collectively referred to as the exposome. The chemical exposome can be summarized as the sum of all xenobiotics that humans are exposed to throughout a lifetime. We review different exposure classes and routes that impact fetal and infant metabolism and the potential toxicological role of mixture effects. We also discuss the progress in human biomonitoring and present possiblemodels for studying maternal-fetal transfer. Data gaps on prenatal and infant exposure to xenobiotic mixtures are identified and include natural biotoxins, in addition to commonly reported synthetic toxicants, to obtain a more holistic assessment of the chemical exposome. We highlight the lack of large-scale studies covering a broad range of xenobiotics. Several recommendations to advance our understanding of the early-life chemical exposome and the subsequent impact on health outcomes are proposed.
Collapse
Affiliation(s)
- Magdaléna Krausová
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Dominik Braun
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Tina Buerki-Thurnherr
- Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Particles Biology Interactions, St. Gallen, Switzerland;
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| | - Eva Schernhammer
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Center for Public Health, Department of Epidemiology, Medical University of Vienna, Vienna, Austria; .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Lukas Wisgrill
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria;
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , , .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| |
Collapse
|
13
|
Tse WH, Higgins S, Patel D, Xing M, West AR, Labouta HI, Keijzer R. The maternal-fetal transfer of passive immunity as a mechanism of transplacental nanoparticle drug delivery for prenatal therapies. Biomater Sci 2022; 10:5243-5253. [PMID: 35912636 DOI: 10.1039/d2bm00293k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoparticles administered into the maternal circulation and across the placenta are a potential clinical therapy to treat congenital diseases. The mechanism by which nanoparticles can safely cross the placenta for targeted drug delivery to the fetus remains poorly understood. We demonstrate that the maternal-fetal transfer of passive immunity through the neonatal Fc Receptor (FcRn) can induce the transplacental transfer of chitosan nanoparticles modifed with IgG antibodies (414 ± 27 nm). The transfer of FITC-tagged IgG-modified chitosan nanoparticles was 2.8 times higher (p = 0.0264) compared to similarly-sized unmodified chitosan nanoparticles (375 ± 17 nm). Co-administration of free IgG competitively diminished the transplacental transfer of IgG-modified nanoparticles, yet unmodified nanoparticles remained unaffected. Colocalization of the FcRn and the IgG-modified chitosan nanoparticles were observed with confocal microscopy. Barrier function before and after nanoparticle administration remained intact as determined by TEER (75-79 Ω cm2) and immmunofluorescence of ZO-1 tight junction proteins. The results provide insight into the clinical applications of nanoparticles for prenatal therapies using the mechanism of the maternal-fetal transfer of passive immunity.
Collapse
Affiliation(s)
- Wai Hei Tse
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| | - Sean Higgins
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada
| | - Daywin Patel
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, R3T 2N2, Manitoba, Canada
| | - Adrian R West
- Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| | - Hagar I Labouta
- Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Apotex Centre, College of Pharmacy, University of Manitoba, R3E 3T5, Manitoba, Canada
| | - Richard Keijzer
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| |
Collapse
|
14
|
Parameshwar PK, Sagrillo-Fagundes L, Azevedo Portilho N, Pastor WA, Vaillancourt C, Moraes C. Engineered models for placental toxicology: Emerging approaches based on tissue decellularization. Reprod Toxicol 2022; 112:148-159. [PMID: 35840119 DOI: 10.1016/j.reprotox.2022.07.003] [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: 04/02/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 11/28/2022]
Abstract
Recent increases in prescriptions and illegal drug use as well as exposure to environmental contaminants during pregnancy have highlighted the critical importance of placental toxicology in understanding and identifying risks to both mother and fetus. Although advantageous for basic science, current in vitro models often fail to capture the complexity of placental response, likely due to their inability to recreate and monitor aspects of the microenvironment including physical properties, mechanical forces and stiffness, protein composition, cell-cell interactions, soluble and physicochemical factors, and other exogenous cues. Tissue engineering holds great promise in addressing these challenges and provides an avenue to better understand basic biology, effects of toxic compounds and potential therapeutics. The key to success lies in effectively recreating the microenvironment. One strategy to do this would be to recreate individual components and then combine them. However, this becomes challenging due to variables present according to conditions such as tissue location, age, health status and lifestyle. The extracellular matrix (ECM) is known to influence cellular fate by working as a storage of factors. Decellularized ECM (dECM) is a recent tool that allows usage of the original ECM in a refurbished form, providing a relatively reliable representation of the microenvironment. This review focuses on using dECM in modified forms such as whole organs, scaffold sheets, electrospun nanofibers, hydrogels, 3D printing, and combinations as building blocks to recreate aspects of the microenvironment to address general tissue engineering and toxicology challenges, thus illustrating their potential as tools for future placental toxicology studies.
Collapse
Affiliation(s)
| | | | - Nathalia Azevedo Portilho
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada; Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - William A Pastor
- Department of Biochemistry, McGill University, Montréal, Québec, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
| | - Cathy Vaillancourt
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada; Department of Obstetrics and Gynecology, Université de Montréal, Montréal, Québec, Canada
| | - Christopher Moraes
- Department of Biological and Biomedical Engineering, McGill University, Montréal, Québec, Canada; Department of Chemical Engineering, McGill University, Montréal, Québec, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada; Division of Experimental Medicine, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
15
|
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
|
16
|
Bai X, Ran J, Zhao X, Liang Y, Yang X, Xi Y. The S100A10-AnxA2 complex is associated with the exocytosis of hepatitis B virus in intrauterine infection. J Transl Med 2022; 102:57-68. [PMID: 34645932 PMCID: PMC8512653 DOI: 10.1038/s41374-021-00681-8] [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: 04/13/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
Mother-to-child transmission (MTCT) is the major cause of chronic infection of hepatitis B virus (HBV) in patients. However, whether and how HBV crosses the placenta to cause infection in utero remains unclear. In this study, we investigate the mechanism as to how the HBV virions pass through layers of the trophoblast. Our data demonstrate the exocytosis of virions from the trophoblast after exposure to HBV where the endocytosed HBV virions co-localized with an S100A10/AnxA2 complex and LC3, an autophagosome membrane marker. Knockdown of either AnxA2 or S100A10 in trophoblast cells led to a reduction of the amount of exo-virus in Transwell assay. Immunohistochemistry also showed a high expression of AnxA2 and S100A10 in the placental tissue samples of HBV-infected mothers with congenital HBV-positive infants (HBV+/+). We conclude that in HBV intrauterine infection and mother-to-child transmission, a proportion of HBV hijacks autophagic protein secretion pathway and translocate across the trophoblast via S100A10/AnxA2 complex and multivesicular body (MVB)-mediated exocytosis. Our study provides a potential target for the interference of the mechanisms of HBV intrauterine infection and mother-to-child transmission.
Collapse
Grants
- National Natural Science Foundation of China (National Science Foundation of China)
- China’s National Key R&D Programs (NKPs) are a new category of projects created after the 2014 reform of the national STI funding system. They have incorporated numerous previously-existing programmes such as MOST’s “863 Programme” for R&D, “Programme 973” for basic research, Key Technologies R&D Programme, and International S&T Cooperation Programme; and NDRC and MIIT’s Industrial Technology R&D Fund. China’s National Key R&D Programmes support R&D in areas of social welfare and people’s livelihood, such as agriculture, energy and resources, environment, and health. They focus in particular on key and strategic technologies, featuring several well-targeted and defined objectives and deliverables to be achieved in a period ranging from three to five years, and reflecting a top-down and industry-university-research cooperation design which integrates basic research, technology application, demonstration and commercialisation.
