1
|
Shimada H, Powell TL, Jansson T. Regulation of placental amino acid transport in health and disease. Acta Physiol (Oxf) 2024; 240:e14157. [PMID: 38711335 PMCID: PMC11162343 DOI: 10.1111/apha.14157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Abnormal fetal growth, i.e., intrauterine growth restriction (IUGR) or fetal growth restriction (FGR) and fetal overgrowth, is associated with increased perinatal morbidity and mortality and is strongly linked to the development of metabolic and cardiovascular disease in childhood and later in life. Emerging evidence suggests that changes in placental amino acid transport may contribute to abnormal fetal growth. This review is focused on amino acid transport in the human placenta, however, relevant animal models will be discussed to add mechanistic insights. At least 25 distinct amino acid transporters with different characteristics and substrate preferences have been identified in the human placenta. Of these, System A, transporting neutral nonessential amino acids, and System L, mediating the transport of essential amino acids, have been studied in some detail. Importantly, decreased placental Systems A and L transporter activity is strongly associated with IUGR and increased placental activity of these two amino acid transporters has been linked to fetal overgrowth in human pregnancy. An array of factors in the maternal circulation, including insulin, IGF-1, and adiponectin, and placental signaling pathways such as mTOR, have been identified as key regulators of placental Systems A and L. Studies using trophoblast-specific gene targeting in mice have provided compelling evidence that changes in placental Systems A and L are mechanistically linked to altered fetal growth. It is possible that targeting specific placental amino acid transporters or their upstream regulators represents a novel intervention to alleviate the short- and long-term consequences of abnormal fetal growth in the future.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Departments of Obstetrics & Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Theresa L Powell
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| | - Thomas Jansson
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| |
Collapse
|
2
|
Cirulli F, De Simone R, Musillo C, Ajmone-Cat MA, Berry A. Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients 2022; 14:nu14153150. [PMID: 35956326 PMCID: PMC9370669 DOI: 10.3390/nu14153150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a main risk factor for the onset and the precipitation of many non-communicable diseases. This condition, which is associated with low-grade chronic systemic inflammation, is of main concern during pregnancy leading to very serious consequences for the new generations. In addition to the prominent role played by the adipose tissue, dysbiosis of the maternal gut may also sustain the obesity-related inflammatory milieu contributing to create an overall suboptimal intrauterine environment. Such a condition here generically defined as “inflamed womb” may hold long-term detrimental effects on fetal brain development, increasing the vulnerability to mental disorders. In this review, we will examine the hypothesis that maternal obesity-related gut dysbiosis and the associated inflammation might specifically target fetal brain microglia, the resident brain immune macrophages, altering neurodevelopmental trajectories in a sex-dependent fashion. We will also review some of the most promising nutritional strategies capable to prevent or counteract the effects of maternal obesity through the modulation of inflammation and oxidative stress or by targeting the maternal microbiota.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| | - Roberta De Simone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Antonietta Ajmone-Cat
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| |
Collapse
|
3
|
Hypoxia and oxidative stress induce sterile placental inflammation in vitro. Sci Rep 2021; 11:7281. [PMID: 33790316 PMCID: PMC8012380 DOI: 10.1038/s41598-021-86268-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Fetal growth restriction (FGR) and stillbirth are associated with placental dysfunction and inflammation and hypoxia, oxidative and nitrative stress are implicated in placental damage. Damage-associated molecular patterns (DAMPs) are elevated in pregnancies at increased risk of FGR and stillbirth and are associated with increase in pro-inflammatory placental cytokines. We hypothesised that placental insults lead to release of DAMPs, promoting placental inflammation. Placental tissue from uncomplicated pregnancies was exposed in vitro to hypoxia, oxidative or nitrative stress. Tissue production and release of DAMPs and cytokines was determined. Oxidative stress and hypoxia caused differential release of DAMPs including uric acid, HMGB1, S100A8, cell-free fetal DNA, S100A12 and HSP70. After oxidative stress pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, IL-8, TNFα, CCL2) were increased both within explants and in conditioned culture medium. Hypoxia increased tissue IL-1α/β, IL-6, IL-8 and TNFα levels, and release of IL-1α, IL-6 and IL-8, whereas CCL2 and IL-10 were reduced. IL1 receptor antagonist (IL1Ra) treatment prevented hypoxia- and oxidative stress-induced IL-6 and IL-8 release. These findings provide evidence that relevant stressors induce a sterile inflammatory profile in placental tissue which can be partially blocked by IL1Ra suggesting this agent has translational potential to prevent placental inflammation evident in FGR and stillbirth.
