1
|
Bondarenko N, Lifantseva N, Voronova S, Melnikova V. The Placenta as the Main Source of Serotonin in Ontogenetic Dynamics: Inflammation-Induced Modulation of Placental Serotonin Can Be Prevented by Immunoglobulin Administration. Int J Mol Sci 2024; 25:13532. [PMID: 39769295 PMCID: PMC11677199 DOI: 10.3390/ijms252413532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Placental serotonin is recognized as a key component of feto-placental physiology and can be influenced by environmental factors such as maternal diet, drugs, stress, and immune activation. In this study, we compared the contribution of placental and fetal sources to the maintenance of serotonin levels required for normal fetal development during ontogenetic dynamics. Our results demonstrated the leading role of the placenta at almost all stages of development. We investigated the modulatory effect of inflammation on placental serotonin levels. The data obtained showed that the susceptibility to prenatal inflammation depends on its severity and varies considerably at different stages of development. According to our results, inflammation-induced modulation of placental serotonin levels can be prevented by immunoglobulin administration at both early and late stages of development. Disturbances in placental serotonin signaling during critical developmental periods may have long-lasting consequences for the health and behavior of the offspring. Therefore, the ability to prevent environmental modulation of placental serotonin, and hence negative effects on the developing fetus, is of great importance.
Collapse
Affiliation(s)
| | | | | | - Victoria Melnikova
- Laboratory of Comparative Developmental Physiology, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow 119334, Russia; (N.B.); (N.L.); (S.V.)
| |
Collapse
|
2
|
Stapleton PA. The Application of Engineered Nanomaterials in Perinatal Therapeutics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2303072. [PMID: 37438678 PMCID: PMC10784409 DOI: 10.1002/smll.202303072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Pregnancy is a vulnerable life stage for the mother and developing fetus. Because of this dual concern, approved therapeutic options for pre-existing conditions or pregnancy-induced pathologies, placental deformities, or fetal concerns are extremely limited. These cases often leave patients and clinicians having to choose between maternal health and fetal development. Recent advancements in nanomedicine and nanotherapeutic devices have made the development of perinatal therapeutics an attractive objective. However, perinatal medicine requires a multifaceted approach given the interactions between maternal, placental, and fetal physiology. Maternal-fetal interactions are centralized to the placenta, a specialized transient barrier organ, to allow for nutrient and waste exchange. Perinatal nanotherapeutics must be designed for placental avoidance or uptake. In this review, pregnancy-related conditions, experimental models, and modes of drug delivery during pregnancy are discussed.
Collapse
Affiliation(s)
- Phoebe A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd., Piscataway, NJ, 08854, USA
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ, 08854, USA
| |
Collapse
|
3
|
Spiess D, Abegg VF, Chauveau A, Treyer A, Reinehr M, Oufir M, Duong E, Potterat O, Hamburger M, Simões-Wüst AP. Placental Passage of Protopine in an Ex Vivo Human Perfusion System. PLANTA MEDICA 2023; 89:194-207. [PMID: 35445384 PMCID: PMC9868778 DOI: 10.1055/a-1829-9546] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/08/2022] [Indexed: 06/14/2023]
Abstract
The placental passage of protopine was investigated with a human ex vivo placental perfusion model. The model was first validated with diazepam and citalopram, 2 compounds known to cross the placental barrier, and antipyrine as a positive control. All compounds were quantified by partially validated U(H)PLC-MS/MS bioanalytical methods. Protopine was transferred from the maternal to the fetal circuit, with a steady-state reached after 90 min. The study compound did not affect placental viability or functionality, as glucose consumption, lactate production, and beta-human chorionic gonadotropin, and leptin release remained constant. Histopathological evaluation of all placental specimens showed unremarkable, age-appropriate parenchymal maturation with no pathologic findings.
Collapse
Affiliation(s)
- Deborah Spiess
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Vanessa Fabienne Abegg
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Antoine Chauveau
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Andrea Treyer
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Michael Reinehr
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Mouhssin Oufir
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Elisa Duong
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Olivier Potterat
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Matthias Hamburger
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Ana Paula Simões-Wüst
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Willi CE, Abdelazim H, Chappell JC. Evaluating cell viability, capillary perfusion, and collateral tortuosity in an ex vivo mouse intestine fluidics model. Front Bioeng Biotechnol 2022; 10:1008481. [PMID: 36568288 PMCID: PMC9780384 DOI: 10.3389/fbioe.2022.1008481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Numerous disease conditions involve the sudden or progressive loss of blood flow. Perfusion restoration is vital for returning affected organs to full health. While a range of clinical interventions can successfully restore flow to downstream tissues, the microvascular responses after a loss-of-flow event can vary over time and may involve substantial microvessel instability. Increased insight into perfusion-mediated capillary stability and access-to-flow is therefore essential for advancing therapeutic reperfusion strategies and improving patient outcomes. To that end, we developed a tissue-based microvascular fluidics model to better understand (i) microvascular stability and access-to-flow over an acute time course post-ischemia, and (ii) collateral flow in vessels neighboring an occlusion site. We utilized murine intestinal tissue regions by catheterizing a feeder artery and introducing perfusate at physiologically comparable flow-rates. The cannulated vessel as well as a portion of the downstream vessels and associated intestinal tissue were cultured while constant perfusion conditions were maintained. An occlusion was introduced in a selected arterial segment, and changes in perfusion within areas receiving varying degrees of collateral flow were observed over time. To observe the microvascular response to perfusion changes, we incorporated (i) tissues harboring cell-reporter constructs, specifically Ng2-DsRed labeling of intestinal pericytes, and (ii) different types of fluorescent perfusates to quantify capillary access-to-flow at discrete time points. In our model, we found that perfusion tracers could enter capillaries within regions downstream of an occlusion upon the initial introduction of perfusion, but at 24 h tissue perfusion was severely decreased. However, live/dead cell discrimination revealed that the tissue overall did not experience significant cell death, including that of microvascular pericytes, even after 48 h. Our findings suggest that altered flow conditions may rapidly initiate cellular responses that reduce capillary access-to-flow, even in the absence of cellular deterioration or hypoxia. Overall, this ex vivo tissue-based microfluidics model may serve as a platform upon which a variety of follow-on studies may be conducted. It will thus enhance our understanding of microvessel stability and access-to-flow during an occlusive event and the role of collateral flow during normal and disrupted perfusion.