Collapse
Affiliation(s)
- Xiaoxia Bai
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China.
| | - Jinshi Ran
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Xianlei Zhao
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Yun Liang
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
| | - Xiaohang Yang
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
- Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Yongmei Xi
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China.
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
17
|
Costa J, Mackay R, de Aguiar Greca SC, Corti A, Silva E, Karteris E, Ahluwalia A. The Role of the 3Rs for Understanding and Modeling the Human Placenta. J Clin Med 2021; 10:jcm10153444. [PMID: 34362227 PMCID: PMC8347836 DOI: 10.3390/jcm10153444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Modeling the physiology of the human placenta is still a challenge, despite the great number of scientific advancements made in the field. Animal models cannot fully replicate the structure and function of the human placenta and pose ethical and financial hurdles. In addition, increasingly stricter animal welfare legislation worldwide is incentivizing the use of 3R (reduction, refinement, replacement) practices. What efforts have been made to develop alternative models for the placenta so far? How effective are they? How can we improve them to make them more predictive of human pathophysiology? To address these questions, this review aims at presenting and discussing the current models used to study phenomena at the placenta level: in vivo, ex vivo, in vitro and in silico. We describe the main achievements and opportunities for improvement of each type of model and critically assess their individual and collective impact on the pursuit of predictive studies of the placenta in line with the 3Rs and European legislation.
Collapse
Affiliation(s)
- Joana Costa
- Centro di Ricerca E.Piaggio, University of Pisa, 56126 Pisa, Italy; (J.C.); (A.C.)
| | - Ruth Mackay
- Centre for Genome Engineering and Maintenance, Department of Mechanical and Aerospace Engineering, Brunel University London, Uxbridge UB8 3PH, UK;
| | | | - Alessandro Corti
- Centro di Ricerca E.Piaggio, University of Pisa, 56126 Pisa, Italy; (J.C.); (A.C.)
- Department of Translational Medicine, University of Pisa, 56126 Pisa, Italy
| | - Elisabete Silva
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (S.-C.d.A.G.); (E.S.); (E.K.)
| | - Emmanouil Karteris
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (S.-C.d.A.G.); (E.S.); (E.K.)
| | - Arti Ahluwalia
- Centro di Ricerca E.Piaggio, University of Pisa, 56126 Pisa, Italy; (J.C.); (A.C.)
- Department of Information Engineering, University of Pisa, 56122 Pisa, Italy
- Interuniversity Centro for the Promotion of 3Rs Principles in Teaching and Research (Centro3R), Italy
- Correspondence:
| |
Collapse
|
18
|
Wong MK, Li EW, Adam M, Selvaganapathy PR, Raha S. Establishment of an in vitro placental barrier model cultured under physiologically relevant oxygen levels. Mol Hum Reprod 2021; 26:353-365. [PMID: 32159799 DOI: 10.1093/molehr/gaaa018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
The human placental barrier facilitates many key functions during pregnancy, most notably the exchange of all substances between the mother and fetus. However, preclinical models of the placental barrier often lacked the multiple cell layers, syncytialization of the trophoblast cells and the low oxygen levels that are present within the body. Therefore, we aimed to design and develop an in vitro model of the placental barrier that would reinstate these factors and enable improved investigations of barrier function. BeWo placental trophoblastic cells and human umbilical vein endothelial cells were co-cultured on contralateral sides of an extracellular matrix-coated transwell insert to establish a multilayered barrier. Epidermal growth factor and forskolin led to significantly increased multi-nucleation of the BeWo cell layer and increased biochemical markers of syncytial fusion, for example syncytin-1 and hCGβ. Our in vitro placental barrier possessed size-specific permeability, with 4000-Da molecules experiencing greater transport and a lower apparent permeability coefficient than 70 000-Da molecules. We further demonstrated that the BeWo layer had greater resistance to smaller molecules compared to the endothelial layer. Chronic, physiologically low oxygen exposure (3-8%) increased the expression of hypoxia-inducible factor 1α and syncytin-1, further increased multi-nucleation of the BeWo cell layer and decreased barrier permeability only against smaller molecules (457 Da/4000 Da). In conclusion, we built a novel in vitro co-culture model of the placental barrier that possessed size-specific permeability and could function under physiologically low oxygen levels. Importantly, this will enable future researchers to better study the maternal-fetal transport of nutrients and drugs during pregnancy.
Collapse
Affiliation(s)
- Michael K Wong
- Graduate Program of Medical Science, McMaster University, Hamilton, Ontario, Canada.,Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Edward W Li
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Mohamed Adam
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Sandeep Raha
- Graduate Program of Medical Science, McMaster University, Hamilton, Ontario, Canada.,Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| |
Collapse
|
19
|
Boos JA, Misun PM, Brunoldi G, Furer LA, Aengenheister L, Modena M, Rousset N, Buerki-Thurnherr T, Hierlemann A. Microfluidic Co-Culture Platform to Recapitulate the Maternal-Placental-Embryonic Axis. Adv Biol (Weinh) 2021; 5:e2100609. [PMID: 34145989 DOI: 10.1002/adbi.202100609] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/28/2021] [Indexed: 12/22/2022]
Abstract
Safety assessment of the effects of developmental toxicants on pregnant women is challenging, and systemic effects in embryo-maternal interactions are largely unknown. However, most developmental toxicity studies rely on animal trials, while in vitro platforms that recapitulate the maternal-placental-embryonic axis are missing. Here, the development of a dedicated microfluidic device for co-cultivation of a placental barrier and 3D embryoid bodies to enable systemic toxicity testing at the embryo-maternal interface is reported. The microfluidic platform features simple handling and recuperation of both tissue models, which facilitates post-hoc in-depth analysis at the tissue and single-cell level. Gravity-driven flow enables inter-tissue communication through the liquid phase as well as simple and robust operation and renders the platform parallelizable. As a proof of concept and to demonstrate platform use for systemic embryotoxicity testing in vitro, maternal exposure to plastic microparticles is emulated, and microparticle effects on the embryo-placental co-culture are investigated.
Collapse
Affiliation(s)
- Julia A Boos
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Patrick M Misun
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Giulia Brunoldi
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Lea A Furer
- Particles@Barriers Group, Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen, 9014, Switzerland
| | - Leonie Aengenheister
- Particles@Barriers Group, Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen, 9014, Switzerland
| | - Mario Modena
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Nassim Rousset
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Tina Buerki-Thurnherr
- Particles@Barriers Group, Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen, 9014, Switzerland
| | - Andreas Hierlemann
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| |
Collapse
|
20
|
Counter-directed leucine gradient promotes amino acid transfer across the human placenta. J Nutr Biochem 2021; 96:108760. [PMID: 33964466 DOI: 10.1016/j.jnutbio.2021.108760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/04/2021] [Accepted: 04/16/2021] [Indexed: 02/01/2023]
Abstract
The developing fetus is highly vulnerable to imbalances in the supply of essential amino acids (AA). Transplacental AA transfer depends on complex interactions between accumulative transporters, exchangers and facilitators, which maintain both intra-extracellular and materno-fetal substrate gradients. We determined physiological AA gradients between maternal and fetal blood and assessed their importance by studying maternal-fetal leucine transfer in human trophoblasts. Maternal-venous and corresponding fetal-arterial/fetal-venous sera were collected from 22 healthy patients at partum. The acquisition of the full AA spectra in serum was performed by ion exchange chromatography. Physiological materno-fetal AA levels were evaluated using paired two-way ANOVA with Tukey's correction. AA concentrations and gradients were tested for associations with anthropometric data by Spearman correlation analysis. Functional effects of a physiological leucine gradient versus equimolar concentrations were tested in BeWo cells using L-[3H]-leucine in conventional and Transwell-based uptake and transfer experiments. The LAT1/SLC7A5-specific inhibitor JPH203 was used to evaluate LAT1-transporter-mediated leucine transport. Maternal AA concentrations correlated with preconceptional and maternal weights at partum. Interestingly, low materno-fetal AA gradients were associated with maternal weight, BMI and gestational weight gain. Leucine uptake was promoted by increased extracellular substrate concentrations. Materno-fetal leucine transfer was significantly increased against a 137µM leucine gradient demonstrating that transplacental leucine transport is stimulated by a counter-directed gradient. Moreover, leucine transfer was inhibited by 10µM JPH203 confirming that Leu transport across the trophoblast monolayer is LAT1-dependent. This study demonstrates a currently underestimated effect of transplacental AA gradients on efficient leucine transfer which could severely affect fetal development.