Collapse
|
4
|
Sun C, Groom KM, Oyston C, Chamley LW, Clark AR, James JL. The placenta in fetal growth restriction: What is going wrong? Placenta 2020; 96:10-18. [PMID: 32421528 DOI: 10.1016/j.placenta.2020.05.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
The placenta is essential for the efficient delivery of nutrients and oxygen from mother to fetus to maintain normal fetal growth. Dysfunctional placental development underpins many pregnancy complications, including fetal growth restriction (FGR) a condition in which the fetus does not reach its growth potential. The FGR placenta is smaller than normal placentae throughout gestation and displays maldevelopment of both the placental villi and the fetal vasculature within these villi. Specialized epithelial cells called trophoblasts exhibit abnormal function and development in FGR placentae. This includes an altered balance between proliferation and apoptotic death, premature cellular senescence, and reduced colonisation of the maternal decidual tissue. Thus, the placenta undergoes aberrant changes at the macroscopic to cellular level in FGR, which can limit exchange capacity and downstream fetal growth. This review aims to compile stereological, in vitro, and imaging data to create a holistic overview of the FGR placenta and its pathophysiology, with a focus on the contribution of trophoblasts.
Collapse
Affiliation(s)
- Cherry Sun
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Katie M Groom
- Liggins Institute, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Charlotte Oyston
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, The University of Auckland, Auckland Bioengineering, House, Level 6/70 Symonds Street, Grafton, Auckland, 1010, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
5
|
Luo Z, Xu X, Sho T, Luo W, Zhang J, Xu W, Yao J, Xu J. Effects of n-acetyl-cysteine supplementation in late gestational diet on maternal-placental redox status, placental NLRP3 inflammasome, and fecal microbiota in sows1. J Anim Sci 2019; 97:1757-1771. [PMID: 30789643 DOI: 10.1093/jas/skz058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 12/14/2022] Open
Abstract
Although n-acetyl-cysteine (NAC) has been shown to efficiently alleviate oxidative stress, inflammatory response, and alter gut microbiota, little attention has been focused on their interactions with placental metabolic status of sows. The effects of NAC on the placental redox status, function, inflammasome, and fecal microbiota in sows were explored to clarify the correlation between the fecal microbiota and placenta. Sows were divided into either the control group or the NAC group which received dietary 0.5% NAC supplementation from day 85 of gestation to delivery. Plasma redox status, placental growth factors, nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome, fecal microbial metabolites, and communities were evaluated. Compared with the control group, although NAC did not ameliorate reproductive performance of sows (P > 0.05), it significantly improved maternal-placental health, which was accompanied by increased activities of glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD), decreased level of malondialdehyde (MDA), and lowered expression of interleukin (IL)-1β and IL-18 through inhibiting NLRP3 inflammasome (P < 0.05). Additionally, NAC significantly increased placental insulin-like growth factors (IGFs) and E-cadherin contents (P < 0.05), elevated the expression of genes involved in angiogenesis and amino acids transporters (P < 0.05), and decreased the microtubule-associated protein light chain 3B (LC3B) and Beclin-1 protein expression (P < 0.05). Furthermore, NAC increased the relative abundances of fecal Prevotella, Clostridium cluster XIVa, and Roseburial/Eubacterium rectale (P < 0.05), which were negatively correlated with placental NLRP3 and positively with solute carrier family 7, member 8 (Slc7a8; P < 0.05). In conclusion, NAC supplementation during late gestation alleviated maternal-placental oxidative stress and inflammatory response, improved placental function, and altered fecal microbial communities.
Collapse
Affiliation(s)
- Zhen Luo
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Xu
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Takami Sho
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Wenli Luo
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhang
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Weina Xu
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jianbo Yao
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV
| | - Jianxiong Xu
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Herrera EA, Cifuentes-Zúñiga F, Figueroa E, Villanueva C, Hernández C, Alegría R, Arroyo-Jousse V, Peñaloza E, Farías M, Uauy R, Casanello P, Krause BJ. N-Acetylcysteine, a glutathione precursor, reverts vascular dysfunction and endothelial epigenetic programming in intrauterine growth restricted guinea pigs. J Physiol 2016; 595:1077-1092. [PMID: 27739590 DOI: 10.1113/jp273396] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/11/2016] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Intrauterine growth restriction (IUGR) is associated with vascular dysfunction, oxidative stress and signs of endothelial epigenetic programming of the umbilical vessels. There is no evidence that this epigenetic programming is occurring on systemic fetal arteries. In IUGR guinea pigs we studied the functional and epigenetic programming of endothelial nitric oxide synthase (eNOS) (Nos3 gene) in umbilical and systemic fetal arteries, addressing the role of oxidative stress in this process by maternal treatment with N-acetylcysteine (NAC) during the second half of gestation. The present study suggests that IUGR endothelial cells have common molecular markers of programming in umbilical and systemic arteries. Notably, maternal treatment with NAC restores fetal growth by increasing placental efficiency and reverting the functional and epigenetic programming of eNOS in arterial endothelium in IUGR guinea pigs. ABSTRACT In humans, intrauterine growth restriction (IUGR) is associated with vascular dysfunction, oxidative stress and signs of endothelial programming in umbilical vessels. We aimed to determine the effects of maternal antioxidant treatment with N-acetylcysteine (NAC) on fetal endothelial function and endothelial nitric oxide synthase (eNOS) programming in IUGR guinea pigs. IUGR was induced by implanting ameroid constrictors on uterine arteries of pregnant guinea pigs at mid gestation, half of the sows receiving NAC in the drinking water (from day 34 until term). Fetal biometry and placental vascular resistance were followed by ultrasound throughout gestation. At term, umbilical arteries and fetal aortae were isolated to assess endothelial function by wire-myography. Primary cultures of endothelial cells (ECs) from fetal aorta, femoral and umbilical arteries were used to determine eNOS mRNA levels by quantitative PCR and analyse DNA methylation in the Nos3 promoter by pyrosequencing. Doppler ultrasound measurements showed that NAC reduced placental vascular resistance in IUGR (P < 0.05) and recovered fetal weight (P < 0.05), increasing fetal-to-placental ratio at term (∼40%) (P < 0.001). In IUGR, NAC treatment restored eNOS-dependent relaxation in aorta and umbilical arteries (P < 0.05), normalizing eNOS mRNA levels in EC fetal and umbilical arteries (P < 0.05). IUGR-derived ECs had a decreased DNA methylation (∼30%) at CpG -170 (from the transcription start site) and this epigenetic signature was absent in NAC-treated fetuses (P < 0.001). These data show that IUGR-ECs have common molecular markers of eNOS programming in umbilical and systemic arteries and this effect is prevented by maternal treatment with antioxidants.
