101
|
Rios L, Campos EE, Menon R, Zago MP, Garg NJ. Epidemiology and pathogenesis of maternal-fetal transmission of Trypanosoma cruzi and a case for vaccine development against congenital Chagas disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165591. [PMID: 31678160 PMCID: PMC6954953 DOI: 10.1016/j.bbadis.2019.165591] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/12/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022]
Abstract
Trypanos o ma cruzi (T. cruzi or Tc) is the causative agent of Chagas disease (CD). It is common for patients to suffer from non-specific symptoms or be clinically asymptomatic with acute and chronic conditions acquired through various routes of transmission. The expecting women and their fetuses are vulnerable to congenital transmission of Tc. Pregnant women face formidable health challenges because the frontline antiparasitic drugs, benznidazole and nifurtimox, are contraindicated during pregnancy. However, it is worthwhile to highlight that newborns can be cured if they are diagnosed and given treatment in a timely manner. In this review, we discuss the pathogenesis of maternal-fetal transmission of Tc and provide a justification for the investment in the development of vaccines against congenital CD.
Collapse
Affiliation(s)
- Lizette Rios
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - E Emanuel Campos
- Instituto de Patología Experimental, Universidad Nacional de Salta - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta, Argentina
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - M Paola Zago
- Instituto de Patología Experimental, Universidad Nacional de Salta - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta, Argentina.
| | - Nisha J Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
102
|
Murthi P, Pinar AA, Dimitriadis E, Samuel CS. Inflammasomes-A Molecular Link for Altered Immunoregulation and Inflammation Mediated Vascular Dysfunction in Preeclampsia. Int J Mol Sci 2020; 21:ijms21041406. [PMID: 32093005 PMCID: PMC7073120 DOI: 10.3390/ijms21041406] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific multisystem disorder and is associated with maladaptation of the maternal cardiovascular system and abnormal placentation. One of the important characteristics in the pathophysiology of PE is a dysfunction of the placenta. Placental insufficiency is associated with poor trophoblast uterine invasion and impaired transformation of the uterine spiral arterioles to high capacity and low impedance vessels and/or abnormalities in the development of chorionic villi. Significant progress in identifying potential molecular targets in the pathophysiology of PE is underway. The human placenta is immunologically functional with the trophoblast able to generate specific and diverse innate immune-like responses through their expression of multimeric self-assembling protein complexes, termed inflammasomes. However, the type of response is highly dependent upon the stimuli, the receptor(s) expressed and activated, the downstream signaling pathways involved, and the timing of gestation. Recent findings highlight that inflammasomes can act as a molecular link for several components at the syncytiotrophoblast surface and also in maternal blood thereby directly influencing each other. Thus, the inflammasome molecular platform can promote adverse inflammatory effects when chronically activated. This review highlights current knowledge in placental inflammasome expression and activity in PE-affected pregnancies, and consequently, vascular dysfunction in PE that must be addressed as an interdependent interactive process.
Collapse
Affiliation(s)
- Padma Murthi
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Victoria 3168, Australia; (A.A.P.); (C.S.S.)
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria 3168, Australia;
- Correspondence: ; Tel.: +61-03-99059917
| | - Anita A. Pinar
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Victoria 3168, Australia; (A.A.P.); (C.S.S.)
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria 3168, Australia;
| | - Chrishan S. Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Victoria 3168, Australia; (A.A.P.); (C.S.S.)
| |
Collapse
|
103
|
Hu J, Zhang J, He G, Zhu S, Tang X, Su J, Li Q, Kong Y, Zhu B. First-trimester maternal serum alpha-fetoprotein is not a good predictor for adverse pregnancy outcomes: a retrospective study of 3325 cases. BMC Pregnancy Childbirth 2020; 20:104. [PMID: 32050927 PMCID: PMC7017534 DOI: 10.1186/s12884-020-2789-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/04/2020] [Indexed: 12/21/2022] Open
Abstract
Background It is well known that second-trimester maternal serum alpha-fetoprotein (MS-AFP) is a predictor for adverse pregnancy outcomes (APOs), such as preterm birth, stillbirth, preeclampsia and small for gestational age (SGA). However, it is unknown whether first-trimester MS-AFP is also predictive of APOs. Methods We retrospectively reviewed the data on the first-trimester MS-AFP levels and pregnancy outcomes of 3325 singleton pregnant women. The cutoff value of 2.5 multiple of the median (MoM) was used to evaluate the risks of APOs regarding MS-AFP. The receiver operating characteristic (ROC) curves were used to evaluate the predictive efficiencies of MS-AFP to these disorders. Results A total of 181 pregnancies resulted in preterm birth, 32 in stillbirth, 81 in preeclampsia, and 362 in SGA. Compared to women with MS-AFP < 2.5MoM, those with MS-AFP ≥ 2.5MoM had increased risks (odds ratio, 95% confidence interval) of preterm birth (2.53, 1.65~3.88), preeclampsia (3.05, 1.71~5.43) and SGA (1.90, 1.34~2.69), and had an earlier distribution of gestational weeks at delivery (P = 0.004) and a lower distribution of neonatal birth weights (P = 0.000), but the actual between-group differences were minuscule. The areas under ROC curves were 0.572 (P = 0.001), 0.579 (P = 0.015) and 0.565 (P = 0.000) for preterm birth, preeclampsia and SGA, respectively. Subdivisions for the disorders did not obviously improve the performances of MS-AFP. Conclusions Elevated first-trimester MS-AFP is associated with increased risk of preterm birth, preeclampsia and SGA. However, the predictive efficiencies were low and it is not a good predictor for these APOs.