Collapse
Affiliation(s)
- Caroline E. Willi
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA, United States,FBRI Center for Vascular and Heart Research, Roanoke, VA, United States
| | - Hanaa Abdelazim
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA, United States,FBRI Center for Vascular and Heart Research, Roanoke, VA, United States
| | - John C. Chappell
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA, United States,FBRI Center for Vascular and Heart Research, Roanoke, VA, United States,Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States,Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States,*Correspondence: John C. Chappell,
| |
Collapse
|
5
|
de Bartolomeis A, Barone A, Vellucci L, Mazza B, Austin MC, Iasevoli F, Ciccarelli M. Linking Inflammation, Aberrant Glutamate-Dopamine Interaction, and Post-synaptic Changes: Translational Relevance for Schizophrenia and Antipsychotic Treatment: a Systematic Review. Mol Neurobiol 2022; 59:6460-6501. [PMID: 35963926 PMCID: PMC9463235 DOI: 10.1007/s12035-022-02976-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/24/2022] [Indexed: 12/16/2022]
Abstract
Evidence from clinical, preclinical, and post-mortem studies supports the inflammatory/immune hypothesis of schizophrenia pathogenesis. Less evident is the link between the inflammatory background and two well-recognized functional and structural findings of schizophrenia pathophysiology: the dopamine-glutamate aberrant interaction and the alteration of dendritic spines architecture, both believed to be the “quantal” elements of cortical-subcortical dysfunctional network. In this systematic review, we tried to capture the major findings linking inflammation, aberrant glutamate-dopamine interaction, and post-synaptic changes under a direct and inverse translational perspective, a paramount picture that at present is lacking. The inflammatory effects on dopaminergic function appear to be bidirectional: the inflammation influences dopamine release, and dopamine acts as a regulator of discrete inflammatory processes involved in schizophrenia such as dysregulated interleukin and kynurenine pathways. Furthermore, the link between inflammation and glutamate is strongly supported by clinical studies aimed at exploring overactive microglia in schizophrenia patients and maternal immune activation models, indicating impaired glutamate regulation and reduced N-methyl-D-aspartate receptor (NMDAR) function. In addition, an inflammatory/immune-induced alteration of post-synaptic density scaffold proteins, crucial for downstream NMDAR signaling and synaptic efficacy, has been demonstrated. According to these findings, a significant increase in plasma inflammatory markers has been found in schizophrenia patients compared to healthy controls, associated with reduced cortical integrity and functional connectivity, relevant to the cognitive deficit of schizophrenia. Finally, the link between altered inflammatory/immune responses raises relevant questions regarding potential new therapeutic strategies specifically for those forms of schizophrenia that are resistant to canonical antipsychotics or unresponsive to clozapine.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy. .,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy.
| | - Annarita Barone
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Licia Vellucci
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Benedetta Mazza
- Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Mark C Austin
- Clinical Psychopharmacology Program, College of Pharmacy, Idaho State University (ISU), Pocatello, ID, USA
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Mariateresa Ciccarelli
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| |
Collapse
|
6
|
Kopacz A, Kloska D, Klimczyk D, Kopec M, Jozkowicz A, Piechota-Polanczyk A. Nrf2 Transcriptional Activity Governs Intestine Development. Int J Mol Sci 2022; 23:ijms23116175. [PMID: 35682851 PMCID: PMC9181470 DOI: 10.3390/ijms23116175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 02/06/2023] Open
Abstract
Our recent findings indicate that Nrf2 transcriptional activity is essential in maintaining the proper large intestinal structure in adult mice. Here, we aimed to verify whether Nrf2-related intestine abnormalities stemmed from the early weaning or gestational periods. Therefore, we analyzed 4-day-old pups and embryos devoid of Nrf2 transcriptional activity (tKO) and their wild-type counterparts. We found significant changes in the intestinal structure of 4-day-old Nrf2 tKO pups including a longer colon, altered crypt distribution, and enlargement of the goblet cells with a markedly higher level of mucin 2. Tracing back the origin of these alterations, we observed that they appeared as early as day 14.5 of embryonic development, independently of sex. Importantly, in this period, we observed a significant increase in the Nrf2 level and a distinctive, untimely pattern of expression of the proliferation factor Ki67. At the latest stage of embryonic development, we detected a premature drop in the differentiation factor Notch1. We suspect that intestine abnormalities in mice lacking Nrf2 transcriptional activity stem from sex-independent disturbed intestinal cell proliferation and could be further exacerbated by altered differentiation. Summing up, we identified Nrf2 transcriptional activity as an important regulator of intestinal formation. It influences the hindgut cell proliferation and differentiation at different stages of embryonic development.
Collapse
|
7
|
Garner KL, Bowdridge EC, DeVallance E, Griffith JA, Kelley EE, Nurkiewicz TR. Using the Isolated Rat Placenta to Assess Fetoplacental Hemodynamics. FRONTIERS IN TOXICOLOGY 2022; 4:814071. [PMID: 35295220 PMCID: PMC8915812 DOI: 10.3389/ftox.2022.814071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/24/2022] [Indexed: 02/05/2023] Open
Abstract
Placental health is critical to fetal growth and maternal health during gestation. However, investigating placental flow in an ex-vivo isolated system where inflow is independently controlled has yet to be developed in the rat. Here, we describe a novel technique, isolated perfused placenta technique that allows for analysis of placental pressure outflow pressure, placental flow in rats at gestational day 20. Using this method, we successfully perfused placentas from dams and were able to observe increases in outflow pressure and flow as the inflow pressure to the placenta was increased in a step wise fashion. This method will help to advance the functional analysis of placental flow and therefore placental resistance and efficiency.