Collapse
Affiliation(s)
- Emilio A Herrera
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Francisca Cifuentes-Zúñiga
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - Esteban Figueroa
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Cristian Villanueva
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Cherie Hernández
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile.,Division of Obstetrics & Gynaecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - René Alegría
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Viviana Arroyo-Jousse
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - Estefania Peñaloza
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - Marcelo Farías
- Division of Obstetrics & Gynaecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - Ricardo Uauy
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - Paola Casanello
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile.,Division of Obstetrics & Gynaecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| | - Bernardo J Krause
- Department of Neonatology, Division of Paediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago 8330024, Santiago, Chile
| |
Collapse
|
7
|
Bertolaccini ML, Contento G, Lennen R, Sanna G, Blower PJ, Ma MT, Sunassee K, Girardi G. Complement inhibition by hydroxychloroquine prevents placental and fetal brain abnormalities in antiphospholipid syndrome. J Autoimmun 2016; 75:30-38. [PMID: 27160365 PMCID: PMC6203312 DOI: 10.1016/j.jaut.2016.04.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/21/2016] [Accepted: 04/24/2016] [Indexed: 11/24/2022]
Abstract
Placental ischemic disease and adverse pregnancy outcomes are frequently observed in patients with antiphospholipid syndrome (APS). Despite the administration of conventional antithrombotic treatment a significant number of women continue to experience adverse pregnancy outcomes, with uncertain prevention and management. Efforts to develop effective pharmacological strategies for refractory obstetric APS cases will be of significant clinical benefit for both mothers and fetuses. Although the antimalarial drug, hydroxychloroquine (HCQ) is increasingly used to treat pregnant women with APS, little is known about its efficacy and mechanism of action of HCQ. Because complement activation plays a crucial and causative role in placental ischemia and abnormal fetal brain development in APS we hypothesised that HCQ prevents these pregnancy complications through inhibition of complement activation. Using a mouse model of obstetric APS that closely resembles the clinical condition, we found that HCQ prevented fetal death and the placental metabolic changes -measured by proton magnetic resonance spectroscopy in APS-mice. Using 111In labelled antiphospholipid antibodies (aPL) we identified the placenta and the fetal brain as the main organ targets in APS-mice. Using this same method, we found that HCQ does not inhibit aPL binding to tissues as was previously suggested from in vitro studies. While HCQ did not affect aPL binding to fetal brain it prevented fetal brain abnormal cortical development. HCQ prevented complement activation in vivo and in vitro. Complement C5a levels in serum samples from APS patients and APS-mice were lower after treatment with HCQ while the antibodies titres remained unchanged. HCQ prevented not only placental insufficiency but also abnormal fetal brain development in APS. By inhibiting complement activation, HCQ might also be an effective antithrombotic therapy.