Collapse
|
21
|
The polarized localization of lipoprotein receptors and cholesterol transporters in the syncytiotrophoblast of the placenta is reproducible in a monolayer of primary human trophoblasts. Placenta 2021; 105:50-60. [PMID: 33548684 DOI: 10.1016/j.placenta.2021.01.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/29/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The uptake of low- and high-density lipoproteins (LDL and HDL) through the LDL receptor (LDLR) and the scavenger receptor class B type I (SR-BI) mediates maternal to fetal cholesterol transfer in syncytiotrophoblast (STB) cells. STB cells deliver cholesterol via cholesterol efflux through the ATP-binding cassette transporters A1 (ABCA1, to ApoA-I), G1 (ABCG1, to HDL), and SR-BI (to HDL). In the human placenta, these proteins are localized in the apical (LDLR, SR-BI, ABCA1) and basal (SR-BI, ABCA1, ABCG1) membrane of STB cells. However, whether these proteins in polarized primary culture models of STB show a similar localization to those in the human placenta is currently unknown. METHODS Primary human trophoblasts (PHT) were isolated from normal placentas and cultured in Transwells® with Matrigel to obtain a polarized STB monolayer, proteins were determined by immunofluorescence and cholesterol efflux determined to different acceptors. RESULTS At day 5, LDLR and ABCA1 localized mainly in the apical membrane, ABCG1 in the basal membrane, and SR-BI in both. Cholesterol efflux towards the apical compartment was higher to adult and neonatal HDL compared to ApoA-I. When acceptors were added in the basal compartment, cholesterol was retained in the Matrigel. DISCUSSION Polarized STB monolayers express LDLR, SR-BI, ABCA1 and ABCG1, and their apical/basal localization resembles the one described in human placental tissue. This study confirms the high physiological value and suitability of this model for use in functional studies. Our findings also suggest that ABCA1 and SR-BI participate in cholesterol efflux to the maternal side of the cells.
Collapse
|
22
|
Aengenheister L, Favaro RR, Morales-Prieto DM, Furer LA, Gruber M, Wadsack C, Markert UR, Buerki-Thurnherr T. Research on nanoparticles in human perfused placenta: State of the art and perspectives. Placenta 2020; 104:199-207. [PMID: 33418345 DOI: 10.1016/j.placenta.2020.12.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022]
Abstract
Increasing human exposure to nanoparticles (NPs) from various sources raises concerns for public health, especially for vulnerable risk groups like pregnant women and their developing fetuses. However, nanomedicine and the prospect of creating safe and effective NP-based formulations of drugs hold great promise to revolutionize treatment during pregnancy. With maternal and fetal health at stake, risks and opportunities of NPs in pregnancy need to be carefully investigated. Importantly, a comprehensive understanding of NP transport and effects at the placenta is urgently needed considering the central position of the placenta at the maternal-fetal interface and its many essential functions to enable successful pregnancy. The perfusion of human placental tissue provides a great opportunity to achieve predictive human relevant insights, circumventing uncertainties due to considerable differences in placental structure and function across species. Here, we have reviewed the current literature on the ex vivo human placenta perfusion of NPs. From 16 available studies, it was evident that placental uptake and transfer of NPs are highly dependent on their characteristics like size and surface modifications, which is in line with previous observations from in vitro and animal transport studies. These studies further revealed that special considerations apply for the perfusion of NPs and we identified relevant controls that should be implemented in future perfusion studies. While current studies mostly focused on placental transfer of NPs to conclude on potential fetal exposure, the ex vivo placental perfusion model has considerable potential to reveal novel insights on NP effects on placental tissue functionality and signaling that could indirectly affect maternal-fetal health.
Collapse
Affiliation(s)
- Leonie Aengenheister
- Laboratory for Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland; Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Rodolfo R Favaro
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Diana M Morales-Prieto
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Lea A Furer
- Laboratory for Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Michael Gruber
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036, Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036, Graz, Austria
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Tina Buerki-Thurnherr
- Laboratory for Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland.
| |
Collapse
|
23
|
Zaugg J, Huang X, Ziegler F, Rubin M, Graff J, Müller J, Moser-Hässig R, Powell T, Gertsch J, Altmann KH, Albrecht C. Small molecule inhibitors provide insights into the relevance of LAT1 and LAT2 in materno-foetal amino acid transport. J Cell Mol Med 2020; 24:12681-12693. [PMID: 33001560 PMCID: PMC7687008 DOI: 10.1111/jcmm.15840] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/20/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
The placenta supplies the foetus with critical nutrients such as essential amino acids (AA, eg leucine) for development and growth. It also represents a cellular barrier which is formed by a polarized, differentiated syncytiotrophoblast (STB) monolayer. Active Na+‐independent leucine transport across the placenta is mainly attributed to the System L transporters LAT1/SLC7A5 and LAT2/SLC7A8. This study explored the influence of trophoblast differentiation on the activity of LAT1/LAT2 and the relevance of LAT1/LAT2 in leucine uptake and transfer in trophoblasts by applying specific small molecule inhibitors (JPH203/JG336/JX009). L‐leucine uptake (total dose = 167 μmol/L) was sensitive to LAT1‐specific inhibition by JPH203 (EC50 = 2.55 µmol/L). The inhibition efficiency of JPH203 was increased by an additional methoxy group in the JPH203‐derivate JG336 (EC50 = 1.99 µmol/L). Interestingly, JX009 showed efficient System L inhibition (EC50 = 2.35 µmol/L) and was the most potent inhibitor of leucine uptake in trophoblasts. The application of JPH203 and JX009 in Transwell®‐based leucine transfer revealed LAT1 as the major accumulative transporter at the apical membrane, but other System L transporters such as LAT2 as rate‐limiting for leucine efflux across the basal membrane. Therefore, differential specificity of the applied inhibitors allowed for estimation of the contribution of LAT1 and LAT2 in materno‐foetal AA transfer and their potential impact in pregnancy diseases associated with impaired foetal growth.