Collapse
Affiliation(s)
- K. L. Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States,Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, United States
| | - E. C. Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States,Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, United States
| | - E. DeVallance
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States,Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, United States
| | - J. A. Griffith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States,Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, United States
| | - E. E. Kelley
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States,Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, United States
| | - T. R. Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States,Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, United States,*Correspondence: T. R. Nurkiewicz,
| |
Collapse
|
8
|
Spiess D, Abegg VF, Chauveau A, Treyer A, Reinehr M, Oufir M, Duong E, Potterat O, Hamburger M, Simões-Wüst AP. Placental Passage of Humulone and Protopine in an Ex Vivo Human Perfusion System. PLANTA MEDICA 2021; 87:1192-1205. [PMID: 34530480 PMCID: PMC8585570 DOI: 10.1055/a-1578-3803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/28/2021] [Indexed: 05/07/2023]
Abstract
The placental passage of humulone and protopine was investigated with a human ex vivo placental perfusion model. The model was first validated with diazepam and citalopram, 2 compounds known to cross the placental barrier, and antipyrine as a positive control. All compounds were quantified by partially validated U(H)PLC-MS/MS bioanalytical methods. Only a small portion of humulone initially present in the maternal circuit reached the fetal circuit. The humulone concentration in the maternal circuit rapidly decreased, likely due to metabolization in the placenta. Protopine was transferred from the maternal to the fetal circuit, with a steady-state reached after 90 min. None of the study compounds affected placental viability or functionality, as glucose consumption, lactate production, beta-human chorionic gonadotropin, and leptin release remained constant. Histopathological evaluation of all placental specimens showed unremarkable, age-appropriate parenchymal maturation with no pathologic findings.
Collapse
Affiliation(s)
- Deborah Spiess
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Vanessa Fabienne Abegg
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Antoine Chauveau
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Andrea Treyer
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Michael Reinehr
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Mouhssin Oufir
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Elisa Duong
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Olivier Potterat
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Matthias Hamburger
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Ana Paula Simões-Wüst
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Fournier SB, D'Errico JN, Stapleton PA. Uterine Vascular Control Preconception and During Pregnancy. Compr Physiol 2021; 11:1871-1893. [PMID: 34061977 DOI: 10.1002/cphy.c190015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Successful pregnancy and reproduction are dependent on adequate uterine blood flow, placental perfusion, and vascular responsivity to fetal demands. The ability to support pregnancy centers on systemic adaptation and endometrial preparation through decidualization, embryonic implantation, trophoblast invasion, arterial/arteriolar reactivity, and vascular remodeling. These adaptations occur through responsiveness to endocrine signaling and local uteroplacental mediators. The purpose of this article is to highlight the current knowledge associated with vascular remodeling and responsivity during uterine preparation for and during pregnancy. We focus on maternal cardiovascular systemic and uterine modifications, endometrial decidualization, implantation and invasion, uterine and spiral artery remodeling, local uterine regulatory mechanisms, placentation, and pathological consequences of vascular dysfunction during pregnancy. © 2021 American Physiological Society. Compr Physiol 11:1-23, 2021.
Collapse
Affiliation(s)
- Sara B Fournier
- Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey, USA
| | - Jeanine N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Phoebe A Stapleton
- Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey, USA.,Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
10
|
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: 47] [Impact Index Per Article: 9.4] [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
|
11
|
Turk EA, Stout JN, Ha C, Luo J, Gagoski B, Yetisir F, Golland P, Wald LL, Adalsteinsson E, Robinson JN, Roberts DJ, Barth WH, Grant PE. Placental MRI: Developing Accurate Quantitative Measures of Oxygenation. Top Magn Reson Imaging 2019; 28:285-297. [PMID: 31592995 PMCID: PMC7323862 DOI: 10.1097/rmr.0000000000000221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The Human Placenta Project has focused attention on the need for noninvasive magnetic resonance imaging (MRI)-based techniques to diagnose and monitor placental function throughout pregnancy. The hope is that the management of placenta-related pathologies would be improved if physicians had more direct, real-time measures of placental health to guide clinical decision making. As oxygen alters signal intensity on MRI and oxygen transport is a key function of the placenta, many of the MRI methods under development are focused on quantifying oxygen transport or oxygen content of the placenta. For example, measurements from blood oxygen level-dependent imaging of the placenta during maternal hyperoxia correspond to outcomes in twin pregnancies, suggesting that some aspects of placental oxygen transport can be monitored by MRI. Additional methods are being developed to accurately quantify baseline placental oxygenation by MRI relaxometry. However, direct validation of placental MRI methods is challenging and therefore animal studies and ex vivo studies of human placentas are needed. Here we provide an overview of the current state of the art of oxygen transport and quantification with MRI. We suggest that as these techniques are being developed, increased focus be placed on ensuring they are robust and reliable across individuals and standardized to enable predictive diagnostic models to be generated from the data. The field is still several years away from establishing the clinical benefit of monitoring placental function in real time with MRI, but the promise of individual personalized diagnosis and monitoring of placental disease in real time continues to motivate this effort.
Collapse
Affiliation(s)
- Esra Abaci Turk
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children’s Hospital, MA, USA
| | - Jeffrey N. Stout
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children’s Hospital, MA, USA
| | - Christopher Ha
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children’s Hospital, MA, USA
| | - Jie Luo
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Borjan Gagoski
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children’s Hospital, MA, USA
| | - Filiz Yetisir
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children’s Hospital, MA, USA
| | - Polina Golland
- Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Lawrence L. Wald
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Elfar Adalsteinsson
- Department of Electrical Engineering and Computer Science Massachusetts Institute of Technology, Cambridge, MA, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Julian N. Robinson
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, USA
| | | | - William H. Barth
- Maternal-Fetal Medicine, Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - P. Ellen Grant
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children’s Hospital, MA, USA
| |
Collapse
|
12
|
Velasquez JC, Zhao Q, Chan Y, Galindo LC, Simasotchi C, Wu D, Hou Z, Herod SM, Oberlander TF, Gil S, Fournier T, Burd I, Andrews AM, Bonnin A. In Utero Exposure to Citalopram Mitigates Maternal Stress Effects on Fetal Brain Development. ACS Chem Neurosci 2019; 10:3307-3317. [PMID: 31184110 DOI: 10.1021/acschemneuro.9b00180] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Human epidemiological and animal-model studies suggest that separate exposure to stress or serotonin-selective reuptake inhibitor (SSRI) antidepressants during pregnancy increases risks for neurodevelopmental disorders in offspring. Yet, little is known about the combined effects of maternal stress and SSRIs with regard to brain development in utero. We found that the placenta is highly permeable to the commonly prescribed SSRI (±)-citalopram (CIT) in humans and mice, allowing rapid exposure of the fetal brain to this drug. We investigated the effects of maternal chronic unpredictable stress in mice with or without maternal oral administration of CIT from embryonic day (E)8 to E17. We assessed fetal brain development using magnetic resonance imaging and quantified changes in serotonergic, thalamocortical, and cortical development. In utero exposure to maternal stress did not affect overall fetal brain growth. However, serotonin tissue content in the fetal forebrain was increased in association with maternal stress; this increase was reversed by maternal CIT. In utero exposure to stress increased the numbers of deep-layer neurons in specific cortical regions, whereas CIT increased overall cell numbers without changing the proportions of layer-specific neurons to offset the effects of stress on deep-layer cortical development. These findings suggest that stress and SSRI exposure in utero differentially impact serotonin-dependent fetal neurodevelopment such that CIT reverses key effects of maternal gestational stress on offspring brain development.