Collapse
Affiliation(s)
| | - Gregorio Contento
- Division of Women's Health, St Thomas' Hospital, King's College London, London, SE1 7EH, UK
| | - Ross Lennen
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Giovanni Sanna
- Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, St Thomas' Hospital, London, SE1 7EH, UK
| | - Philip J Blower
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, SE1 7EH, UK
| | - Michelle T Ma
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, SE1 7EH, UK
| | - Kavitha Sunassee
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, SE1 7EH, UK
| | - Guillermina Girardi
- Division of Women's Health, St Thomas' Hospital, King's College London, London, SE1 7EH, UK; MRC Centre for Inflammation Research, Queen's Medical Research Institute University of Edinburgh, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
8
|
Maternal obesity is associated with a reduction in placental taurine transporter activity. Int J Obes (Lond) 2014; 39:557-64. [PMID: 25547282 PMCID: PMC4389721 DOI: 10.1038/ijo.2014.212] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/04/2014] [Accepted: 11/21/2014] [Indexed: 02/07/2023]
Abstract
Background/Objectives: Maternal obesity increases the risk of poor pregnancy outcome including stillbirth, pre-eclampsia, fetal growth restriction and fetal overgrowth. These pregnancy complications are associated with dysfunctional syncytiotrophoblast, the transporting epithelium of the human placenta. Taurine, a β-amino acid with antioxidant and cytoprotective properties, has a role in syncytiotrophoblast development and function and is required for fetal growth and organ development. Taurine is conditionally essential in pregnancy and fetal tissues depend on uptake of taurine from maternal blood. We tested the hypothesis that taurine uptake into placental syncytiotrophoblast by the taurine transporter protein (TauT) is lower in obese women (body mass index (BMI)⩾30 kg m−2) than in women of ideal weight (BMI 18.5–24.9 kg m−2) and explored potential regulatory factors. Subjects/Methods: Placentas were collected from term (37–42-week gestation), uncomplicated, singleton pregnancies from women with BMI 19–49 kg m−2. TauT activity was measured as the Na+-dependent uptake of 3H-taurine into placental villous fragments. TauT expression in membrane-enriched placental samples was investigated by western blot. In vitro studies using placental villous explants examined whether leptin or IL-6, adipokines/cytokines that are elevated in maternal obesity, regulates TauT activity. Results: Placental TauT activity was significantly lower in obese women (BMI⩾30) than women of ideal weight (P<0.03) and inversely related to maternal BMI (19–49 kg m−2; P<0.05; n=61). There was no difference in TauT expression between placentas of ideal weight and obese class III (BMI⩾40) subjects. Long-term exposure (48 h) of placental villous explants to leptin or IL-6 did not affect TauT activity. Conclusions: Placental TauT activity at term is negatively related to maternal BMI. We propose that the reduction in placental TauT activity in maternal obesity could lower syncytiotrophoblast taurine concentration, compromise placental development and function, and reduce the driving force for taurine efflux to the fetus, thereby increasing the risk of poor pregnancy outcome.
Collapse
|
9
|
Abad C, Vallejos C, De Gregorio N, Díaz P, Chiarello DI, Mendoza M, Piñero S, Proverbio T, Botana D, Rojas P, Riquelme G, Proverbio F, Marín R. Na⁺, K⁺-ATPase and Ca²⁺-ATPase activities in basal and microvillous syncytiotrophoblast membranes from preeclamptic human term placenta. Hypertens Pregnancy 2014; 34:65-79. [PMID: 25356531 DOI: 10.3109/10641955.2014.973038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The aim of this study is to evaluate the effect of preeclampsia on the level of lipid peroxidation, activity and expression of both plasma membrane Ca(2+)- and Na(+), K(+)-ATPases in syncytiotrophoblast. METHODS The level of lipid peroxidation was estimated by measuring TBARS. ATPase activities were quantified by a colorimetric method measuring the amount of inorganic phosphate during the assay. Expression of Ca(2+)- and Na(+), K(+)-ATPases in syncytiotrophoblast plasma membranes and term placenta tissue sections was investigated using Western blot and immunohistochemistry, respectively. RESULTS Our results show a higher level of lipid peroxidation of syncytiotrophoblast plasma membranes from preeclamptic, as compared to uncomplicated pregnant women. Preeclampsia also significantly reduced the activity of Ca(2+)- and Na(+), K(+)-ATPases; however, expression of both ATPases was unaffected. CONCLUSION Our findings suggest that the reduction of Ca(2+)- and Na(+), K(+)-ATPase activities during preeclampsia could be at least partially due to an increased level of lipid peroxidation of the syncytiotrophoblast plasma membranes.
Collapse
Affiliation(s)
- Cilia Abad
- Laboratorio de Bioenergética Celular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC) , Caracas , Venezuela , and
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Araújo JR, Correia-Branco A, Pereira AC, Pinho MJ, Keating E, Martel F. Oxidative stress decreases uptake of neutral amino acids in a human placental cell line (BeWo cells). Reprod Toxicol 2013; 40:76-81. [PMID: 23806338 DOI: 10.1016/j.reprotox.2013.06.073] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 05/29/2013] [Accepted: 06/15/2013] [Indexed: 02/07/2023]
Abstract
Increased oxidative stress (OS) is implicated in the pathophysiology of several pregnancy disorders. We aimed to investigate the effect of tert-butylhydroperoxide (TBHP)-induced OS upon the placental transport of the neutral amino acids L-methionine (L-Met) and L-alanine (L-Ala), by using a human trophoblast cell model (BeWo cells). TBHP reduced both total and Na(+)-independent (14)C-L-Met intracellular steady-state accumulation over time (Amax), by reducing non-system L-mediated uptake - most probably system y(+) - while having no effect on system L. Moreover, TBHP reduced total (14)C-L-Ala Amax through an inhibition of system A. The effect of TBHP upon total, but not system A-mediated, (14)C-L-Ala uptake was dependent upon phosphoinositide 3-kinase (PI3K) and protein kinase C (PKC) activation, and was completely prevented by the polyphenol quercetin. In conclusion, a reduction in placental uptake of neutral amino acids may contribute to the deleterious effects of pregnancy disorders associated with OS.