Collapse
Affiliation(s)
- Jilin Hu
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Jinman Zhang
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Guilin He
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Shu Zhu
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Xinhua Tang
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Jie Su
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Qian Li
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Yamin Kong
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China
| | - Baosheng Zhu
- National Health Commission Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Clinical Medicine Research Center for Birth Defects and Rare Diseases, Department of Obstetrics and Gynecology, the First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, Yunnan Province, 650032, People's Republic of China.
| |
Collapse
|
104
|
Elkin ER, Bridges D, Harris SM, Loch-Caruso RK. Exposure to Trichloroethylene Metabolite S-(1,2-Dichlorovinyl)-L-cysteine Causes Compensatory Changes to Macronutrient Utilization and Energy Metabolism in Placental HTR-8/SVneo Cells. Chem Res Toxicol 2020; 33:1339-1355. [PMID: 31951115 PMCID: PMC7299793 DOI: 10.1021/acs.chemrestox.9b00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Trichloroethylene
(TCE) is a widespread environmental contaminant
following decades of use as an industrial solvent, improper disposal,
and remediation challenges. Consequently, TCE exposure continues to
constitute a risk to human health. Despite epidemiological evidence
associating exposure with adverse birth outcomes, the effects of TCE
and its metabolite S-(1, 2-dichlorovinyl)-L-cysteine
(DCVC) on the placenta remain undetermined. Flexible and efficient
macronutrient and energy metabolism pathway utilization is essential
for placental cell physiological adaptability. Because DCVC is known
to compromise cellular energy status and disrupt energy metabolism
in renal proximal tubular cells, this study investigated the effects
of DCVC on cellular energy status and energy metabolism pathways in
placental cells. Human extravillous trophoblast cells, HTR-8/SVneo,
were exposed to 5–20 μM DCVC for 6 or 12 h. After establishing
concentration and exposure duration thresholds for DCVC-induced cytotoxicity,
targeted metabolomics was used to evaluate overall energy status and
metabolite concentrations from energy metabolism pathways. The data
revealed glucose metabolism perturbations including a time-dependent
accumulation of glucose-6-phosphate+frutose-6-phosphate (G6P+F6P)
as well as independent shunting of glucose intermediates that diminished
with time, with modest energy status decline but in the absence of
significant changes in ATP concentrations. Furthermore, metabolic
profiling suggested that DCVC stimulated compensatory utilization
of glycerol, lipid, and amino acid metabolism to provide intermediate
substrates entering downstream in the glycolytic pathway or the tricarboxylic
acid cycle. Lastly, amino acid deprivation increased susceptibility
to DCVC-induced cytotoxicity. Taken together, these results suggest
that DCVC caused metabolic perturbations necessitating adaptations
in macronutrient and energy metabolism pathway utilization to maintain
adequate ATP levels.
Collapse
Affiliation(s)
- Elana R Elkin
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| | - Sean M Harris
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| | - Rita Karen Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| |
Collapse
|
105
|
Eddy AC, Howell JA, Chapman H, Taylor E, Mahdi F, George EM, Bidwell GL. Biopolymer-Delivered, Maternally Sequestered NF-κB (Nuclear Factor-κB) Inhibitory Peptide for Treatment of Preeclampsia. Hypertension 2019; 75:193-201. [PMID: 31786977 DOI: 10.1161/hypertensionaha.119.13368] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy that causes significant acute and long-term risk to the mother and the baby. The multifaceted maternal syndrome is driven by overproduction of circulating anti-angiogenic factors, widespread inflammation, and endothelial dysfunction. Nuclear factor-κB (NF-κB) is a transcription factor that plays a central role in the inflammatory response. Its activity is increased in the preeclamptic placenta, and it promotes the systemic endothelial dysfunction present in preeclampsia. There is an acute need for new therapeutics targeted to the causative pathways of preeclampsia. Our group has developed a drug delivery system based on the bioengineered protein ELP (elastin-like polypeptide) that is capable of stabilizing therapeutics in the maternal circulation and preventing their placental transfer. Here we used the ELP carrier system to deliver a peptide known to inhibit the NF-κB pathway. This polypeptide, containing a cell-penetrating peptide and an NF-κB inhibitory peptide derived from the p50 nuclear localization sequence (abbreviated SynB1-ELP-p50i), blocked NF-κB activation and prevented TNF-α (tumor necrosis factor alpha)-induced endothelin production in vitro. Fusion of the p50i peptide to the SynB1-ELP carrier slowed its plasma clearance and prevented its placental transfer in pregnant rats, resulting in increased deposition in the maternal kidney, liver, and placenta relative to the free peptide. When administered in a rat model of placental ischemia, SynB1-ELP-p50i partially ameliorated placental ischemia-induced hypertension and reduced placental TNF-α levels with no signs of toxicity. These data support the continued development of ELP-delivered NF-κB inhibitors as maternally sequestered anti-inflammatory agents for preeclampsia therapy.
Collapse
Affiliation(s)
- Adrian C Eddy
- From the Department of Physiology and Biophysics (A.C.E., H.C., E.T., E.M.G.), University of Mississippi Medical Center
| | - John Aaron Howell
- Graduate Program in Neuroscience (J.A.H.), University of Mississippi Medical Center.,Department of Neurology (J.A.H., F.M., G.L.B.), University of Mississippi Medical Center
| | - Heather Chapman
- From the Department of Physiology and Biophysics (A.C.E., H.C., E.T., E.M.G.), University of Mississippi Medical Center
| | - Erin Taylor
- From the Department of Physiology and Biophysics (A.C.E., H.C., E.T., E.M.G.), University of Mississippi Medical Center
| | - Fakhri Mahdi
- Department of Neurology (J.A.H., F.M., G.L.B.), University of Mississippi Medical Center
| | - Eric M George
- From the Department of Physiology and Biophysics (A.C.E., H.C., E.T., E.M.G.), University of Mississippi Medical Center.,Department of Cell and Molecular Biology (E.M.G., G.L.B.), University of Mississippi Medical Center
| | - Gene L Bidwell
- Department of Neurology (J.A.H., F.M., G.L.B.), University of Mississippi Medical Center.,Department of Cell and Molecular Biology (E.M.G., G.L.B.), University of Mississippi Medical Center.,Department of Pharmacology and Toxicology (G.L.B.), University of Mississippi Medical Center
| |
Collapse
|
106
|
Grandi SM, Filion KB, Yoon S, Ayele HT, Doyle CM, Hutcheon JA, Smith GN, Gore GC, Ray JG, Nerenberg K, Platt RW. Cardiovascular Disease-Related Morbidity and Mortality in Women With a History of Pregnancy Complications. Circulation 2019; 139:1069-1079. [PMID: 30779636 DOI: 10.1161/circulationaha.118.036748] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Women with a history of certain pregnancy complications are at higher risk for cardiovascular (CVD) disease. However, most clinical guidelines only recommend postpartum follow-up of those with a history of preeclampsia, gestational diabetes mellitus, or preterm birth. This systematic review was undertaken to determine if there is an association between a broader array of pregnancy complications and the future risk of CVD. METHODS We systematically searched PubMed, MEDLINE and EMBASE (via Ovid), CINAHL, and the Cochrane Library from inception to September 22, 2017, for observational studies of the association between the hypertensive disorders of pregnancy, placental abruption, preterm birth, gestational diabetes mellitus, low birth weight, small-for-gestational-age birth, stillbirth, and miscarriage and subsequent CVD. Likelihood ratio meta-analyses were performed to generate pooled odds ratios (OR) and 95% intrinsic confidence intervals (ICI). RESULTS Our systematic review included 84 studies (28 993 438 patients). Sample sizes varied from 250 to 2 000 000, with a median follow-up of 7.5 years postpartum. The risk of CVD was highest in women with gestational hypertension (OR 1.7; 95% ICI, 1.3-2.2), preeclampsia (OR 2.7; 95% ICI, 2.5-3.0), placental abruption (OR 1.8; 95% ICI, 1.4-2.3), preterm birth (OR 1.6; 95% ICI, 1.4-1.9), gestational diabetes mellitus (OR 1.7; 95% ICI, 1.1-2.5), and stillbirth (OR 1.5; 95% ICI, 1.1-2.1). A consistent trend was seen for low birth weight and small-for-gestational-age birth weight but not for miscarriage. CONCLUSIONS Women with a broader array of pregnancy complications, including placental abruption and stillbirth, are at increased risk of future CVD. The findings support the need for assessment and risk factor management beyond the postpartum period.