Collapse
Affiliation(s)
- Juan C. Velasquez
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Qiuying Zhao
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Yen Chan
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Ligia C.M. Galindo
- Department of Anatomy, Federal University of Pernambuco, Recife 50670, Brazil
| | | | - Dan Wu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Zhipeng Hou
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Skyla M. Herod
- Department of Biology and Chemistry, Azusa Pacific University, Azusa, California 91702, United States
| | - Tim F. Oberlander
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3 V4, Canada
| | - Sophie Gil
- UMR-S 1139 INSERM/University of Paris Descartes, 75006 Paris, France
- PremUp Foundation, 75014 Paris, France
| | - Thierry Fournier
- UMR-S 1139 INSERM/University of Paris Descartes, 75006 Paris, France
- PremUp Foundation, 75014 Paris, France
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Anne M. Andrews
- Terry and Jane Semel Institute for Neuroscience & Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, and Departments of Psychiatry and Biobehavioral Sciences and Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Alexandre Bonnin
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
13
|
D'Errico JN, Doherty C, Fournier SB, Renkel N, Kallontzi S, Goedken M, Fabris L, Buckley B, Stapleton PA. Identification and quantification of gold engineered nanomaterials and impaired fluid transfer across the rat placenta via ex vivo perfusion. Biomed Pharmacother 2019; 117:109148. [PMID: 31347503 DOI: 10.1016/j.biopha.2019.109148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
Development and implementation of products incorporating nanoparticles are occurring at a rapid pace. These particles are widely utilized in domestic, occupational, and biomedical applications. Currently, it is unclear if pregnant women will be able to take advantage of the potential biomedical nanoproducts out of concerns associated with placental transfer and fetal interactions. We recently developed an ex vivo rat placental perfusion technique to allow for the evaluation of xenobiotic transfer and placental physiological perturbations. In this study, a segment of the uterine horn and associated placenta was isolated from pregnant (gestational day 20) Sprague-Dawley rats and placed into a modified pressure myography vessel chamber. The proximal and distal ends of the maternal uterine artery and the vessels of the umbilical cord were cannulated, secured, and perfused with physiological salt solution (PSS). The proximal uterine artery and umbilical artery were pressurized at 80 mmHg and 50 mmHg, respectively, to allow countercurrent flow through the placenta. After equilibration, a single 900 μL bolus dose of 20 nm gold engineered nanoparticles (Au-ENM) was introduced into the proximal maternal artery. Distal uterine and umbilical vein effluents were collected every 10 min for 180 min to measure placental fluid dynamics. The quantification of Au-ENM transfer was conducted via inductively coupled plasma mass spectrometry (ICP-MS). Overall, we were able to measure Au-ENM within uterine and umbilical effluent with 20 min of material infusion. This novel methodology may be widely incorporated into studies of pharmacology, toxicology, and placental physiology.
Collapse
Affiliation(s)
- J N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - C Doherty
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - S B Fournier
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - N Renkel
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - S Kallontzi
- Department of Material Science and Engineering, School of Engineering, Rutgers University, 607 Taylor Rd., Piscataway, NJ 08854, USA
| | - M Goedken
- Research Pathology Services, Rutgers University, Piscataway, NJ 08854, USA
| | - L Fabris
- Department of Material Science and Engineering, School of Engineering, Rutgers University, 607 Taylor Rd., Piscataway, NJ 08854, USA
| | - B Buckley
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - P A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd., Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA.
| |
Collapse
|
14
|
Abstract
The placenta is a key organ during pregnancy that serves as a barrier to fetal xenobiotic exposure and mediates the exchange of nutrients for waste. An assay is described here to perfuse an isolated rat placenta and evaluate the maternal-to-fetal translocation of xenobiotics ex vivo. In addition, the evaluation of physiological processes such as fluid flow to the fetus and placental metabolism may be conducted with this methodology. This technique is suitable for evaluating maternal-to-fetal kinetics of pharmaceutical candidates or environmental contaminants. In contrast to current alternative approaches, this methodology allows the evaluation of the isolated maternal-fetal vasculature, with the systemic neural or immune involvement removed, allowing any observed changes in physiological function to be attributable to local factors within the isolated tissue.
Collapse
Affiliation(s)
- Jeanine N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University
| | | | - Phoebe A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University; Environmental and Occupational Health Sciences Institute;
| |
Collapse
|
15
|
Abukabda AB, Bowdridge EC, McBride CR, Batchelor TP, Goldsmith WT, Garner KL, Friend S, Nurkiewicz TR. Maternal titanium dioxide nanomaterial inhalation exposure compromises placental hemodynamics. Toxicol Appl Pharmacol 2019; 367:51-61. [PMID: 30711534 DOI: 10.1016/j.taap.2019.01.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 01/12/2023]
Abstract
The fetal consequences of gestational engineered nanomaterial (ENM) exposure are unclear. The placenta is a barrier protecting the fetus and allowing transfer of substances from the maternal circulation. The purpose of this study was to determine the effects of maternal pulmonary titanium dioxide nanoparticle (nano-TiO2) exposure on the placenta and umbilical vascular reactivity. We hypothesized that pulmonary nano-TiO2 inhalation exposure increases placental vascular resistance and impairs umbilical vascular responsiveness. Pregnant Sprague-Dawley rats were exposed via whole-body inhalation to nano-TiO2 with an aerodynamic diameter of 188 ± 0.36 nm. On gestational day (GD) 11, rats began inhalation exposures (6 h/exposure). Daily lung deposition was 87.5 ± 2.7 μg. Animals were exposed for 6 days for a cumulative lung burden of 525 ± 16 μg. On GD 20, placentas, umbilical artery and vein were isolated, cannulated, and treated with acetylcholine (ACh), angiotensin II (ANGII), S-nitroso-N-acetyl-DL-penicillamine (SNAP), or calcium-free superfusate (Ca2+-free). Mean outflow pressure was measured in placental units. ACh increased outflow pressure to 53 ± 5 mmHg in sham-controls but only to 35 ± 4 mmHg in exposed subjects. ANGII decreased outflow pressure in placentas from exposed animals (17 ± 7 mmHg) compared to sham-controls (31 ± 6 mmHg). Ca2+-free superfusate yielded maximal outflow pressures in sham-control (63 ± 5 mmHg) and exposed (30 ± 10 mmHg) rats. Umbilical artery endothelium-dependent dilation was decreased in nano-TiO2 exposed fetuses (30 ± 9%) compared to sham-controls (58 ± 6%), but ANGII sensitivity was increased (-79 ± 20% vs -36 ± 10%). These results indicate that maternal gestational pulmonary nano-TiO2 exposure increases placental vascular resistance and impairs umbilical vascular reactivity.