Collapse
Affiliation(s)
- João Ricardo Araújo
- Department of Biochemistry (U38-FCT), Faculty of Medicine, University of Porto, Porto, Portugal.
| | | | | | | | | | | |
Collapse
|
11
|
Desforges M, Parsons L, Westwood M, Sibley CP, Greenwood SL. Taurine transport in human placental trophoblast is important for regulation of cell differentiation and survival. Cell Death Dis 2013; 4:e559. [PMID: 23519128 PMCID: PMC3618382 DOI: 10.1038/cddis.2013.81] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The outer epithelial cell layer of human placenta, the syncytiotrophoblast, is a specialised terminally differentiated multinucleate tissue. It is generated and renewed from underlying cytotrophoblast cells that undergo proliferation, differentiation and fusion with syncytiotrophoblast. Acquisition of fresh cellular components is thought to be balanced by apoptosis and shedding of aged nuclei. This process of trophoblast cell turnover maintains a functional syncytiotrophoblast, capable of sufficient nutrient transfer from mother to foetus. Foetal growth restriction (FGR) is a pregnancy complication associated with aberrant trophoblast turnover and reduced activity of certain amino acid transporters, including the taurine transporter (TauT). Taurine is the most abundant amino acid in human placenta implying an important physiological role within this tissue. Unlike other amino acids, taurine is not incorporated into proteins and in non-placental cell types represents an important osmolyte involved in cell volume regulation, and is also cytoprotective. Here, we investigated the role of taurine in trophoblast turnover using RNA interference to deplete primary human trophoblast cells of TauT and reduce intracellular taurine content. Trophoblast differentiation was compromised in TauT-deficient cells, and susceptibility of these cells to an inflammatory cytokine that is elevated in FGR was increased, evidenced by elevated levels of apoptosis. These data suggest an important role for taurine in trophoblast turnover and cytoprotection.
Collapse
Affiliation(s)
- M Desforges
- Maternal and Foetal Health Research Centre, Institute of Human Development, University of Manchester, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK.
| | | | | | | | | |
Collapse
|
12
|
Regulation of nutrient transport across the placenta. J Pregnancy 2012; 2012:179827. [PMID: 23304511 PMCID: PMC3523549 DOI: 10.1155/2012/179827] [Citation(s) in RCA: 281] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 09/02/2012] [Indexed: 12/20/2022] Open
Abstract
Abnormal fetal growth, both growth restriction and overgrowth, is associated with perinatal complications and an increased risk of metabolic and cardiovascular disease later in life. Fetal growth is dependent on nutrient availability, which in turn is related to the capacity of the placenta to transport these nutrients. The activity of a range of nutrient transporters has been reported to be decreased in placentas of growth restricted fetuses, whereas at least some studies indicate that placental nutrient transport is upregulated in fetal overgrowth. These findings suggest that changes in placental nutrient transport may directly contribute to the development of abnormal fetal growth. Detailed information on the mechanisms by which placental nutrient transporters are regulated will therefore help us to better understand how important pregnancy complications develop and may provide a foundation for designing novel intervention strategies. In this paper we will focus on recent studies of regulatory mechanisms that modulate placental transport of amino acids, fatty acids, and glucose.
Collapse
|
13
|
Huang LT, Hsieh CS, Chang KA, Tain YL. Roles of nitric oxide and asymmetric dimethylarginine in pregnancy and fetal programming. Int J Mol Sci 2012. [PMID: 23203083 PMCID: PMC3509599 DOI: 10.3390/ijms131114606] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nitric oxide (NO) regulates placental blood flow and actively participates in trophoblast invasion and placental development. Asymmetric dimethylarginine (ADMA) can inhibit NO synthase, which generates NO. ADMA has been associated with uterine artery flow disturbances such as preeclampsia. Substantial experimental evidence has reliably supported the hypothesis that an adverse in utero environment plays a role in postnatal physiological and pathophysiological programming. Growing evidence suggests that the placental nitrergic system is involved in epigenetic fetal programming. In this review, we discuss the roles of NO and ADMA in normal and compromised pregnancies as well as the link between placental insufficiency and epigenetic fetal programming.
Collapse
Affiliation(s)
- Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
- Department of Traditional Chinese Medicine, Chang Gung University, Linkow 333, Taiwan
| | - Chih-Sung Hsieh
- Department of Pediatric Surgery, Pingtung Christian Hospital, Pingtung 900, Taiwan; E-Mail:
- Department of Nursing, MeiHo University, Pingtung 900, Taiwan
| | - Kow-Aung Chang
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-975056995; Fax: +886-7-7338009
| |
Collapse
|
14
|
Placental amino acids transport in intrauterine growth restriction. J Pregnancy 2012; 2012:972562. [PMID: 22997583 PMCID: PMC3401547 DOI: 10.1155/2012/972562] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 04/19/2012] [Indexed: 01/08/2023] Open
Abstract
The placenta represents a key organ for fetal growth as it acts as an interface between mother and fetus, regulating the fetal-maternal exchange of nutrients, gases, and waste products. During pregnancy, amino acids represent one of the major nutrients for fetal life, and both maternal and fetal concentrations are significantly different in pregnancies with intrauterine growth restriction when compared to uncomplicated pregnancies. The transport of amino acids across the placenta is a complex process that includes the influx of neutral, anionic, and cationic amino acids across the microvilluos plasma membrane of the syncytiotrophoblast, the passage through the cytoplasm of the trophoblasts, and the transfer outside the trophoblasts across the basal membrane into the fetal circulation. In this paper, we review the transport mechanisms of amino acids across the placenta in normal pregnancies and in pregnancies complicated by intrauterine growth restriction.