Collapse
Affiliation(s)
- Jonas Zaugg
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Xiao Huang
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Fabian Ziegler
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Matthias Rubin
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Julien Graff
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland.,Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Jennifer Müller
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland.,Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Ruedi Moser-Hässig
- Division of Gynecology and Obstetrics, Lindenhofgruppe, Bern, Switzerland
| | - Theresa Powell
- Department of Pediatrics, Neonatology Section, University of Colorado, Denver, CO, USA
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Karl-Heinz Altmann
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland.,Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Kreuder AE, Bolaños-Rosales A, Palmer C, Thomas A, Geiger MA, Lam T, Amler AK, Markert UR, Lauster R, Kloke L. Inspired by the human placenta: a novel 3D bioprinted membrane system to create barrier models. Sci Rep 2020; 10:15606. [PMID: 32973223 PMCID: PMC7515925 DOI: 10.1038/s41598-020-72559-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022] Open
Abstract
Barrier organ models need a scaffold structure to create a two compartment culture. Technical filter membranes used most often as scaffolds may impact cell behaviour and present a barrier themselves, ultimately limiting transferability of test results. In this work we present an alternative for technical filter membrane systems: a 3D bioprinted biological membrane in 24 well format. The biological membrane, based on extracellular matrix (ECM), is highly permeable and presents a natural 3D environment for cell culture. Inspired by the human placenta we established a coculture of a trophoblast-derived cell line (BeWo b30), together with primary placental fibroblasts within the biological membrane (simulating villous stroma) and primary human placental endothelial cells-representing three cellular components of the human placental villus. All cell types maintained their cell type specific marker expression after two weeks of coculture on the biological membrane. In permeability assays the trophoblast layer developed a barrier on the biological membrane, which was even more pronounced when cocultured with fibroblasts. In this work we present a filter membrane free scaffold, we characterize its properties and assess its suitability for cell culture and barrier models. Further we show a novel placenta inspired model in a complex bioprinted coculture. In the absence of an artificial filter membrane, we demonstrate barrier architecture and functionality.
Collapse
Affiliation(s)
- Anna-Elisabeth Kreuder
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany.
- Cellbricks GmbH, Berlin, 13355, Germany.
| | - Aramis Bolaños-Rosales
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
- Cellbricks GmbH, Berlin, 13355, Germany
| | | | - Alexander Thomas
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
- Cellbricks GmbH, Berlin, 13355, Germany
| | | | | | - Anna-Klara Amler
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
- Cellbricks GmbH, Berlin, 13355, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, 07747, Jena, Germany
| | - Roland Lauster
- Medical Biotechnology, Technical University of Berlin, Berlin, 13355, Germany
| | - Lutz Kloke
- Cellbricks GmbH, Berlin, 13355, Germany.
| |
Collapse
|
25
|
Karahoda R, Abad C, Horackova H, Kastner P, Zaugg J, Cerveny L, Kucera R, Albrecht C, Staud F. Dynamics of Tryptophan Metabolic Pathways in Human Placenta and Placental-Derived Cells: Effect of Gestation Age and Trophoblast Differentiation. Front Cell Dev Biol 2020; 8:574034. [PMID: 33072756 PMCID: PMC7530341 DOI: 10.3389/fcell.2020.574034] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
L-Tryptophan is an essential amino acid and a precursor of several physiologically active metabolites. In the placenta, the serotonin and kynurenine metabolic pathways of tryptophan metabolism have been identified, giving rise to various molecules of neuroactive or immunoprotective properties, such as serotonin, melatonin, kynurenine, kynurenic acid, or quinolinic acid. Current literature suggests that optimal levels of these molecules in the fetoplacental unit are crucial for proper placenta functions, fetal development and programming. Placenta is a unique endocrine organ that, being equipped with a battery of biotransformation enzymes and transporters, precisely orchestrates homeostasis of tryptophan metabolic pathways. However, because pregnancy is a dynamic process and placental/fetal needs are continuously changing throughout gestation, placenta must adapt to these changes and ensure proper communication in the feto-placental unit. Therefore, in this study we investigated alterations of placental tryptophan metabolic pathways throughout gestation. Quantitative polymerase chain reaction (PCR) analysis of 21 selected genes was carried out in first trimester (n = 13) and term (n = 32) placentas. Heatmap analysis with hierarchical clustering revealed differential gene expression of serotonin and kynurenine pathways across gestation. Subsequently, digital droplet PCR, Western blot, and functional analyses of the rate-limiting enzymes suggest preferential serotonin synthesis early in pregnancy with a switch to kynurenine production toward term. Correspondingly, increased function and/or protein expression of serotonin degrading enzyme and transporters at term indicates efficient placental uptake and metabolic degradation of serotonin. Lastly, gene expression analysis in choriocarcinoma-derived cell lines (BeWo, BeWo b30, JEG-3) revealed dissimilar expression patterns and divergent effect of syncytialization compared to primary trophoblast cells isolated from human term placentas; these findings show that the commonly used in vitro placental models are not suitable to study placental handling of tryptophan. Altogether, our data provide the first comprehensive evidence of changes in placental homeostasis of tryptophan and its metabolites as a function of gestational age, which is critical for proper placental function and fetal development.
Collapse
Affiliation(s)
- Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Hana Horackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Petr Kastner
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Jonas Zaugg
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Lukas Cerveny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Radim Kucera
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.,Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| |
Collapse
|
26
|
Zubizarreta ME, Xiao S. Bioengineering models of female reproduction. Biodes Manuf 2020; 3:237-251. [PMID: 32774987 PMCID: PMC7413245 DOI: 10.1007/s42242-020-00082-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 05/23/2020] [Indexed: 12/25/2022]
Abstract
The female reproductive system consists of the ovaries, the female gonads, and the reproductive track organs of the fallopian tubes, uterus, cervix, and vagina. It functions to provide hormonal support and anatomical structure for the production of new offspring. A number of endogenous and exogenous factors can impact female reproductive health and fertility, including genetic vulnerability, medications, environmental exposures, age, nutrition, and diseases, etc. To date, due to the ethical concerns of using human subjects in biomedical research, the majority of studies use in vivo animal models and 2D cell/tissue culture models to study female reproduction. However, the complexity and species difference of the female reproductive system in humans makes it difficult to compare to those of animals. Moreover, the monolayered cells cultured on flat plastics or glass lose their 3D architecture as well as the physical and/or biochemical contacts with other cells in vivo. Further, all reproductive organs do not work alone but interconnect with each other and also with non-reproductive organs to support female reproductive, endocrine, and systemic health. These facts suggest that there is an urgent and unmet need to develop representative, effective, and efficient in vitro models for studying human female reproduction. The prodigious advancements of bioengineering (e.g. biomaterials, 3D printing, and organ-on-a-chip) allow us to study female reproduction in an entirely new way. Here, we review recent advances that use bioengineering methods to study female reproduction, including the bioengineering models of the ovary, fallopian tube, uterus, embryo implantation, placenta, and reproductive disease.
Collapse
Affiliation(s)
- Maria E. Zubizarreta
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Shuo Xiao
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental Health Sciences Institute, Rutgers University, Piscataway, NJ, 08854, USA
| |
Collapse
|
27
|
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
|
28
|
Rothbauer M, Eilenberger C, Spitz S, Bachmann B, Pajenda J, Schwaighofer A, Höll G, Helmke PS, Kohl Y, Lendl B, Ertl P. FTIR spectroscopy as a novel analytical approach for investigation of glucose transport and glucose transport inhibition studies in transwell in vitro barrier models. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 237:118388. [PMID: 32361318 DOI: 10.1016/j.saa.2020.118388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 06/11/2023]
Abstract
Glucose transport is key for cellular metabolism as well as physiological function and is maintained via passive facilitated and active sodium-glucose linked transport routes. Here, we present for the first time Fourier-transform infrared spectroscopy as a novel approach for quantification of apical-to-basolateral glucose transport of in vitro cell barrier models using liver, lung, intestinal and placental cancer cell lines. Results of our comparative study revealed that distinct differences could be observed upon subjection to transport inhibitors.