Collapse
Affiliation(s)
- Alaeddin B Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Carroll R McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas P Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William T Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista L Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA; Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA; National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
16
|
Fitzgerald W, Gomez-Lopez N, Erez O, Romero R, Margolis L. Extracellular vesicles generated by placental tissues ex vivo: A transport system for immune mediators and growth factors. Am J Reprod Immunol 2018; 80:e12860. [PMID: 29726582 PMCID: PMC6021205 DOI: 10.1111/aji.12860] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/12/2022] Open
Abstract
PROBLEM To study the mechanisms of placenta function and the role of extracellular vesicles (EVs) in pregnancy, it is necessary to develop an ex vivo system that retains placental cytoarchitecture and the primary metabolic aspects, in particular the release of EVs and soluble factors. Here, we developed such a system and investigated the pattern of secretion of cytokines, growth factors, and extracellular vesicles by placental villous and amnion tissues ex vivo. METHODS OF STUDY Placental villous and amnion explants were cultured for 2 weeks at the air/liquid interface and their morphology and the released cytokines and EVs were analyzed. Cytokines were analyzed with multiplexed bead assays, and individual EVs were analyzed with recently developed techniques that involved EV capture with magnetic nanoparticles coupled to anti-EV antibodies and flow cytometry. RESULTS Ex vivo tissues (i) remained viable and preserved their cytoarchitecture; (ii) maintained secretion of cytokines and growth factors; (iii) released EVs of syncytiotrophoblast and amnion epithelial cell origins that contain cytokines and growth factors. CONCLUSION A system of ex vivo placental villous and amnion tissues can be used as an adequate model to study placenta metabolic activity in normal and complicated pregnancies, in particular to characterize EVs by their surface markers and by encapsulated proteins. Establishment and benchmarking the placenta ex vivo system may provide new insight in the functional status of this organ in various placental disorders, particularly regarding the release of EVs and cytokines. Such EVs may have a prognostic value for pregnancy complications.
Collapse
Affiliation(s)
- Wendy Fitzgerald
- Section of Intercellular Interactions, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Leonid Margolis
- Section of Intercellular Interactions, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| |
Collapse
|
17
|
Fournier SB, D'Errico JN, Stapleton PA. Engineered nanomaterial applications in perinatal therapeutics. Pharmacol Res 2018; 130:36-43. [PMID: 29477479 PMCID: PMC5965276 DOI: 10.1016/j.phrs.2018.02.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 02/07/2023]
Abstract
Engineered nanomaterials (ENM) are widely used in commercial, domestic, and more recently biomedical applications. While the majority of exposures to ENM are unintentional, biomedical platforms are being evaluated for use in individualized and/or tissue-targeted therapies. Treatments are often avoided during prenatal periods to reduce adverse effects on the developing fetus. The placenta is central to maternal-fetal medicine. Perturbation of placental functions can limit transfer of necessary nutrients, alter production of hormones needed during pregnancy, or allow undesired passage of xenobiotics to the developing fetus. The development of therapeutics to target specific maternal, placental, or fetal tissues would be especially important to reduce or circumvent toxicities. Therefore, this review will discuss the potential use of ENM in perinatal medicine, the applicable physiochemical properties of ENM in therapeutic use, and current methodologies of ENM testing in perinatal medicine, and identify maternal, fetal, and offspring concerns associated with ENM exposure during gestation. As potential nanoparticle-based therapies continue to develop, so does the need for thorough consideration and evaluation for use in perinatal medicine.
Collapse
Affiliation(s)
- S B Fournier
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - J N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - P A Stapleton
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA; Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd., Piscataway, NJ 08854, USA.
| |
Collapse
|
18
|
Goeden N, Notarangelo FM, Pocivavsek A, Beggiato S, Bonnin A, Schwarcz R. Prenatal Dynamics of Kynurenine Pathway Metabolism in Mice: Focus on Kynurenic Acid. Dev Neurosci 2017; 39:519-528. [PMID: 29080891 DOI: 10.1159/000481168] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/31/2017] [Indexed: 01/09/2023] Open
Abstract
The kynurenine pathway (KP), the major catabolic route of tryptophan in mammals, contains several neuroactive metabolites, including kynurenic acid (KYNA) and 3-hydroxykynurenine (3-HK). KP metabolism, and especially the fate of KYNA, during pregnancy is poorly understood, yet it may play a significant role in the development of psychiatric disorders later in life. The present study was designed to investigate the prenatal features of KP metabolism in vivo, with special focus on KYNA. To this end, pregnant CD-1 mice were treated systemically with kynurenine (100 mg/kg), KYNA (10 mg/kg), or saline on embryonic day 18. As expected, administration of either kynurenine or KYNA increased KYNA levels in the maternal plasma and placenta. Maternal kynurenine treatment also raised kynurenine levels in the fetal plasma and brain, demonstrating the ability of this pivotal KP metabolite to cross the placenta and increase the levels of both KYNA and 3-HK in the fetal brain. In contrast, maternal administration of KYNA caused only a small, nonsignificant elevation in KYNA levels in fetal plasma and brain. Complementary experiments using an ex vivo placental perfusion procedure confirmed the significant transplacental transfer of kynurenine and demonstrated that only a very small fraction of maternal kynurenine is converted to KYNA in the placenta and released into the fetal compartment under physiological conditions. Jointly, these results help to clarify the contributions of the maternal circulation and the placenta to fetal KYNA in the late prenatal period.