Collapse
|
15
|
Lappas M, Hiden U, Desoye G, Froehlich J, Hauguel-de Mouzon S, Jawerbaum A. The role of oxidative stress in the pathophysiology of gestational diabetes mellitus. Antioxid Redox Signal 2011; 15:3061-100. [PMID: 21675877 DOI: 10.1089/ars.2010.3765] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Normal human pregnancy is considered a state of enhanced oxidative stress. In pregnancy, it plays important roles in embryo development, implantation, placental development and function, fetal development, and labor. However, pathologic pregnancies, including gestational diabetes mellitus (GDM), are associated with a heightened level of oxidative stress, owing to both overproduction of free radicals and/or a defect in the antioxidant defenses. This has important implications on the mother, placental function, and fetal well-being. Animal models of diabetes have confirmed the important role of oxidative stress in the etiology of congenital malformations; the relative immaturity of the antioxidant system facilitates the exposure of embryos and fetuses to the damaging effects of oxidative stress. Of note, there are only a few clinical studies evaluating the potential beneficial effects of antioxidants in GDM. Thus, whether or not increased antioxidant intake can reduce the complications of GDM in both mother and fetus needs to be explored. This review provides an overview and updated data on our current understanding of the complications associated with oxidative changes in GDM.
Collapse
Affiliation(s)
- Martha Lappas
- Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
16
|
Koo TS, Kwon HJ, Kim MH, Kim DD, Shim CK, Chung SJ, Chong S. Functional impairment of rat taurine transporter by activation of nitrogen oxide through superoxide. Drug Metab Pharmacokinet 2011; 27:286-93. [PMID: 22166889 DOI: 10.2133/dmpk.dmpk-11-rg-076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The objective of this study was to identify the nitrogen oxide form(s) involved in the functional impairment of the rat taurine transport system. Taurine uptake activity in the rat renal brush border membrane vesicle (RBBMV) preparation or Xenopus laevis oocytes that express the rat taurine transporter was compared after the pretreatment with nitrogen oxide donors from which nitric oxide (NO) is released at different rates. The functional impairment was associated with a reduced Vmax, but did not involve an alteration in the Km, of taurine uptake in the RBBMV preparation that had been pretreated with sodium nitroprusside, a slow release nitric oxide (NO) donor. When the preparation was pretreated with S-nitroso-N-acetyl penicillamine, a rapid release NO donor, the activity of taurine uptake was unaffected. The activity was not statistically different from the control after the pretreatment with sodium nitroprusside and superoxide dismutase. Consistent with the study with RBBMV, a similar alteration in the activity of taurine uptake by NO donors was observed in oocytes expressing the transporter. Considering the fact that peroxynitrite, a highly reactive nitrogen oxide form, is formed by the reaction between NO and superoxide, the taurine transporter, and probably other transport systems as well, may be functionally impaired by peroxynitrite.
Collapse
Affiliation(s)
- Tae-Sung Koo
- Life Science R&D Park, SK Biopharmaceuticals Co., Ltd., Daejeon, Korea
| | | | | | | | | | | | | |
Collapse
|
17
|
Lee NY, Kang YS. Regulation of taurine transport at the blood-placental barrier by calcium ion, PKC activator and oxidative stress conditions. J Biomed Sci 2010; 17 Suppl 1:S37. [PMID: 20804613 PMCID: PMC2994386 DOI: 10.1186/1423-0127-17-s1-s37] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the present study, we investigated the changes of uptake and efflux transport of taurine under various stress conditions using rat conditionally immortalized syncytiotrophoblast cell line (TR-TBT cells), as in vitro blood-placental barrier (BPB) model. METHODS The transport of taurine in TR-TBT cells were characterized by cellular uptake study using radiolabeled taurine. The efflux of taurine was measured from the amount of radiolabeled taurine remaining in the cells after the uptake of radiolabeled taurine for 60 min. RESULTS Taurine uptake was significantly decreased by phosphorylation of protein kinase C (PKC) activator in TR-TBT cells. Also, calcium ion (Ca2+) was involved in taurine transport in TR-TBT cells. Taurine uptake was inhibited and efflux was enhanced under calcium free conditions in the cells. In addition, oxidative stress induced the change of taurine transport in TR-TBT cells, but the changes were different depending on the types of oxidative stress inducing agents. Tumor necrosis factor-alpha (TNF-alpha), lipopolysaccharide (LPS) and diethyl maleate (DEM) significantly increased taurine uptake, but H2O2 and nitric oxide (NO) donor decreased taurine uptake in the cells. Taurine efflux was down-regulated by TNF-alpha in TR-TBT cells. CONCLUSION Taurine transport in TR-TBT cells were regulated diversely at extracellular Ca2+ level, PKC activator and oxidative stress conditions. It suggested that variable stresses affected the taurine supplies from maternal blood to fetus and taurine level of fetus.