Collapse
Affiliation(s)
- Mario Rothbauer
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria.
| | - Christoph Eilenberger
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Sarah Spitz
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Barbara Bachmann
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Jasmin Pajenda
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Andreas Schwaighofer
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria
| | - Gregor Höll
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Palle Steen Helmke
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering, 66280 Sulzbach, Germany
| | - Bernhard Lendl
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria
| | - Peter Ertl
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria.
| |
Collapse
|
29
|
Gundacker C, Ellinger I. The unique applicability of the human placenta to the Adverse Outcome Pathway (AOP) concept: the placenta provides fundamental insights into human organ functions at multiple levels of biological organization. Reprod Toxicol 2020; 96:273-281. [PMID: 32768559 DOI: 10.1016/j.reprotox.2020.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022]
Abstract
Despite the short lifespan of the human placenta, the proper formation and function of the organ is of crucial importance for fetal development. Placental dysfunction increases the risk of complications for mother and child during pregnancy and childbirth and beyond as it predisposes to fetal programming. The placenta is an upstream organ of the fetus. It performs the functions of fetal lungs, liver, intestines, kidneys and glands as long as these organs are not fully functional. Furthermore, it is the only human organ that is non-invasively available either after elective abortion or after birth. This is a crucial point given that the conceptual framework of Adverse Outcome Pathway (AOP) requires data on organ function. In vitro and ex vivo placental studies, combined with epidemiological and clinical data on pregnant women, newborns, and infants can uniquely cover all levels of information needed to develop new AOPs and complement existing AOPs related to reproductive toxicity and beyond. To stimulate further research in this area and to support researchers in future studies dealing with the development of AOPs related to the placenta, this review first gives a brief description of placental structure, placental development and relevant pregnancy diseases. The state of knowledge about the available placental models, their particularities and limitations are briefly discussed. Finally, the use of placental research for the development of AOPs is presented with an illustrative example.
Collapse
Affiliation(s)
- Claudia Gundacker
- Institute of Medical Genetics, Medical University Vienna, Vienna, Austria.
| | - Isabella Ellinger
- Institute for Pathophysiology and Allergy Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
30
|
Li Z, Kurosawa O, Iwata H. A Novel Human Placental Barrier Model Based on Trophoblast Stem Cells Derived from Human Induced Pluripotent Stem Cells. Tissue Eng Part A 2020; 26:780-791. [PMID: 32323636 DOI: 10.1089/ten.tea.2019.0342] [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] [Indexed: 12/29/2022] Open
Abstract
The placenta acts as an interface between the fetus and the expecting mother. Various drugs and environmental pollutants can pass through the human placental barrier and may harm the developing fetus. Currently available in vitro placental barrier models are often inadequate, because they are lacking the functional trophoblast cells. Therefore, we developed and characterized a new human placental model using trophoblast stem cells (TSCs) derived from human induced pluripotent stem cells. Umbilical vein endothelial cells, fibroblast, and TSCs were cocultured using micromesh cell culture technique. These cells formed a tight three-layered structure. This coculture model induced progressive fusion of TSCs and formed a syncytialized epithelium that resembles the in vivo syncytiotrophoblast. Our model allowed the cultured trophoblasts to form microvilli and to reconstitute expression and physiological localization of membrane transport proteins, such as transporter for ATP-binding cassette subfamily B member 1, ATP-binding cassette subfamily C member 3, and glucose transporter-1. Drug permeability assays were performed using five compounds. The results from the permeability assays were comparable to the ones obtained with ex vivo placental models. In conclusion, we developed a novel coculture model mimicking human placenta that provides a useful tool for the studies on transfer of substances between the mother and fetus. Impact statement Compared with the currently available in vitro placental barrier models, a novel three-dimensional coculture placental barrier model presented in this study morphologically and functionally modeled the true placental barrier. The use of human trophoblast stem cells from human induced pluripotent stem cells substantially improved the current model. The use of micromesh sheet as a bioscaffold facilitated the formation of a good multilayer structure, which is closer to the physical appearance of the placenta observed in human.
Collapse
Affiliation(s)
- Zhuosi Li
- The "Compass to Healthy Life" Research Complex Program, RIKEN Institute, Kobe, Japan
| | - Osamu Kurosawa
- The "Compass to Healthy Life" Research Complex Program, RIKEN Institute, Kobe, Japan
| | - Hiroo Iwata
- The "Compass to Healthy Life" Research Complex Program, RIKEN Institute, Kobe, Japan.,Research Promotion Institution for COI Site, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Mishra JS, Zhao H, Hattis S, Kumar S. Elevated Glucose and Insulin Levels Decrease DHA Transfer across Human Trophoblasts via SIRT1-Dependent Mechanism. Nutrients 2020; 12:nu12051271. [PMID: 32365792 PMCID: PMC7284516 DOI: 10.3390/nu12051271] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/30/2022] Open
Abstract
Gestational diabetes mellitus (GDM) results in reduced docosahexaenoic acid (DHA) transfer to the fetus, likely due to placental dysfunction. Sirtuin-1 (SIRT1) is a nutrient sensor and regulator of lipid metabolism. This study investigated whether the high glucose and insulin condition of GDM regulates DHA transfer and expression of fatty acid transporters and if this effect is related to SIRT1 expression and function. Syncytialized primary human trophoblasts were treated with and without glucose (25 mmol/L) and insulin (10-7 mol/L) for 72 h to mimic the insulin-resistance conditions of GDM pregnancies. In control conditions, DHA transfer across trophoblasts increased in a time- and dose-dependent manner. Exposure to GDM conditions significantly decreased DHA transfer, but increased triglyceride accumulation and fatty acid transporter expression (CD36, FABP3, and FABP4). GDM conditions significantly suppressed SIRT1 mRNA and protein expression. The SIRT1 inhibitor decreased DHA transfer across control trophoblasts, and recombinant SIRT1 and SIRT1 activators restored the decreased DHA transport induced by GDM conditions. The results demonstrate a novel role of SIRT1 in the regulation of DHA transfer across trophoblasts. The suppressed SIRT1 expression and the resultant decrease in placental DHA transfer caused by high glucose and insulin levels suggest new insights of molecular mechanisms linking GDM to fetal DHA deficiency.
Collapse
Affiliation(s)
- Jay S. Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
| | - Hanjie Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
| | - Sari Hattis
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
- Correspondence: ; Tel.: +1-608-265-1046
| |
Collapse
|
32
|
Cholesterol uptake and efflux are impaired in human trophoblast cells from pregnancies with maternal supraphysiological hypercholesterolemia. Sci Rep 2020; 10:5264. [PMID: 32210256 PMCID: PMC7093446 DOI: 10.1038/s41598-020-61629-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Maternal physiological (MPH) or supraphysiological hypercholesterolaemia (MSPH) occurs during pregnancy. Cholesterol trafficking from maternal to foetal circulation requires the uptake of maternal LDL and HDL by syncytiotrophoblast and cholesterol efflux from this multinucleated tissue to ApoA-I and HDL. We aimed to determine the effects of MSPH on placental cholesterol trafficking. Placental tissue and primary human trophoblast (PHT) were isolated from pregnant women with total cholesterol <280 md/dL (MPH, n = 27) or ≥280 md/dL (MSPH, n = 28). The lipid profile in umbilical cord blood from MPH and MSPH neonates was similar. The abundance of LDL receptor (LDLR) and HDL receptor (SR-BI) was comparable between MSPH and MPH placentas. However, LDLR was localized mainly in the syncytiotrophoblast surface and was associated with reduced placental levels of its ligand ApoB. In PHT from MSPH, the uptake of LDL and HDL was lower compared to MPH, without changes in LDLR and reduced levels of SR-BI. Regarding cholesterol efflux, in MSPH placentas, the abundance of cholesterol transporter ABCA1 was increased, while ABCG1 and SR-BI were reduced. In PHT from MSPH, the cholesterol efflux to ApoA-I was increased and to HDL was reduced, along with reduced levels of ABCG1, compared to MPH. Inhibition of SR-BI did not change cholesterol efflux in PHT. The TC content in PHT was comparable in MPH and MSPH cells. However, free cholesterol was increased in MSPH cells. We conclude that MSPH alters the trafficking and content of cholesterol in placental trophoblasts, which could be associated with changes in the placenta-mediated maternal-to-foetal cholesterol trafficking.