Collapse
Affiliation(s)
- Nick Goeden
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
19
|
Maternal Inflammation Disrupts Fetal Neurodevelopment via Increased Placental Output of Serotonin to the Fetal Brain. J Neurosci 2017; 36:6041-9. [PMID: 27251625 DOI: 10.1523/jneurosci.2534-15.2016] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 04/13/2016] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Maternal inflammation during pregnancy affects placental function and is associated with increased risk of neurodevelopmental disorders in the offspring. The molecular mechanisms linking placental dysfunction to abnormal fetal neurodevelopment remain unclear. During typical development, serotonin (5-HT) synthesized in the placenta from maternal l-tryptophan (TRP) reaches the fetal brain. There, 5-HT modulates critical neurodevelopmental processes. We investigated the effects of maternal inflammation triggered in midpregnancy in mice by the immunostimulant polyriboinosinic-polyribocytidylic acid [poly(I:C)] on TRP metabolism in the placenta and its impact on fetal neurodevelopment. We show that a moderate maternal immune challenge upregulates placental TRP conversion rapidly to 5-HT through successively transient increases in substrate availability and TRP hydroxylase (TPH) enzymatic activity, leading to accumulation of exogenous 5-HT and blunting of endogenous 5-HT axonal outgrowth specifically within the fetal forebrain. The pharmacological inhibition of TPH activity blocked these effects. These results establish altered placental TRP conversion to 5-HT as a new mechanism by which maternal inflammation disrupts 5-HT-dependent neurogenic processes during fetal neurodevelopment. SIGNIFICANCE STATEMENT The mechanisms linking maternal inflammation during pregnancy with increased risk of neurodevelopmental disorders in the offspring are poorly understood. In this study, we show that maternal inflammation in midpregnancy results in an upregulation of tryptophan conversion to serotonin (5-HT) within the placenta. Remarkably, this leads to exposure of the fetal forebrain to increased concentrations of this biogenic amine and to specific alterations of crucially important 5-HT-dependent neurogenic processes. More specifically, we found altered serotonergic axon growth resulting from increased 5-HT in the fetal forebrain. The data provide a new understanding of placental function playing a key role in fetal brain development and how this process is altered by adverse prenatal events such as maternal inflammation. The results uncover important future directions for understanding the early developmental origins of mental disorders.
Collapse
|
20
|
Muller CL, Anacker AMJ, Rogers TD, Goeden N, Keller EH, Forsberg CG, Kerr TM, Wender CLA, Anderson GM, Stanwood GD, Blakely RD, Bonnin A, Veenstra-VanderWeele J. Impact of Maternal Serotonin Transporter Genotype on Placental Serotonin, Fetal Forebrain Serotonin, and Neurodevelopment. Neuropsychopharmacology 2017; 42:427-436. [PMID: 27550733 PMCID: PMC5399236 DOI: 10.1038/npp.2016.166] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/16/2016] [Accepted: 08/04/2016] [Indexed: 12/29/2022]
Abstract
Biomarker, neuroimaging, and genetic findings implicate the serotonin transporter (SERT) in autism spectrum disorder (ASD). Previously, we found that adult male mice expressing the autism-associated SERT Ala56 variant have altered central serotonin (5-HT) system function, as well as elevated peripheral blood 5-HT levels. Early in gestation, before midbrain 5-HT projections have reached the cortex, peripheral sources supply 5-HT to the forebrain, suggesting that altered maternal or placenta 5-HT system function could impact the developing embryo. We therefore used different combinations of maternal and embryo SERT Ala56 genotypes to examine effects on blood, placenta and embryo serotonin levels and neurodevelopment at embryonic day E14.5, when peripheral sources of 5-HT predominate, and E18.5, when midbrain 5-HT projections have reached the forebrain. Maternal SERT Ala56 genotype was associated with decreased placenta and embryonic forebrain 5-HT levels at E14.5. Low 5-HT in the placenta persisted, but forebrain levels normalized by E18.5. Maternal SERT Ala56 genotype effects on forebrain 5-HT levels were accompanied by a broadening of 5-HT-sensitive thalamocortical axon projections. In contrast, no effect of embryo genotype was seen in concepti from heterozygous dams. Blood 5-HT levels were dynamic across pregnancy and were increased in SERT Ala56 dams at E14.5. Placenta RNA sequencing data at E14.5 indicated substantial impact of maternal SERT Ala56 genotype, with alterations in immune and metabolic-related pathways. Collectively, these findings indicate that maternal SERT function impacts offspring placental 5-HT levels, forebrain 5-HT levels, and neurodevelopment.
Collapse
Affiliation(s)
| | - Allison MJ Anacker
- Department of Psychiatry, Columbia University Medical School, New York, NY, USA
| | - Tiffany D Rogers
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nick Goeden
- Department of Cell and Neurobiology and Zilkha Neurogenetic Institute, University of Southern California School of Medicine, Los Angeles, CA, USA
| | - Elizabeth H Keller
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - C Gunnar Forsberg
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Travis M Kerr
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Carly LA Wender
- Department of Psychiatry, Columbia University Medical School, New York, NY, USA
| | - George M Anderson
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - Gregg D Stanwood
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Randy D Blakely
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alexandre Bonnin
- Department of Cell and Neurobiology and Zilkha Neurogenetic Institute, University of Southern California School of Medicine, Los Angeles, CA, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University Medical School, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Center for Autism and the Developing Brain, New York Presbyterian Hospital, New York, NY, USA
- Sackler Institute for Developmental Psychobiology, Columbia University, New York, NY, USA
| |
Collapse
|
21
|
Velasquez JC, Goeden N, Herod SM, Bonnin A. Maternal Pharmacokinetics and Fetal Disposition of (±)-Citalopram during Mouse Pregnancy. ACS Chem Neurosci 2016; 7:327-38. [PMID: 26765210 PMCID: PMC5384759 DOI: 10.1021/acschemneuro.5b00287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While selective-serotonin reuptake inhibitor (SSRI) antidepressants are commonly prescribed in the treatment of depression, their use during pregnancy leads to fetal drug exposures. According to recent reports, such exposures could affect fetal development and long-term offspring health. A central question is how pregnancy-induced physical and physiological changes in mothers, fetuses, and the placenta influence fetal SSRI exposures during gestation. In this study, we examined the effects of gestational stage on the maternal pharmacokinetics and fetal disposition of the SSRI (±)-citalopram (CIT) in a mouse model. We determined the maternal and fetal CIT serum concentration-time profiles following acute maternal administration on gestational days (GD)14 and GD18, as well as the fetal brain drug disposition. The results show that pregnancy affects the pharmacokinetics of CIT and that maternal drug clearance increases as gestation progresses. The data further show that CIT and its primary metabolite desmethylcitalopram (DCIT) readily cross the placenta into the fetal compartment, and fetal exposure to CIT exceeds that of the mother during gestation 2 h after maternal administration. Enzymatic activity assays revealed that fetal drug metabolic capacity develops in late gestation, resulting in elevated circulating and brain concentrations of DCIT at embryonic day (E)18. Fetal exposure to the SSRI CIT in murine pregnancy is therefore influenced by both maternal gestational stage and embryonic development, suggesting potential time-dependent effects on fetal brain development.