Collapse
Affiliation(s)
- Sonia M Grandi
- Department of Epidemiology, Biostatisticcs and Occupational Health, McGill University, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.)
| | - Kristian B Filion
- Department of Epidemiology, Biostatisticcs and Occupational Health, McGill University, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Department of Medicine, McGill University, Montreal, QC, Canada (K.F.)
| | - Sarah Yoon
- Department of Epidemiology, Biostatisticcs and Occupational Health, McGill University, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.)
| | - Henok T Ayele
- Department of Epidemiology, Biostatisticcs and Occupational Health, McGill University, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.)
| | - Carla M Doyle
- Department of Epidemiology, Biostatisticcs and Occupational Health, McGill University, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.)
| | - Jennifer A Hutcheon
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Canada (J.H.)
| | - Graeme N Smith
- Department of Obstetrics and Gynaecology, School of Medicine, Queen's University, Kingston, ON, Canada (G.S.)
| | - Genevieve C Gore
- Schulich Library of Science and Engineering, McGill University, Montreal, QC, Canada (G.G.)
| | - Joel G Ray
- Department of Medicine and Institute of Health Policy, Management and Evaluation, University of Toronto, ON, Canada (J.R.)
| | - Kara Nerenberg
- University of Calgary, Department of Medicine, Cumming School of Medicine, AB, Canada (K.N.)
| | - Robert W Platt
- Department of Epidemiology, Biostatisticcs and Occupational Health, McGill University, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada (S.G., K.F., S.Y., H.A., C.D., R.P.).,McGill University Health Center Research Institute and Department of Pediatrics, McGill University, Montreal, QC, Canada (R.P.)
| |
Collapse
|
107
|
Maric-Bilkan C, Abrahams VM, Arteaga SS, Bourjeily G, Conrad KP, Catov JM, Costantine MM, Cox B, Garovic V, George EM, Gernand AD, Jeyabalan A, Karumanchi SA, Laposky AD, Miodovnik M, Mitchell M, Pemberton VL, Reddy UM, Santillan MK, Tsigas E, Thornburg KLR, Ward K, Myatt L, Roberts JM. Research Recommendations From the National Institutes of Health Workshop on Predicting, Preventing, and Treating Preeclampsia. Hypertension 2019; 73:757-766. [PMID: 30686084 DOI: 10.1161/hypertensionaha.118.11644] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Christine Maric-Bilkan
- From the Division of Cardiovascular Sciences (C.M.-B., S.S.A., V.L.P.), National Institutes of Health, Bethesda, MD
| | - Vikki M Abrahams
- Department of Ob/Gyn and Reproductive Sciences, Yale University School of Medicine, New Haven, CT (V.M.A.)
| | - S Sonia Arteaga
- From the Division of Cardiovascular Sciences (C.M.-B., S.S.A., V.L.P.), National Institutes of Health, Bethesda, MD
| | - Ghada Bourjeily
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI (G.B.)
| | - Kirk P Conrad
- Department of Physiology and Functional Genomics and Ob/Gyn, University of Florida College of Medicine, Gainesville (K.P.C.)
| | - Janet M Catov
- Department of Ob/Gyn and Reproductive Sciences, Magee-Women's Research Institute and Clinical and Translational Sciences Research Institute, University of Pittsburgh, PA (J.M.C., A.J., J.M.R.)
| | - Maged M Costantine
- Department of Ob/Gyn, University of Texas Medical Branch, Galveston (M.M.C.)
| | - Brian Cox
- Department of Physiology and Ob/Gyn, University of Toronto, ON, Canada (B.C.)
| | - Vesna Garovic
- Department of Internal Medicine and Division of Nephrology and Hypertension, Department of Ob/Gyn, Mayo Clinic, Rochester, MN (V.G.)
| | - Eric M George
- Department of Physiology and Biophysics and Cell and Molecular Biology, University of Mississippi Medical Center, Jackson (E.M.G.)
| | - Alison D Gernand
- Department of Nutritional Sciences, The Pennsylvania State University (A.D.G.)
| | - Arun Jeyabalan
- Department of Ob/Gyn and Reproductive Sciences, Magee-Women's Research Institute and Clinical and Translational Sciences Research Institute, University of Pittsburgh, PA (J.M.C., A.J., J.M.R.)
| | - S Ananth Karumanchi
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA (S.A.K.)
| | - Aaron D Laposky
- Division of Lung Diseases, National Center on Sleep Disorders Research (A.D.L.), National Institutes of Health, Bethesda, MD
| | - Menachem Miodovnik
- National Heart, Lung, and Blood Institute and Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (M. Miodovnik, U.M.R.), National Institutes of Health, Bethesda, MD
| | - Megan Mitchell
- Division of Extramural Research Activities (M. Mitchell), National Institutes of Health, Bethesda, MD
| | - Victoria L Pemberton
- From the Division of Cardiovascular Sciences (C.M.-B., S.S.A., V.L.P.), National Institutes of Health, Bethesda, MD
| | - Uma M Reddy
- National Heart, Lung, and Blood Institute and Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (M. Miodovnik, U.M.R.), National Institutes of Health, Bethesda, MD
| | - Mark K Santillan
- Department of Ob/Gyn, University of Iowa Carver College of Medicine (M.K.S.)
| | | | - Kent L R Thornburg
- Bob & Charlee Moore Institute for Nutrition & Wellness, Oregon Health and Science University, Portland (K.L.R.T.)