Collapse
Affiliation(s)
- Na-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Science, Sookmyung Women's University, Seoul, 140-742, Republic of Korea.
| | | |
Collapse
|
18
|
Roos S, Kanai Y, Prasad PD, Powell TL, Jansson T. Regulation of placental amino acid transporter activity by mammalian target of rapamycin. Am J Physiol Cell Physiol 2009; 296:C142-50. [DOI: 10.1152/ajpcell.00330.2008] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The activity of placental amino acid transporters is decreased in intrauterine growth restriction (IUGR), but the underlying regulatory mechanisms have not been established. Inhibition of the mammalian target of rapamycin (mTOR) signaling pathway has been shown to decrease the activity of the system L amino acid transporter in human placental villous fragments, and placental mTOR activity is decreased in IUGR. In the present study, we used cultured primary trophoblast cells to study mTOR regulation of placental amino acid transporters in more detail and to test the hypothesis that mTOR alters amino acid transport activity by changes in transporter expression. Inhibition of mTOR by rapamycin significantly reduced the activity of system A (−17%), system L (−28%), and taurine (−40%) amino acid transporters. mRNA expression of isoforms of the three amino acid transporter systems in response to mTOR inhibition was measured using quantitative real-time PCR. mRNA expression of l-type amino acid transporter 1 (LAT1; a system L isoform) and taurine transporter was reduced by 13% and 50%, respectively; however, mTOR inhibition did not alter the mRNA expression of system A isoforms (sodium-coupled neutral amino acid transporter-1, -2, and -4), LAT2, or 4F2hc. Rapamycin treatment did not significantly affect the protein expression of any of the transporter isoforms. We conclude that mTOR signaling regulates the activity of key placental amino acid transporters and that this effect is not due to a decrease in total protein expression. These data suggest that mTOR regulates placental amino acid transporters by posttranslational modifications or by affecting transporter translocation to the plasma membrane.
Collapse
|
19
|
Jansson T, Powell TL. Role of the placenta in fetal programming: underlying mechanisms and potential interventional approaches. Clin Sci (Lond) 2007; 113:1-13. [PMID: 17536998 DOI: 10.1042/cs20060339] [Citation(s) in RCA: 355] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adverse influences during fetal life alter the structure and function of distinct cells, organ systems or homoeostatic pathways, thereby ‘programming’ the individual for an increased risk of developing cardiovascular disease and diabetes in adult life. Fetal programming can be caused by a number of different perturbations in the maternal compartment, such as altered maternal nutrition and reduced utero–placental blood flow; however, the underlying mechanisms remain to be fully established. Perturbations in the maternal environment must be transmitted across the placenta in order to affect the fetus. Here, we review recent insights into how the placenta responds to changes in the maternal environment and discuss possible mechanisms by which the placenta mediates fetal programming. In IUGR (intrauterine growth restriction) pregnancies, the increased placental vascular resistance subjects the fetal heart to increased work load, representing a possible direct link between altered placental structure and fetal programming of cardiovascular disease. A decreased activity of placental 11β-HSD-2 (type 2 isoform of 11β-hydroxysteroid dehydrogenase) activity can increase fetal exposure to maternal cortisol, which programmes the fetus for later hypertension and metabolic disease. The placenta appears to function as a nutrient sensor regulating nutrient transport according to the ability of the maternal supply line to deliver nutrients. By directly regulating fetal nutrient supply and fetal growth, the placenta plays a central role in fetal programming. Furthermore, perturbations in the maternal compartment may affect the methylation status of placental genes and increase placental oxidative/nitrative stress, resulting in changes in placental function. Intervention strategies targeting the placenta in order to prevent or alleviate altered fetal growth and/or fetal programming include altering placental growth and nutrient transport by maternally administered IGFs (insulin-like growth factors) and altering maternal levels of methyl donors.
Collapse
Affiliation(s)
- Thomas Jansson
- Department of Obstetrics and Gynecology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| | | |
Collapse
|
20
|
Shibata E, Powers RW, Rajakumar A, von Versen-Höynck F, Gallaher MJ, Lykins DL, Roberts JM, Hubel CA. Angiotensin II decreases system A amino acid transporter activity in human placental villous fragments through AT1 receptor activation. Am J Physiol Endocrinol Metab 2006; 291:E1009-16. [PMID: 16787961 DOI: 10.1152/ajpendo.00134.2006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reduced transport of amino acids from mother to fetus can lead to fetal intrauterine growth restriction (IUGR). The activities of several amino acid transport systems, including system A, are decreased in placental syncytiotrophoblast of IUGR pregnancies. Na(+)-K(+)-ATPase activity provides an essential driving force for Na(+)-coupled system A transport, is decreased in the placenta of IUGR pregnancies, and is decreased by angiotensin II in several tissues. Several reports have shown activation of the fetoplacental renin-angiotensin system (RAS) in IUGR. We investigated the effect of angiotensin II on placental system A transport and Na(+)-K(+)-ATPase activity in placental villi. Placental system A activity in single primary villous fragments was measured as the Na(+)-dependent uptake of alpha-(methylamino)isobutyric acid, and Na(+)/K(+) ATPase activity was measured as ouabain-sensitive uptake of (86)rubidium. Angiotensin II decreased system A activity in a concentration-dependent fashion (10-500 nmol/l). Angiotensin II type 1 receptor (AT1-R) antagonists losartan and AT1-R anti-peptide blocked the angiotensin II effect, but the angiotensin II type 2 receptor antagonist PD-123319 was without effect. System A activity was not altered by preincubation with AT1-R-independent vasoconstrictors, and antioxidants did not prevent the decrease in activity mediated by angiotensin II. Angiotensin II decreased Na(+)-K(+)-ATPase activity by an AT1-R dependent mechanism, and inhibition of Na(+)-K(+)-ATPase activity decreased system A activity in a dose-response fashion. These data suggest that angiotensin II, via AT1-R signaling, decreases system A activity by suppressing Na(+)-K(+)-ATPase in human placental villi, consistent with possible adverse effects of enhanced placental RAS on fetal growth.