Collapse
Affiliation(s)
| | | | - Skyla M. Herod
- Department
of Biology and Chemistry, Azusa Pacific University, Azusa, California 91702, United States
| | | |
Collapse
|
22
|
Huang X, Lüthi M, Ontsouka EC, Kallol S, Baumann MU, Surbek DV, Albrecht C. Establishment of a confluent monolayer model with human primary trophoblast cells: novel insights into placental glucose transport. Mol Hum Reprod 2016; 22:442-56. [PMID: 26931579 DOI: 10.1093/molehr/gaw018] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/24/2016] [Indexed: 11/14/2022] Open
Abstract
STUDY HYPOTHESIS Using optimized conditions, primary trophoblast cells isolated from human term placenta can develop a confluent monolayer in vitro, which morphologically and functionally resembles the microvilli structure found in vivo. STUDY FINDING We report the successful establishment of a confluent human primary trophoblast monolayer using pre-coated polycarbonate inserts, where the integrity and functionality was validated by cell morphology, biophysical features, cellular marker expression and secretion, and asymmetric glucose transport. WHAT IS KNOWN ALREADY Human trophoblast cells form the initial barrier between maternal and fetal blood to regulate materno-fetal exchange processes. Although the method for isolating pure human cytotrophoblast cells was developed almost 30 years ago, a functional in vitro model with primary trophoblasts forming a confluent monolayer is still lacking. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Human term cytotrophoblasts were isolated by enzymatic digestion and density gradient separation. The purity of the primary cells was evaluated by flow cytometry using the trophoblast-specific marker cytokeratin 7, and vimentin as an indicator for potentially contaminating cells. We screened different coating matrices for high cell viability to optimize the growth conditions for primary trophoblasts on polycarbonate inserts. During culture, cell confluency and polarity were monitored daily by determining transepithelial electrical resistance (TEER) and permeability properties of florescent dyes. The time course of syncytia-related gene expression and hCG secretion during syncytialization were assessed by quantitative RT-PCR and enzyme-linked immunosorbent assay, respectively. The morphology of cultured trophoblasts after 5 days was determined by light microscopy, scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Membrane makers were visualized using confocal microscopy. Additionally, glucose transport studies were performed on the polarized trophoblasts in the same system. MAIN RESULTS AND THE ROLE OF CHANCE During 5-day culture, the highly pure trophoblasts were cultured on inserts coated with reconstituted basement membrane matrix . They exhibited a confluent polarized monolayer, with a modest TEER and a size-dependent apparent permeability coefficient (Papp) to fluorescently labeled compounds (MW ∼400-70 000 Da). The syncytialization progress was characterized by gradually increasing mRNA levels of fusogen genes and elevating hCG secretion. SEM analyses confirmed a confluent trophoblast layer with numerous microvilli, and TEM revealed a monolayer with tight junctions. Immunocytochemistry on the confluent trophoblasts showed positivity for the cell-cell adhesion molecule E-cadherin, the tight junction protein 1 (ZO-1) and the membrane proteins ATP-binding cassette transporter A1 (ABCA1) and glucose transporter 1 (GLUT1). Applying this model to study the bidirectional transport of a non-metabolizable glucose derivative indicated a carrier-mediated placental glucose transport mechanism with asymmetric kinetics. LIMITATIONS, REASONS FOR CAUTION The current study is only focused on primary trophoblast cells isolated from healthy placentas delivered at term. It remains to be evaluated whether this system can be extended to pathological trophoblasts isolated from diverse gestational diseases. WIDER IMPLICATIONS OF THE FINDINGS These findings confirmed the physiological properties of the newly developed human trophoblast barrier, which can be applied to study the exchange of endobiotics and xenobiotics between the maternal and fetal compartment, as well as intracellular metabolism, paracellular contributions and regulatory mechanisms influencing the vectorial transport of molecules. LARGE-SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This study was supported by the Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Switzerland, and the Swiss National Science Foundation (grant no. 310030_149958, C.A.). All authors declare that their participation in the study did not involve factual or potential conflicts of interests.