Collapse
|
33
|
Renggli K, Rousset N, Lohasz C, Nguyen OTP, Hierlemann A. Integrated Microphysiological Systems: Transferable Organ Models and Recirculating Flow. ADVANCED BIOSYSTEMS 2019; 3:e1900018. [PMID: 32627410 PMCID: PMC7610576 DOI: 10.1002/adbi.201900018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Studying and understanding of tissue and disease mechanisms largely depend on the availability of suitable and representative biological model systems. These model systems should be carefully engineered and faithfully reproduce the biological system of interest to understand physiological effects, pharmacokinetics, and toxicity to better identify new drug compounds. By relying on microfluidics, microphysiological systems (MPSs) enable the precise control of culturing conditions and connections of advanced in vitro 3D organ models that better reproduce in vivo environments. This review focuses on transferable in vitro organ models and integrated MPSs that host these transferable biological units and enable interactions between different tissue types. Interchangeable and transferrable in vitro organ models allow for independent quality control of the biological model before system assembly and building MPS assays on demand. Due to the complexity and different maturation times of individual in vitro tissues, off-chip production and quality control entail improved stability and reproducibility of the systems and results, which is important for large-scale adoption of the technology. Lastly, the technical and biological challenges and open issues for realizing and implementing integrated MPSs with transferable in vitro organ models are discussed.
Collapse
Affiliation(s)
- Kasper Renggli
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | | | | | | |
Collapse
|
34
|
Placental secretion of apolipoprotein A1 and E: the anti-atherogenic impact of the placenta. Sci Rep 2019; 9:6225. [PMID: 30996342 PMCID: PMC6470155 DOI: 10.1038/s41598-019-42522-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
High levels of atherogenic lipids in pregnancy are associated with health complications for the mother, the fetus and the newborn. As endocrine secretory tissue, the human placenta releases apolipoproteins (apos), particularly apoA1 and apoE. However, the magnitude and the directionality of the apo secretions remain unknown. We aimed to 1) determine the amount and orientation (apical-maternal versus basal-fetal) of placentally secreted apoA1 and apoE using human perfused placenta and primary trophoblast cell (PTC) culture, 2) compare apoA1 and apoE secretions of PTC with that of hepatocytes and 3) associate the obtained results with human blood levels by determining apoA1 and apoE concentrations in maternal and fetal serum samples. In perfused placenta and serum samples, apoA1 and apoE concentrations were significantly higher at the maternal compared to the fetal side. For apoE a similar trend was found in PTC. For apoA1, the secretion to the apical side declined over time while release to the basal side was stable resulting in significantly different apoA1 concentrations between both sides. Unexpectedly, PTC secreted significantly higher amounts of apoA1 and apoE compared to hepatocytes. Our data indicate that the placenta may play an important role in maternal and fetal cholesterol homeostasis via secretion of anti-atherogenic apos.
Collapse
|
35
|
Transcriptomic analysis of the interaction of choriocarcinoma spheroids with receptive vs. non-receptive endometrial epithelium cell lines: an in vitro model for human implantation. J Assist Reprod Genet 2019; 36:857-873. [PMID: 30972518 DOI: 10.1007/s10815-019-01442-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/19/2019] [Indexed: 10/27/2022] Open
Abstract
PURPOSE Several in vitro systems have been reported to model human implantation; however, the molecular dynamics of the trophoblast vs. the epithelial substrate during attachment have not been described. We have established an in vitro model which allowed us to dissect the transcriptional responses of the trophoblast and the receptive vs. non-receptive epithelium after co-culture. METHODS We established an in vitro system based on co-culture of (a) immortalized cells representing receptive (Ishikawa) or non-receptive (HEC-1-A) endometrial epithelium with (b) spheroids of a trophoblastic cell line (JEG-3) modified to express green fluorescent protein (GFP). After 48 h of co-culture, GFP+ (trophoblast cells) and GFP- cell fractions (receptive or non-receptive epithelial cells) were isolated by fluorescence-activated flow cytometry (FACS) and subjected to RNA-seq profiling and gene set enrichment analysis (GSEA). RESULTS Compared to HEC-1-A, the trophoblast challenge to Ishikawa cells differentially regulated the expression of 495 genes, which mainly involved cell adhesion and extracellular matrix (ECM) molecules. GSEA revealed enrichment of pathways related to cell division, cell cycle regulation, and metabolism in the Ishikawa substrate. Comparing the gene expression profile of trophoblast spheroids revealed that 1877 and 323 genes were upregulated or downregulated when co-cultured on Ishikawa substrates (compared to HEC-1-A), respectively. Pathways favorable to development, including tissue remodeling, organogenesis, and angiogenesis, were enhanced in the trophoblast compartment after co-culture of spheroids with receptive epithelium. By contrast, the co-culture with less receptive epithelium enriched pathways mainly related to trophoblast cell proliferation and cell cycle regulation. CONCLUSIONS Endometrial receptivity requires a transcriptional signature that determines the trophoblast response and drives attachment.
Collapse
|
36
|
When 3 Rs meet a forth R: Replacement, reduction and refinement of animals in research on reproduction. J Reprod Immunol 2019; 132:54-59. [DOI: 10.1016/j.jri.2019.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
|
37
|
Tamò L, Hibaoui Y, Kallol S, Alves MP, Albrecht C, Hostettler KE, Feki A, Rougier JS, Abriel H, Knudsen L, Gazdhar A, Geiser T. Generation of an alveolar epithelial type II cell line from induced pluripotent stem cells. Am J Physiol Lung Cell Mol Physiol 2018; 315:L921-L932. [DOI: 10.1152/ajplung.00357.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Differentiation of primary alveolar type II epithelial cells (AEC II) to AEC type I in culture is a major barrier in the study of the alveolar epithelium in vitro. The establishment of an AEC II cell line derived from induced pluripotent stem cells (iPSC) represents a novel opportunity to study alveolar epithelial cell biology, for instance, in the context of lung injury, fibrosis, and repair. In the present study, we generated long-lasting AEC II from iPSC (LL-iPSC-AEC II). LL-iPSC-AEC II displayed morphological characteristics of AEC II, including growth in a cobblestone monolayer, the presence of lamellar bodies, and microvilli, as shown by electron microscopy. Also, LL-iPSC-AEC II expressed AEC type II proteins, such as cytokeratin, surfactant protein C, and LysoTracker DND 26 (a marker for lamellar bodies). Furthermore, the LL-iPSC-AEC II exhibited functional properties of AEC II by an increase of transepithelial electrical resistance over time, secretion of inflammatory mediators in biologically relevant quantities (IL-6 and IL-8), and efficient in vitro alveolar epithelial wound repair. Consistent with the AEC II phenotype, the cell line showed the ability to uptake and release surfactant protein B, to secrete phospholipids, and to differentiate into AEC type I. In summary, we established a long-lasting, but finite AEC type II cell line derived from iPSC as a novel cellular model to study alveolar epithelial cell biology in lung health and disease.