| | | | - Leslie Myatt
- Bob & Charlee Moore Institute for Nutrition & Wellness and Department of Ob/Gyn, Oregon Health and Science University, Portland (L.M.)
| | - James M Roberts
- Department of Ob/Gyn and Reproductive Sciences, Magee-Women's Research Institute and Clinical and Translational Sciences Research Institute, University of Pittsburgh, PA (J.M.C., A.J., J.M.R.)
| |
Collapse
|
108
|
Montagnana M, Benati M, Tagetti A, Raffaelli R, Danese E, Zatti N, Marcon D, Branz L, Micheli C, Franchi M, Fava C, Lippi G. Evaluation of circ_100219 and miR-135b in serum and exosomes of healthy pregnant women. J Matern Fetal Neonatal Med 2019; 34:3645-3650. [PMID: 31722587 DOI: 10.1080/14767058.2019.1689556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Aim: Circular RNAs (circRNAs) are recently discovered and highly stable noncoding RNAs acting as gene regulators. These circRNAs can function as miRNA sponges, thereby upregulating or downregulating miRNA target gene expression. MiR-135b is expressed in placenta tissue and can be found in maternal circulation, thus playing a functional role in pregnancy. This miR is a target of circ_100219. This preliminary study was aimed to evaluate circ_100219 and miR-135b expression in pregnant and nonpregnant women, and explore the relationship between circ_100219 and miR-135b in serum and exosomes.Methods: Total RNA was isolated from serum and exosomes of 30 healthy pregnant women (32.9 ± 5.1 years) between 23-27 gestational weeks and 30 healthy nonpregnant women (31.3 ± 5.4 years). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to quantify circ_100219 and miR-135b expression. GAPDH and U6 snRNA were chosen as reference for normalizing expression levels. The differences between pregnant and nonpregnant women were assessed with Mann-Whitney test and correlation with Spearman's test.Results: The circ_100219 expression levels were significantly lower both in serum and exosomes of second trimester pregnant women compared to the control group (p < .0001), whilst Mir-135b expression levels were significantly higher in pregnant than in the control group (p < .0001). A significant negative correlation was observed between circ_100219 and miR-135b expression levels in both serum and exosomes (r = -0.34 and p = .009; r = -0.31 and p = .01, respectively). The circ_100219:miR-135b ratio was significantly increased in nonpregnant women compared to the pregnant group, in both serum and exosomes (49.0 versus 1.1, p < .0001 and 2042.4 versus 28.5, p < .0001, respectively).Conclusions: Our results confirm a role for circ_100219 and miR-135b in physiological pregnancy. Further studies are needed to investigate the circ_100219:miR-135b ratio in pregnancy complications.
Collapse
Affiliation(s)
- Martina Montagnana
- Department Neurosciences, Biomedicine and Movement Sciences, Clinical Biochemistry Section, University of Verona, Verona, Italy
| | - Marco Benati
- Department Neurosciences, Biomedicine and Movement Sciences, Clinical Biochemistry Section, University of Verona, Verona, Italy
| | - Angela Tagetti
- Department of Medicine, Medicine C Section, University of Verona, Verona, Italy
| | - Ricciarda Raffaelli
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, Obstetrics and Gynecology Section, University of Verona, Verona, Italy
| | - Elisa Danese
- Department Neurosciences, Biomedicine and Movement Sciences, Clinical Biochemistry Section, University of Verona, Verona, Italy
| | - Nicoletta Zatti
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, Obstetrics and Gynecology Section, University of Verona, Verona, Italy
| | - Denise Marcon
- Department of Medicine, Medicine C Section, University of Verona, Verona, Italy
| | - Lorella Branz
- Department of Medicine, Medicine C Section, University of Verona, Verona, Italy
| | - Chiara Micheli
- Department of Medicine, Medicine C Section, University of Verona, Verona, Italy
| | - Massimo Franchi
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, Obstetrics and Gynecology Section, University of Verona, Verona, Italy
| | - Cristiano Fava
- Department of Medicine, Medicine C Section, University of Verona, Verona, Italy
| | - Giuseppe Lippi
- Department Neurosciences, Biomedicine and Movement Sciences, Clinical Biochemistry Section, University of Verona, Verona, Italy
| |
Collapse
|
109
|
Unique transcriptomic landscapes identified in idiopathic spontaneous and infection related preterm births compared to normal term births. PLoS One 2019; 14:e0225062. [PMID: 31703110 PMCID: PMC6839872 DOI: 10.1371/journal.pone.0225062] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/27/2019] [Indexed: 02/07/2023] Open
Abstract
Preterm birth (PTB) is leading contributor to infant death in the United States and globally, yet the underlying mechanistic causes are not well understood. Histopathological studies of preterm birth suggest advanced villous maturity may have a role in idiopathic spontaneous preterm birth (isPTB). To better understand pathological and molecular basis of isPTB, we compared placental villous transcriptomes from carefully phenotyped cohorts of PTB due to infection or isPTB between 28–36 weeks gestation and healthy term placentas. Transcriptomic analyses revealed a unique expression signature for isPTB distinct from the age-matched controls that were delivered prematurely due to infection. This signature included the upregulation of three IGF binding proteins (IGFBP1, IGFBP2, and IGFBP6), supporting a role for aberrant IGF signaling in isPTB. However, within the isPTB expression signature, we detected secondary signature of inflammatory markers including TNC, C3, CFH, and C1R, which have been associated with placental maturity. In contrast, the expression signature of the gestational age-matched infected samples included upregulation of proliferative genes along with cell cycling and mitosis pathways. Together, these data suggest an isPTB molecular signature of placental hypermaturity, likely contributing to the premature activation of inflammatory pathways associated with birth and providing a molecular basis for idiopathic spontaneous birth.