Collapse
Affiliation(s)
- Xiao Huang
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| | - Michael Lüthi
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| | - Edgar C Ontsouka
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| | - Sampada Kallol
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| | - Marc U Baumann
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland Department of Obstetrics and Gynecology, University Hospital, University of Bern, Bern, Switzerland
| | - Daniel V Surbek
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland Department of Obstetrics and Gynecology, University Hospital, University of Bern, Bern, Switzerland
| | - Christiane Albrecht
- Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| |
Collapse
|
23
|
Staud F, Ceckova M. Regulation of drug transporter expression and function in the placenta. Expert Opin Drug Metab Toxicol 2015; 11:533-55. [DOI: 10.1517/17425255.2015.1005073] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
24
|
Gil-Ortega M, Fernández-Alfonso MS, Somoza B, Casteilla L, Sengenès C. Ex vivo microperfusion system of the adipose organ: a new approach to studying the mobilization of adipose cell populations. Int J Obes (Lond) 2013; 38:1255-62. [PMID: 24357852 DOI: 10.1038/ijo.2013.243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/28/2013] [Accepted: 12/16/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND/OBJECTIVES Adipose tissue (AT) is a dynamic organ that expands and contracts rapidly. It is composed of adipocytes and of cell populations among which immune cells and mesenchymal progenitors known as adipose stromal cells (ASCs). The AT cell turnover has been extensively studied. Surprisingly it has only been viewed as the result of both cell proliferation/death and cell infiltration. Nevertheless, both immune cells and ASCs exhibit migration abilities; therefore their egress from AT in response to physiological/pathophysiological stimuli has to be considered. To do so, the aim of the present work was to develop a model allowing the study of cell release from the adipose organ. SUBJECTS/METHODS Mesenteric (Mes) ATs were isolated from 9-week-old C57BL/6 male mice and were catheterized via the superior mesenteric artery and were perfused with a saline solution. After an equilibration period, the mesenteric fat pad was perfused with CXCL12 (stromal-derived factor-1, SDF-1) or sphingosine 1-phosphate (S1P) to trigger cell mobilization and perfusates were collected every 30 min for subsequent flow cytometry analyses. RESULTS We report here that CXCL12 induces the specific release of ASCs from MesAT thus demonstrating that ASCs are specifically mobilized from fat depots by a CXCL12-dependent pathway. Moreover, we showed that leukocyte mobilization can be triggered via a S1P-dependent pathway. CONCLUSIONS We have developed a microperfusion model of an intact fat depot allowing the study of AT cell release in response to various molecules. The perfusion system described here demonstrates that ASCs and leukocytes can be pharmacologically mobilized from AT. Therefore, AT microperfusion might constitute an appropriate and reliable approach for evaluating the mobilization of different cell populations from AT in various physiological and pathophysiological contexts. Such a model might help in identifying factors and drugs controlling AT cell release, impacting the medical fields of regenerative medicine and of obesity or its associated comorbidities.
Collapse
Affiliation(s)
- M Gil-Ortega
- 1] Inserm U1031 STROMAlab BP 84 225-F-31 432, Toulouse, France [2] CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, BP 84 225-F-31 432, Toulouse, France [3] EFS (Etablissement Français du Sang), STROMAlab BP 84 225-F-31 432, Toulouse, France [4] Université Toulouse III, UPS UMR5273 STROMAlab BP 84 225-F-31 432, Toulouse, France
| | - M S Fernández-Alfonso
- Instituto Pluridisciplinar, Facultad de Farmacia, Universidad Complutense de Madrid, Juan XXIII 1, 28040 Madrid, Spain
| | - B Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28668 Madrid, Spain
| | - L Casteilla
- 1] Inserm U1031 STROMAlab BP 84 225-F-31 432, Toulouse, France [2] CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, BP 84 225-F-31 432, Toulouse, France [3] EFS (Etablissement Français du Sang), STROMAlab BP 84 225-F-31 432, Toulouse, France [4] Université Toulouse III, UPS UMR5273 STROMAlab BP 84 225-F-31 432, Toulouse, France
| | - C Sengenès
- 1] Inserm U1031 STROMAlab BP 84 225-F-31 432, Toulouse, France [2] CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, BP 84 225-F-31 432, Toulouse, France [3] EFS (Etablissement Français du Sang), STROMAlab BP 84 225-F-31 432, Toulouse, France [4] Université Toulouse III, UPS UMR5273 STROMAlab BP 84 225-F-31 432, Toulouse, France
| |
Collapse
|
25
|
Oury F, Khrimian L, Denny CA, Gardin A, Chamouni A, Goeden N, Huang YY, Lee H, Srinivas P, Gao XB, Suyama S, Langer T, Mann JJ, Horvath TL, Bonnin A, Karsenty G. Maternal and offspring pools of osteocalcin influence brain development and functions. Cell 2013; 155:228-41. [PMID: 24074871 DOI: 10.1016/j.cell.2013.08.042] [Citation(s) in RCA: 295] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/05/2013] [Accepted: 08/22/2013] [Indexed: 10/26/2022]
Abstract
The powerful regulation of bone mass exerted by the brain suggests the existence of bone-derived signals modulating this regulation or other functions of the brain. We show here that the osteoblast-derived hormone osteocalcin crosses the blood-brain barrier, binds to neurons of the brainstem, midbrain, and hippocampus, enhances the synthesis of monoamine neurotransmitters, inhibits GABA synthesis, prevents anxiety and depression, and favors learning and memory independently of its metabolic functions. In addition to these postnatal functions, maternal osteocalcin crosses the placenta during pregnancy and prevents neuronal apoptosis before embryos synthesize this hormone. As a result, the severity of the neuroanatomical defects and learning and memory deficits of Osteocalcin(-/-) mice is determined by the maternal genotype, and delivering osteocalcin to pregnant Osteocalcin(-/-) mothers rescues these abnormalities in their Osteocalcin(-/-) progeny. This study reveals that the skeleton via osteocalcin influences cognition and contributes to the maternal influence on fetal brain development.
Collapse
Affiliation(s)
- Franck Oury
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Velasquez JC, Goeden N, Bonnin A. Placental serotonin: implications for the developmental effects of SSRIs and maternal depression. Front Cell Neurosci 2013; 7:47. [PMID: 23630464 PMCID: PMC3632750 DOI: 10.3389/fncel.2013.00047] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/03/2013] [Indexed: 01/15/2023] Open
Abstract
In addition to its role in the pathophysiology of numerous psychiatric disorders, increasing evidence points to serotonin (5-HT) as a crucial molecule for the modulation of neurodevelopmental processes. Recent evidence indicates that the placenta is involved in the synthesis of 5-HT from maternally derived tryptophan (TRP). This gives rise to the possibility that genetic and environmental perturbations directly affecting placental TRP metabolism may lead to abnormal brain circuit wiring in the developing embryo, and therefore contribute to the developmental origin of psychiatric disorders. In this review, we discuss how perturbations of the placental TRP metabolic pathway may lead to abnormal brain development and function throughout life. Of particular interest is prenatal exposure to maternal depression and antidepressants, both known to alter fetal development. We review existing evidence on how antidepressants can alter placental physiology in its key function of maintaining fetal homeostasis and have long-term effects on fetal forebrain development.
Collapse
Affiliation(s)
- Juan C Velasquez
- Department of Cell and Neurobiology, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California Los Angeles, CA, USA
| | | | | |
Collapse
|