Collapse
Affiliation(s)
- Luca Tamò
- Department of Pulmonary Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Youssef Hibaoui
- Department of Gynecology and Obstetrics, University Hospital Geneva, Geneva, Switzerland
| | - Sampada Kallol
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Marco P. Alves
- Department of Pediatric Pneumology, University Hospital Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Virology and Immunology, Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Swiss National Center of Competence in Research, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Katrin E. Hostettler
- Clinics of Respiratory Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anis Feki
- Department of Gynecology and Obstetrics, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | | | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Swiss National Center of Competence in Research, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
38
|
Kallol S, Moser-Haessig R, Ontsouka CE, Albrecht C. Comparative expression patterns of selected membrane transporters in differentiated BeWo and human primary trophoblast cells. Placenta 2018; 72-73:48-52. [PMID: 30501881 DOI: 10.1016/j.placenta.2018.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/25/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023]
Abstract
Although placental membrane transporters have an important impact on materno-fetal nutrient transfer, placental cell models are poorly characterized regarding transporter expression. We assessed the mRNA expression of 26 physiologically important solute carriers and ABC transporters in BeWo (b30 clone) and primary human trophoblast cells (PHT) before and after syncytialization. 77% of the transporters showed similar mRNA expression changes between BeWo and PHT after syncytialization. Selected transporters, however, were either lacking in BeWo or showed different trends after syncytialization. In conclusion, BeWo cells generally represent an apt model for transporter studies, but their suitability should be confirmed for each transporter by comparison with PHT.
Collapse
Affiliation(s)
- Sampada Kallol
- Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Switzerland
| | | | - Corneille Edgar Ontsouka
- Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Switzerland.
| |
Collapse
|
39
|
In Vitro Models for Studying Transport Across Epithelial Tissue Barriers. Ann Biomed Eng 2018; 47:1-21. [DOI: 10.1007/s10439-018-02124-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
|
40
|
Kallol S, Huang X, Müller S, Ontsouka CE, Albrecht C. Novel Insights into Concepts and Directionality of Maternal⁻Fetal Cholesterol Transfer across the Human Placenta. Int J Mol Sci 2018; 19:ijms19082334. [PMID: 30096856 PMCID: PMC6121295 DOI: 10.3390/ijms19082334] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/14/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022] Open
Abstract
Cholesterol is indispensable for cellular membrane composition and function. It is also a precursor for the synthesis of steroid hormones, which promote, among others, the maturation of fetal organs. A role of the ATP-binding-cassette-transporter-A1 (ABCA1) in the transport of maternal cholesterol to the fetus was suggested by transferring cholesterol to apolipoprotein-A-1 (apo-A1), but the directionality of the apoA-1/ABCA1-dependent cholesterol transport remains unclear. We isolated primary trophoblasts from term placentae to test the hypotheses that (1) apoA-1/ABCA1 dispatches cholesterol mainly towards the fetus to support fetal developmental maturation at term, and (2) differentiated syncytiotrophoblasts (STB) exert higher cholesterol transport activity than undifferentiated cytotrophoblasts (CTB). As experimental models, we used (1) trophoblast monolayers grown on Transwell® system consisting of apical (maternal-like) and basal (fetal-like) compartments, and (2) trophoblasts grown on conventional culture plates at CTB and STB stages. Surprisingly, apoA-1-mediated cholesterol efflux operated almost exclusively at the apical-maternal side, where ABCA1 was also localized by immunofluorescence. We found greater cholesterol efflux capacity in STB, which was increased by liver-X-receptor agonist treatment and decreased by ABCA1 inhibition. We conclude that at term the apoA-1/ABCA1 pathway is rather involved in cholesterol transport to the mother than in transfer to the fully developed fetus.
Collapse
Affiliation(s)
- Sampada Kallol
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, CH-3012 Bern, Switzerland.
| | - Xiao Huang
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, CH-3012 Bern, Switzerland.
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland.
| | - Stefan Müller
- Department of BioMedical Research, University of Bern, CH-3012 Bern, Switzerland.
| | - Corneille Edgar Ontsouka
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, CH-3012 Bern, Switzerland.
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, CH-3012 Bern, Switzerland.
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland.
| |
Collapse
|
41
|
Biomimetic Placenta-Fetus Model Demonstrating Maternal-Fetal Transmission and Fetal Neural Toxicity of Zika Virus. Ann Biomed Eng 2018; 46:1963-1974. [PMID: 30003503 DOI: 10.1007/s10439-018-2090-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022]
Abstract
Recent global epidemics of viral infection such as Zika virus (ZIKV) and associated birth defects from maternal-fetal viral transmission highlights the critical unmet need for experimental models that adequately recapitulates the biology of the human maternal-fetal interface and downstream fetal development. Herein, we report an in vitro biomimetic placenta-fetus model of the maternal-fetal interface and downstream fetal cells. Using a tissue engineering approach, we built a 3D model incorporating placental trophoblast and endothelial cells into an extracellular matrix environment and validated formation of the maternal-fetal interface. We utilized this model to study ZIKV exposure to the placenta and neural progenitor cells. Our results indicated ZIKV infects both trophoblast and endothelial cells, leading to a higher viral load exposed to fetal cells downstream of the barrier. Viral inhibition by chloroquine reduced the amount of virus both in the placenta and transmitted to fetal cells. A sustained downstream neural cell viability in contrast to significantly reduced viability in an acellular model indicates that the placenta sequesters ZIKV consistent with clinical observations. These findings suggest that the placenta can modulate ZIKV exposure-induced fetal damage. Moreover, such tissue models can enable rigorous assessment of potential therapeutics for maternal-fetal medicine.
Collapse
|
42
|
Aengenheister L, Keevend K, Muoth C, Schönenberger R, Diener L, Wick P, Buerki-Thurnherr T. An advanced human in vitro co-culture model for translocation studies across the placental barrier. Sci Rep 2018; 8:5388. [PMID: 29599470 PMCID: PMC5876397 DOI: 10.1038/s41598-018-23410-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/12/2018] [Indexed: 01/10/2023] Open
Abstract
Although various drugs, environmental pollutants and nanoparticles (NP) can cross the human placental barrier and may harm the developing fetus, knowledge on predictive placental transfer rates and the underlying transport pathways is mostly lacking. Current available in vitro placental transfer models are often inappropriate for translocation studies of macromolecules or NPs and do not consider barrier function of placental endothelial cells (EC). Therefore, we developed a human placental in vitro co-culture transfer model with tight layers of trophoblasts (BeWo b30) and placental microvascular ECs (HPEC-A2) on a low-absorbing, 3 µm porous membrane. Translocation studies with four model substances and two polystyrene (PS) NPs across the individual and co-culture layers revealed that for most of these compounds, the trophoblast and the EC layer both demonstrate similar, but not additive, retention capacity. Only the paracellular marker Na-F was substantially more retained by the BeWo layer. Furthermore, simple shaking, which is often applied to mimic placental perfusion, did not alter translocation kinetics compared to static exposure. In conclusion, we developed a novel placental co-culture model, which provides predictive values for translocation of a broad variety of molecules and NPs and enables valuable mechanistic investigations on cell type-specific placental barrier function.