Collapse
|
110
|
Gillet V, Ouellet A, Stepanov Y, Rodosthenous RS, Croft EK, Brennan K, Abdelouahab N, Baccarelli A, Takser L. miRNA Profiles in Extracellular Vesicles From Serum Early in Pregnancies Complicated by Gestational Diabetes Mellitus. J Clin Endocrinol Metab 2019; 104:5157-5169. [PMID: 31058973 PMCID: PMC6760296 DOI: 10.1210/jc.2018-02693] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
CONTEXT Underlying mechanisms leading to gestational diabetes mellitus (GDM) are still under investigation, and it is unclear whether the placenta plays a role in triggering glucose intolerance or if its functions are modified in response to the hyperglycemia. Circulating miRNAs are involved in placental development and function and are encapsulated in extracellular vesicles (EVs). OBJECTIVE To compare differential expression of miRNAs in circulating EVs in pregnancies complicated by GDM vs controls. METHODS This was a case-control study nested in a prospective pregnancy cohort including 23 women with GDM and 46 matched controls. The presence of serum EVs in early pregnancy was validated by transmission electron microscopy. Placental dimensions were assessed at 11 to 13 weeks of gestation. Differential expression of 17 miRNAs encapsulated in EVs (miR‒122-5p, miR‒132-3p, miR-1323, miR‒182-3p, miR‒210-3p, miR‒29a-3p, miR‒29b-3p, miR‒342-3p, miR‒517-5p, miR‒517a-3p, miR‒518b, miR-520h, miR‒525-5p, miR‒136-5p, miR‒342-3p, miR‒376c-5p, and miR‒494-3p) was assessed using quantitative reverse transcription PCR. RESULTS EVs were present in the early phase of placentation (6 to 15 weeks of gestation) in both cases and controls. No differences were observed for placental dimensions and estimated placental volume between GDM and control groups. Ten miRNAs (miR‒122-5p; miR‒132-3p; miR‒1323; miR‒136-5p; miR‒182-3p; miR‒210-3p; miR‒29a-3p; miR‒29b-3p; miR‒342-3p, and miR-520h) showed significantly higher levels in GDM cases than in controls (P ≤ 0.05). Bioinformatics analysis showed that these miRNAs are involved in trophoblast proliferation/differentiation as well as in insulin secretion/regulation and glucose transport in pregnant women. CONCLUSION The miRNA content of blood EVs may be a promising avenue for studying the early effect of impaired glucose metabolism on placental development.
Collapse
Affiliation(s)
- Virginie Gillet
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Correspondence and Reprint Requests: Virginie Gillet, MSc, or Larissa Takser, MD, PhD, Faculté de Médecine et des Sciences de la Santé 3001, 12ème Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada. E-mail: or
| | - Annie Ouellet
- Département d’Obstétrique et de Gynécologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Yulia Stepanov
- Département d’Obstétrique et de Gynécologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | - Erika Kathe Croft
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kasey Brennan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Nadia Abdelouahab
- Département d’Obstétrique et de Gynécologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Andrea Baccarelli
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Larissa Takser
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Département de Psychiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Correspondence and Reprint Requests: Virginie Gillet, MSc, or Larissa Takser, MD, PhD, Faculté de Médecine et des Sciences de la Santé 3001, 12ème Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada. E-mail: or
| |
Collapse
|
111
|
Kelley AS, Smith YR, Padmanabhan V. A Narrative Review of Placental Contribution to Adverse Pregnancy Outcomes in Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2019; 104:5299-5315. [PMID: 31393571 PMCID: PMC6767873 DOI: 10.1210/jc.2019-00383] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is the most common endocrinopathy of reproductive-aged women. In pregnancy, women with PCOS experience increased risk of miscarriage, gestational diabetes, preeclampsia, and extremes of fetal birth weight, and their offspring are predisposed to reproductive and cardiometabolic dysfunction in adulthood. Pregnancy complications, adverse fetal outcomes, and developmental programming of long-term health risks are known to have placental origins. These findings highlight the plausibility of placental compromise in pregnancies of women with PCOS. EVIDENCE SYNTHESIS A comprehensive PubMed search was performed using terms "polycystic ovary syndrome," "placenta," "developmental programming," "hyperandrogenism," "androgen excess," "insulin resistance," "hyperinsulinemia," "pregnancy," and "pregnancy complications" in both human and animal experimental models. CONCLUSIONS There is limited human placental research specific to pregnancy of women with PCOS. Gestational androgen excess and insulin resistance are two clinical hallmarks of PCOS that may contribute to placental dysfunction and underlie the higher rates of maternal-fetal complications observed in pregnancies of women with PCOS. Additional research is needed to prevent adverse maternal and developmental outcomes in women with PCOS and their offspring.
Collapse
Affiliation(s)
- Angela S Kelley
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Yolanda R Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
- Correspondence and Reprint Requests: Vasantha Padmanabhan, PhD, Department of Pediatrics, University of Michigan, 7510 MSRB 1, 1500 West Medical Center Drive, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
112
|
Liu Y, Gao B, Zeng X, Yang J, Zhang L, Xu G, Jia R, Xu Z. Association between thyroid-stimulating hormone and maternal hemodynamics in hypertensive disorders of pregnancy: an observational study. BMC Pregnancy Childbirth 2019; 19:396. [PMID: 31675921 PMCID: PMC6824088 DOI: 10.1186/s12884-019-2556-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/14/2019] [Indexed: 11/10/2022] Open
Abstract
Background Hypertensive disorders of pregnancy (HDP) are characterized by hemodynamic disturbances. Altered thyroid function is a risk factor for poor outcomes of pregnancy. However, the associations between thyroid function biomarkers and maternal hemodynamics during pregnancy in HDP remain unclear. Methods From January 2016 to January 2018, pregnant women diagnosed with HDP admitted to the Nanjing Maternity and Child Health Care Hospital were prospectively enrolled in the third trimester. Normally distributed variables were expressed as mean ± standard deviation and skewed variables were expressed as median (25th percentile, 75th percentile). Correlations between thyroid-stimulating hormone (TSH) or free thyroxine (FT4) and maternal hemodynamic parameters were assessed by Pearson’s correlation coefficient and 95% confidence interval (95%CI). Bonferroni’s correction for multiple correlations was performed. Logistic regression models with odd ratio (OR) and 95%CI were applied to confirm the associations. Results A total of 163 third-trimester pregnant women with HDP with a mean gestational age of 35.62 ± 2.83 weeks were recruited. The infant birth weight of patients with elevated TSH levels was lower than that of patients with normal TSH levels (2635 ± 867 g vs. 3037 ± 673 g, p = 0.002). Reduced cardiac output (CO) was defined as CO < 3.5 L/min. The infant birth weight of patients with reduced CO was lower than that of patients with normal CO (2250 ± 510 g vs. 2890 ± 774 g, p = 0.002). TSH levels were significantly and negatively correlated with CO (r = − 0.260, 95%CI: − 0.392- -0.103, p < 0.001). FT4 levels were not significantly correlated with any of the maternal hemodynamic parameters (all p > 0.05). TSH level (OR = 1.371, 95%CI: 1.086–1.733, p = 0.008) was confirmed associated with reduced CO in the logistic regression analysis. Conclusions Elevated TSH levels are associated with reduced CO in HDP during the third trimester.