Collapse
Affiliation(s)
- Eiji Shibata
- Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
McCord N, Ayuk P, McMahon M, Boyd RCA, Sargent I, Redman C. System y+ arginine transport and NO production in peripheral blood mononuclear cells in pregnancy and preeclampsia. Hypertension 2005; 47:109-15. [PMID: 16344361 DOI: 10.1161/01.hyp.0000197952.22711.c4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Systemic inflammation and oxidative stress are features of normal pregnancy and, in excess, contribute to the pathogenesis of preeclampsia. Inflammatory cell activation stimulates uptake of arginine (the precursor for nitric oxide) by transport system y+, expression of one of its genes (CAT-2) together with inducible nitric oxide synthase, leading to nitric oxide production. We investigated whether these changes occur in peripheral blood mononuclear cells in normal pregnancy and are exaggerated in preeclampsia. Samples from matched trios of nonpregnant, normal pregnant, and preeclamptic women were studied. Arginine transport was characterized, and the expression of inducible nitric oxide synthase and cell-specific nitric oxide production were measured. Arginine uptake by system y+ was significantly increased (P<0.001) in peripheral blood mononuclear cells in normal pregnancy but not in preeclampsia. CAT-2 mRNA was not detected in cells from nonpregnant women but was detected in 3 of 10 normal pregnant and 8 of 10 of preeclamptic women (P<0.001). Inducible nitric oxide synthase protein expression was significantly increased in normal pregnant women (P<0.05) but not preeclamptic women. No significant differences in cell-specific nitric oxide production were observed. These changes confirm the predictions for normal pregnancy but not for preeclampsia in which, despite increases in CAT-2 expression, arginine uptake is not additionally increased. This may create a relative deficiency of arginine in PBMCs favoring superoxide and peroxynitrite production and contribute to oxidative and nitrosative stress in preeclampsia.
Collapse
Affiliation(s)
- Nicola McCord
- Nuffield Department of Obstetrics and Gynaecology, Oxford University, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
22
|
Roos S, Powell TL, Jansson T. Human placental taurine transporter in uncomplicated and IUGR pregnancies: cellular localization, protein expression, and regulation. Am J Physiol Regul Integr Comp Physiol 2004; 287:R886-93. [PMID: 15166008 DOI: 10.1152/ajpregu.00232.2004] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Transplacental transfer is the fetus' primary source of taurine, an essential amino acid during fetal life. In intrauterine growth restriction (IUGR), placental transport capacity of taurine is reduced and fetal taurine levels are decreased. We characterized the protein expression of the taurine transporter (TAUT) in human placenta using immunocytochemistry and Western blotting, tested the hypothesis that placental protein expression of TAUT is reduced in IUGR, and investigated TAUT regulation by measuring the Na(+)-dependent taurine uptake in primary villous fragments after 1 h of incubation with different effectors. TAUT was primarily localized in the syncytiotrophoblast microvillous plasma membrane (MVM). TAUT was detected as a single 70-kDa band, and MVM TAUT expression was unaltered in IUGR. The PKC activator PMA and the nitric oxide (NO) donor 3-morpholinosydnonimine decreased TAUT activity (P < 0.05, n = 7-15). However, none of the tested hormones, e.g., leptin and growth hormone, altered TAUT activity significantly. PKC activity measured in MVM from control and IUGR placentas was not different. In conclusion, syncytiotrophoblast TAUT is strongly polarized to the maternal-facing plasma membrane. MVM TAUT expression is unaltered in IUGR, suggesting that the reduced MVM taurine transport in IUGR is due to changes in transporter activity. NO release downregulates placental TAUT activity, and it has previously been shown that IUGR is associated with increased fetoplacental NO levels. NO may therefore play an important role in downregulating MVM TAUT activity in IUGR.
Collapse
Affiliation(s)
- S Roos
- Perinatal Center, Dept. of Physiology and Pharmacology, Göteborg Univ., PO Box 432, S-405 30 Göteborg, Sweden.
| | | | | |
Collapse
|