Collapse
Affiliation(s)
- Leonie Aengenheister
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Kerda Keevend
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Carina Muoth
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - René Schönenberger
- UTOX, EAWAG, Swiss Federal Institute of Aquatic Science and Technology, Ueberlandstrasse 133, 8600, Dübendorf, Switzerland
| | - Liliane Diener
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Peter Wick
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Tina Buerki-Thurnherr
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland.
| |
Collapse
|
43
|
Decreased expression of fibroblast growth factor 13 in early-onset preeclampsia is associated with the increased trophoblast permeability. Placenta 2018; 62:43-49. [DOI: 10.1016/j.placenta.2017.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
|
44
|
Methylmercury Uptake into BeWo Cells Depends on LAT2-4F2hc, a System L Amino Acid Transporter. Int J Mol Sci 2017; 18:ijms18081730. [PMID: 28786956 PMCID: PMC5578120 DOI: 10.3390/ijms18081730] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 12/24/2022] Open
Abstract
The organic mercury compound methylmercury (MeHg) is able to target the fetal brain. However, the uptake of the toxicant into placental cells is incompletely understood. MeHg strongly binds to thiol-S containing molecules such as cysteine. This MeHg-l-cysteine exhibits some structural similarity to methionine. System L plays a crucial role in placental transport of essential amino acids such as leucine and methionine and thus has been assumed to also transport MeHg-l-cysteine across the placenta. The uptake of methylmercury and tritiated leucine and methionine into the choriocarcinoma cell line BeWo was examined using transwell assay and small interfering (si)RNA mediated gene knockdown. Upon the downregulation of large neutral amino acids transporter (LAT)2 and 4F2 cell-surface antigen heavy chain (4F2hc), respectively, the levels of [3H]leucine in BeWo cells are significantly reduced compared to controls treated with non-targeting siRNA (p < 0.05). The uptake of [3H]methionine was reduced upon LAT2 down-regulation as well as methylmercury uptake after 4F2hc silencing (p < 0.05, respectively). These findings suggest an important role of system L in the placental uptake of the metal. Comparing the cellular accumulation of mercury, leucine, and methionine, it can be assumed that (1) MeHg is transported through system L amino acid transporters and (2) system L is responsible for the uptake of amino acids and MeHg primarily at the apical membrane of the trophoblast. The findings together can explain why mercury in contrast to other heavy metals such as lead or cadmium is efficiently transported to fetal blood.
Collapse
|
45
|
Rothbauer M, Patel N, Gondola H, Siwetz M, Huppertz B, Ertl P. A comparative study of five physiological key parameters between four different human trophoblast-derived cell lines. Sci Rep 2017; 7:5892. [PMID: 28724925 PMCID: PMC5517571 DOI: 10.1038/s41598-017-06364-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/06/2017] [Indexed: 12/16/2022] Open
Abstract
The human placenta plays a crucial role as the interface between mother and fetus. It represents a unique tissue that undergoes morphological as well as functional changes on the cellular and tissue level throughout pregnancy. To better understand how the placenta works, a variety of techniques has been developed to re-create this complex physiological barrier in vitro. However, due to the low availability of freshly isolated primary cells, choriocarcinoma cell lines remain the usual suspects as in vitro models for placental research. Here, we present a comparative study on the functional aspects of the choriocarcinoma cell lines BeWo, JAR and Jeg-3, as well as the first trimester trophoblast cell line ACH-3P as placental in vitro barrier models for endocrine and transport studies. Functional assays including tight junction immunostaining, sodium fluorescein retardation, trans epithelial resistance, glucose transport, hormone secretion as well as size-dependent polystyrene nanoparticle transport were performed using the four cell types to evaluate key functional parameters of each cell line to act a relevant in vitro placental barrier model.
Collapse
Affiliation(s)
- Mario Rothbauer
- Vienna University of Technology, Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry & Institute of Chemical Technologies and Analytics, Getreidemarkt 9, 1060, Vienna, Austria.
| | - Nilaykumar Patel
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090, Vienna, Austria
| | - Hajnalka Gondola
- Vienna University of Technology, Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry & Institute of Chemical Technologies and Analytics, Getreidemarkt 9, 1060, Vienna, Austria
| | - Monika Siwetz
- Medical University of Graz, Institute of Cell Biology, Histology and Embryology, Harrachgasse 21/VII, 8010, Graz, Austria
| | - Berthold Huppertz
- Medical University of Graz, Institute of Cell Biology, Histology and Embryology, Harrachgasse 21/VII, 8010, Graz, Austria
| | - Peter Ertl
- Vienna University of Technology, Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry & Institute of Chemical Technologies and Analytics, Getreidemarkt 9, 1060, Vienna, Austria
| |
Collapse
|
46
|
|
47
|
Saadeldin IM, Swelum AAA, Elsafadi M, Moumen AF, Alzahrani FA, Mahmood A, Alfayez M, Alowaimer AN. Isolation and characterization of the trophectoderm from the Arabian camel (Camelus dromedarius). Placenta 2017; 57:113-122. [PMID: 28863999 DOI: 10.1016/j.placenta.2017.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
Abstract
We isolated and characterized trophoblast from in vivo-derived camel embryos and compared with embryonic stem-like cells. Camel embryos were flushed on day 8 post-insemination and used to derive trophectoderm and embryonic stem-like cells under feeder-free culture conditions using a basement membrane matrix. Embryos were evaluated for the expression of POU5F1, MYC, KLF4, SOX2, CDX2, and KRT8 mRNA transcripts by relative quantitative polymerase chain reaction. Camel embryos grew and expanded to ∼4.5 mm and maintained their vesicular shape in vitro for 21 days post-insemination. Trophoblast and embryonic stem-like cell lines grew under feeder-free culture conditions and showed distinct morphological criteria and normal chromosomal counts. Embryonic stem-like cells showed positive staining in the alkaline phosphatase reaction. Trophoblast cells showed a significant increase in CDX2, KRT8, KLF4, and SOX2 expression compared with embryonic stem-like cells and whole embryos. Embryonic stem-like cells showed a significant decrease in CDX2 expression and increase in SOX2 and KRT8 expression compared to embryonic expression. POU5F1 and MYC expression showed no difference between embryos and both cell lines. We characterized embryo survival in vitro, particularly the derivation of trophectoderm and embryonic stem-like cells, providing a foundation for further analysis of early embryonic development and placentation in camels.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia; Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, 44519 Zagazig, Egypt.
| | - Ayman Abdel-Aziz Swelum
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia; Department of Theriogeneology, Faculty of Veterinary Medicine, Zagazig University, 44519 Zagazig, Egypt
| | - Mona Elsafadi
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Moumen
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Faisal A Alzahrani
- Department of Biological Sciences, Rabigh College of Science and Arts, King Abdulaziz University, Rabigh Branch, Rabigh 21911, Saudi Arabia
| | - Amer Mahmood
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Musaad Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah N Alowaimer
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia
| |
Collapse
|
48
|
Mikkelsen E, Lauridsen H, Nielsen PM, Qi H, Nørlinger T, Andersen MD, Uldbjerg N, Laustsen C, Sandager P, Pedersen M. The chinchilla as a novel animal model of pregnancy. ROYAL SOCIETY OPEN SCIENCE 2017; 4:161098. [PMID: 28484627 PMCID: PMC5414264 DOI: 10.1098/rsos.161098] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/28/2017] [Indexed: 06/07/2023]
Abstract
Several parameters are important when choosing the most appropriate animal to model human obstetrics, including gestation period, number of fetuses per gestation and placental structure. The domesticated long-tailed chinchilla (Chinchilla lanigera) is a well-suited and appropriate animal model of pregnancy that often will carry only one offspring and has a long gestation period of 105-115 days. Furthermore, the chinchilla placenta is of the haemomonochorial labyrinthine type and is therefore comparable to the human villous haemomonochorial placenta. This proof-of-concept study demonstrated the feasibility in laboratory settings, and demonstrated the potential of the pregnant chinchilla as an animal model for obstetric research and its potential usefulness for non-invasive measurements in the placenta. We demonstrate measurements of the placental and fetal metabolism (demonstrated in vivo by hyperpolarized MRI and in vitro by qPCR analyses), placental vessels (demonstrated ex vivo by contrast-enhanced CT angiography) and overall anatomy (demonstrated in vivo by whole-body CT).
Collapse
Affiliation(s)
- Emmeli Mikkelsen
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Henrik Lauridsen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Per Mose Nielsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Haiyun Qi
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Thomas Nørlinger
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Maria Dahl Andersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Niels Uldbjerg
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Puk Sandager
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Michael Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| |
Collapse
|