Collapse
Affiliation(s)
- Yu Liu
- Department of Obstetrics and Gynecology, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, 210004, China
| | - Bo Gao
- Department of Clinical Nutrition, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, 321 Zhongshan Road, Gulou District, Nanjing, China
| | - Xin Zeng
- Department of Obstetrics and Gynecology, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, 210004, China.,Nanjing Maternal and Child Health Institute, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, China
| | - Jing Yang
- Department of Science and Education, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, China
| | - Lei Zhang
- Department of Obstetrics and Gynecology, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, 210004, China
| | - Ganwei Xu
- Department of Medical Records, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, China
| | - Ruizhe Jia
- Department of Obstetrics and Gynecology, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, 210004, China.
| | - Zhengfeng Xu
- Department of Obstetrics and Gynecology, The affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University; Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, 210004, China. .,Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing, China.
| |
Collapse
|
113
|
Zheng L, Huang J, Su Y, Wang F, Kong H, Xin H. Vitexin ameliorates preeclampsia phenotypes by inhibiting TFPI-2 and HIF-1α/VEGF in a l-NAME induced rat model. Drug Dev Res 2019; 80:1120-1127. [PMID: 31486114 DOI: 10.1002/ddr.21596] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022]
Abstract
Preeclampsia (PE) is a leading cause of maternal and perinatal morbidity and mortality with few safe, effective, and minimally invasive therapeutics. Inflammation, oxidative stress, and angiogenic imbalance have been reported to contribute to PE pathogenesis. Vitexin (VI) possesses various pharmacological activities including the potent regulation of the above biological processes in different conditions. This study aims to investigate whether VI has therapeutic potential to PE and the underlying mechanisms. Sprague-Dawley pregnant rats pretreated with or without VI were fed with l-NAME-containing water to induce experimental PE. Results showed that VI decreased high systolic blood pressure and urinary protein in PE rats time- and dose-dependently. Meanwhile, VI of higher dosage (45, 60 mg/kg) corrected abnormal pregnancy outcomes, including low pup weight and low pups/placenta ratio. In addition, VI of high dosage (60 mg/kg) decreased sFlt-1, increased PlGF and alleviated oxidative stress both in blood and placental samples compared with nontreated PE group. Furthermore, VI alleviated placental TFPI-2, HIF 1α, and VEGF in PE rats. In short, the present study suggests that the inhibition of placental TFPI-2 and HIF-1α/VEGF might be one of the potential mechanisms underlying the protective effects of VI to experimental PE induced by l-NAME.
Collapse
Affiliation(s)
- Lili Zheng
- Department of Obstetrics, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Huang
- Department of Obstetrics, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuan Su
- Department of Obstetrics, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fang Wang
- Department of Obstetrics, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongfang Kong
- Department of Obstetrics, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hong Xin
- Department of Obstetrics, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
114
|
Elkin ER, Bridges D, Loch-Caruso R. The trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine induces progressive mitochondrial dysfunction in HTR-8/SVneo trophoblasts. Toxicology 2019; 427:152283. [PMID: 31476333 DOI: 10.1016/j.tox.2019.152283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/24/2019] [Accepted: 08/28/2019] [Indexed: 02/08/2023]
Abstract
Trichloroethylene is an industrial solvent and common environmental pollutant. Despite efforts to ban trichloroethylene, its availability and usage persist globally, constituting a hazard to human health. Recent studies reported associations between maternal trichloroethylene exposure and increased risk for low birth weight. Despite these associations, the toxicological mechanism underlying trichloroethylene adverse effects on pregnancy remains largely unknown. The trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine (DCVC) induces mitochondrial-mediated apoptosis in a trophoblast cell line. To gain further understanding of mitochondrial-mediated DCVC placental toxicity, this study investigated the effects of DCVC exposure on mitochondrial function using non-cytolethal concentrations in placental cells. Human trophoblasts, HTR-8/SVneo, were exposed in vitro to a maximum of 20 μM DCVC for up to 12 h. Cell-based oxygen consumption and extracellular acidification assays were used to evaluate key aspects of mitochondrial function. Following 6 h of exposure to 20 μM DCVC, elevated oxygen consumption, mitochondrial proton leak and sustained energy coupling deficiency were observed. Similarly, 12 h of exposure to 20 μM DCVC decreased mitochondrial-dependent basal, ATP-linked and maximum oxygen consumption rates. Using the fluorochrome TMRE, dissipation of mitochondrial membrane potential was detected after a 12-h exposure to 20 μM DCVC, and (±)-α-tocopherol, a known suppressor of lipid peroxidation, attenuated DCVC-stimulated mitochondrial membrane depolarization but failed to rescue oxygen consumption perturbations. Together, these results suggest that DCVC caused progressive mitochondrial dysfunction, resulting in lipid peroxidation-associated mitochondrial membrane depolarization. Our findings contribute to the biological plausibility of DCVC-induced placental impairment and provide new insights into the role of the mitochondria in DCVC-induced toxicity.
Collapse
Affiliation(s)
- Elana R Elkin
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA.
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA.
| | - Rita Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA.
| |
Collapse
|
115
|
Jhee JH, Lee S, Park Y, Lee SE, Kim YA, Kang SW, Kwon JY, Park JT. Prediction model development of late-onset preeclampsia using machine learning-based methods. PLoS One 2019; 14:e0221202. [PMID: 31442238 PMCID: PMC6707607 DOI: 10.1371/journal.pone.0221202] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 08/01/2019] [Indexed: 11/18/2022] Open
Abstract
Preeclampsia is one of the leading causes of maternal and fetal morbidity and mortality. Due to the lack of effective preventive measures, its prediction is essential to its prompt management. This study aimed to develop models using machine learning to predict late-onset preeclampsia using hospital electronic medical record data. The performance of the machine learning based models and models using conventional statistical methods were also compared. A total of 11,006 pregnant women who received antenatal care at Yonsei University Hospital were included. Maternal data were retrieved from electronic medical records during the early second trimester to 34 weeks. The prediction outcome was late-onset preeclampsia occurrence after 34 weeks’ gestation. Pattern recognition and cluster analysis were used to select the parameters included in the prediction models. Logistic regression, decision tree model, naïve Bayes classification, support vector machine, random forest algorithm, and stochastic gradient boosting method were used to construct the prediction models. C-statistics was used to assess the performance of each model. The overall preeclampsia development rate was 4.7% (474 patients). Systolic blood pressure, serum blood urea nitrogen and creatinine levels, platelet counts, serum potassium level, white blood cell count, serum calcium level, and urinary protein were the most influential variables included in the prediction models. C-statistics for the decision tree model, naïve Bayes classification, support vector machine, random forest algorithm, stochastic gradient boosting method, and logistic regression models were 0.857, 0.776, 0.573, 0.894, 0.924, and 0.806, respectively. The stochastic gradient boosting model had the best prediction performance with an accuracy and false positive rate of 0.973 and 0.009, respectively. The combined use of maternal factors and common antenatal laboratory data of the early second trimester through early third trimester could effectively predict late-onset preeclampsia using machine learning algorithms. Future prospective studies are needed to verify the clinical applicability algorithms.
Collapse
Affiliation(s)
- Jong Hyun Jhee
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - SungHee Lee
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
- Biostatics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Yejin Park
- Division of Maternal-Fetal Medicine, Institute of Women’s Medical Life Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Eun Lee
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
- Biostatics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ah Kim
- Department of Medical Informatics, Yonsei University Health System, Seoul, Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Ja-Young Kwon
- Division of Maternal-Fetal Medicine, Institute of Women’s Medical Life Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JTP); (JYK)
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
- * E-mail: (JTP); (JYK)
| |
Collapse
|
116
|
Dominguez JE, Krystal AD, Habib AS. Obstructive Sleep Apnea in Pregnant Women: A Review of Pregnancy Outcomes and an Approach to Management. Anesth Analg 2019; 127:1167-1177. [PMID: 29649034 DOI: 10.1213/ane.0000000000003335] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Among obese pregnant women, 15%-20% have obstructive sleep apnea (OSA) and this prevalence increases along with body mass index and in the presence of other comorbidities. Prepregnancy obesity and pregnancy-related weight gain are certainly risk factors for sleep-disordered breathing in pregnancy, but certain physiologic changes of pregnancy may also increase a woman's risk of developing or worsening OSA. While it has been shown that untreated OSA in postmenopausal women is associated with a range of cardiovascular, pulmonary, and metabolic comorbidities, a body of literature is emerging that suggests OSA may also have serious implications for the health of mothers and fetuses during and after pregnancy. In this review, we discuss the following: pregnancy as a vulnerable period for the development or worsening of OSA; the associations between OSA and maternal and fetal outcomes; the current screening modalities for OSA in pregnancy; and current recommendations regarding peripartum management of OSA.
Collapse
Affiliation(s)
| | - Andrew D Krystal
- Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina.,Department of Psychiatry, University of California, San Francisco, San Francisco, California
| | | |
Collapse
|
117
|
Pregnancy Hypertension and a Commonly Inherited IGF1R Variant (rs2016347) Reduce Breast Cancer Risk by Enhancing Mammary Gland Involution. JOURNAL OF ONCOLOGY 2019; 2019:6018432. [PMID: 31687025 PMCID: PMC6800903 DOI: 10.1155/2019/6018432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/28/2019] [Accepted: 07/28/2019] [Indexed: 12/21/2022]
Abstract
Background Terminal duct lobular units (TDLUs) are the anatomic sites of breast cancer initiation, and breast tissue involution resulting in lower TDLU counts has been associated with decreased breast cancer risk. The insulin-like growth factor (IGF) pathway plays a role in breast involution, and systemic changes in this developmental pathway occur with hypertensive disorders of pregnancy (HDP), which have also been associated with lower breast cancer risk, especially in women carrying a functional variant of IGF1R SNP rs2016347. We proposed that this breast cancer protective effect might be explained by increased breast tissue involution. Materials and Methods We conducted a retrospective cohort study utilizing the Komen Tissue Bank, which collects breast tissue core biopsies from women without a history of breast cancer. Eighty white non-Hispanic women with a history of HDP were selected along with 120 nonexposed participants, and after genotyping for rs2016347, TDLU parameters were histologically measured blinded to participant characteristics from fixed biopsy sections. Results Stratified models by HDP status demonstrated that among HDP+ participants, those carrying two T alleles of rs2016347 had a decrease in TDLU counts of 53.2% when compared to those with no T alleles (p=0.049). Trend analysis demonstrated a multiplicative decrease in counts of 31.6% per T allele (p=0.050). Although no statistically significant interaction was seen between HDP status and T alleles, interaction terms showed increasingly negative values reaching a p value of 0.124 for HDP × 2T alleles. Conclusions The observed statistically significant decrease in TDLU counts signifies increased breast epithelial involution in women with prior HDP who inherited the TT genotype of IGF1R SNP rs2016347. The increasing degree of breast involution with greater rs2016347 T allele copy number is consistent with the known progressive reduction in IGF1R expression in breast and other normal tissues.
Collapse
|
118
|
Hu J, Zhang J, Zhu B. Protective effect of metformin on a rat model of lipopolysaccharide-induced preeclampsia. Fundam Clin Pharmacol 2019; 33:649-658. [PMID: 31334867 DOI: 10.1111/fcp.12501] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/05/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
Abstract
Recent in vitro and clinical studies have found that metformin (MET) may play a preventive or therapeutic role in preeclampsia (PE) and may be a candidate drug for the prevention and/or treatment of PE. In this study, we used lipopolysaccharide (LPS) to induce a PE-like rat model and investigated the intervention effect of MET from the perspectives of clinical manifestations, placental morphology, serum marker for placental injury, systemic inflammatory response and oxidative/nitrative stress, and placental nuclear factor-κB (NF-κB) signaling. The results showed that MET improved LPS-induced hypertension, proteinuria, fetal growth restriction (FGR) and stillbirth, alleviated placental injury and decreased maternal serum marker alpha-fetoprotein (MS-AFP) level; MET suppressed LPS-induced TNF-α and IL-6 productions, reduced oxidative/nitrative stress as evidenced by increased superoxide dismutase (SOD) activity, decreased inducible nitric oxide synthase (iNOS) activity, and decreased levels of malondialdehyde (MDA) and nitric oxide (NO); MET inhibited LPS-induced NF-κB activation in placentas. Based on these findings, it can be concluded that MET is beneficial to the PE-like rat model by protecting placentas from injury, suppressing systemic inflammatory response and oxidative/nitrative stress, and inhibiting placental NF-κB signaling pathway. MET is a promising drug for prevention and/or treatment of PE.
Collapse
Affiliation(s)
- Jilin Hu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.,Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jinman Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.,National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, 650032, China
| | - Baosheng Zhu
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.,National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, 650032, China
| |
Collapse
|
119
|
Tanacan A, Beksac MS, Orgul G, Duru S, Sener B, Karaagaoglu E. Impact of extractable nuclear antigen, anti-double stranded DNA, antiphospholipid antibody, and anticardiolipin antibody positivity on obstetrical complications and pregnancy outcomes. Hum Antibodies 2019; 27:135-141. [PMID: 30856108 DOI: 10.3233/hab-180359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Extractable nuclear antigen (ENA) and anti-double stranded DNA (anti-dsDNA) positivity and related diseases like systemic lupus erythematosus, Sjögren syndrome, and other autoimmune diseases are known to be associated with obstetrical complications and poor perinatal outcomes. OBJECTIVE To demonstrate the importance of ENA, anti-dsDNA, antiphospholipid (APL), and anticardiolipin (ACL) antibody positivity on pregnancy outcomes. METHODS Ninety one pregnant women with known ENA, anti-dsDNA, APL IgG and IgM, and ACL IgG and IgM antibody positivity were retrospectively compared with 91 randomly selected pregnant woman in terms of obstetrical complications and pregnancy outcomes. Beksac Obstetrics Index-pregnancy (BOIp), calculated as (number of children + (π/10))/gravidity in the current pregnancy, was used to compare the risk level between groups. RESULTS Significant differences were found in the median maternal age, gravidity, number of previous miscarriages, and BOIp between the groups (p= 0.04, p< 0.001, p< 0.001, and p< 0.001, respectively). Significant differences were also found between the study and control groups in the median gestational age at birth, birth weight, and APGAR1 score (p< 0.001 for all). Similarly, significant differences were found between groups in the rates of intra-uterine growth restriction, oligohydramnios, and gestational hypertension (p< 0.001, p= 0.05, and p= 0.05, respectively). There were 3 (3.3%) stillbirths in the study group and none in the control group (p= 0.123). CONCLUSION We evaluated the impact of anti-dsDNA, ENA, APL, and ACL antibody positivity, which may cause immunologic inflammation at placenta and thereby affect pregnancy outcomes.
Collapse
Affiliation(s)
- Atakan Tanacan
- Division of Perinatology, Department of Obstetrics and Gynecology, Hacettepe University, Ankara, Turkey
| | - Mehmet Sinan Beksac
- Division of Perinatology, Department of Obstetrics and Gynecology, Hacettepe University, Ankara, Turkey
| | - Gokcen Orgul
- Division of Perinatology, Department of Obstetrics and Gynecology, Hacettepe University, Ankara, Turkey
| | - Sinem Duru
- Division of Perinatology, Department of Obstetrics and Gynecology, Hacettepe University, Ankara, Turkey
| | - Burcin Sener
- Department of Clinical Biochemistry, Hacettepe University, Ankara, Turkey
| | | |
Collapse
|
120
|
Hu J, Zhang J, Chan Y, Zhu B. A rat model of placental inflammation explains the unexplained elevated maternal serum alpha-fetoprotein associated with adverse pregnancy outcomes. J Obstet Gynaecol Res 2019; 45:1980-1988. [PMID: 31381236 DOI: 10.1111/jog.14085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/13/2019] [Indexed: 01/09/2023]
Abstract
AIM It has been reported in numerous studies that elevated maternal serum alpha-fetoprotein (MS-AFP) is associated with adverse pregnancy outcomes (APO), such as pre-eclampsia, stillbirth, preterm birth and fetal growth restriction. However, the mechanism linking elevated MS-AFP and APO is obscure. In this study, we tried to explore the mechanism by using pregnant rats. METHODS Lipopolysaccharide (LPS) was used to induce placental inflammation in pregnant rats. Maternal serum and placental inflammatory cytokines and placental morphology were used to assess the level of placental inflammation. The incidences of APO and the levels of MS-AFP were evaluated. The expressions of alpha-fetoprotein (AFP) in the related organs and fetal serum AFP levels were detected. RESULTS Compared to saline-treated pregnant rats, LPS led to elevated maternal serum and placental inflammatory cytokines and a higher rate of placental inflammation. Lipopolysaccharide resulted in the features of APO and at the same time elevated MS-AFP. Maternal serum alpha-fetoprotein levels were significantly correlated to the evaluation parameters of APO. Lipopolysaccharide did not increase the expressions of AFP in fetal liver, maternal liver and placenta, but reduced the fetal serum AFP levels. CONCLUSION The phenomenon that elevated MS-AFP is associated with APO, which has been reported in human pregnancies, is observed in our rat model. Placental inflammation can be the potential cause linking the two manifestations together. Although the source of elevated MS-AFP is not identified, fetal blood circulation is suspected. Our study may provide an animal model for the future studies on this subject.
Collapse
Affiliation(s)
- Jilin Hu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinman Zhang
- Medical School, Kunming University of Science and Technology, Kunming, China.,National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, China
| | - Ying Chan
- National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, China
| | - Baosheng Zhu
- Medical School, Kunming University of Science and Technology, Kunming, China.,National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
121
|
Exposure of trophoblast cells to fine particulate matter air pollution leads to growth inhibition, inflammation and ER stress. PLoS One 2019; 14:e0218799. [PMID: 31318865 PMCID: PMC6638881 DOI: 10.1371/journal.pone.0218799] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 06/10/2019] [Indexed: 01/07/2023] Open
Abstract
Ambient air pollution is considered a major environmental health threat to pregnant women. Our previous work has shown an association between exposure to airborne particulate matter (PM) and an increased risk of developing pre-eclamspia. It is now recognized that many pregnancy complications are due to underlying placental dysfunction, and this tissue plays a pivotal role in pre-eclamspia. Recent studies have shown that PM can enter the circulation and reach the human placenta but the effects of PM on human placental function are still largely unknown. In this work we investigated the effects of airborne PM on trophoblast cells. Human, first trimester trophoblast cells (HTR-8/SV) were exposed to urban pollution particles (Malmö PM2.5; Prague PM10) for up to seven days in vitro and were analysed for uptake, levels of hCGβ and IL-6 secretion and proteomic analysis. HTR-8/SVneo cells rapidly endocytose PM within 30 min of exposure and particles accumulate in the cell in perinuclear vesicles. High doses of Prague and Malmö PM (500-5000 ng/ml) significantly decreased hCGβ secretion and increased IL-6 secretion after 48 h exposure. Exposure to PM (50 ng/ml) for 48h or seven days led to reduced cellular growth and altered protein expression. The differentially expressed proteins are involved in networks that regulate cellular processes such as inflammation, endoplasmic reticulum stress, cellular survival and molecular transport pathways. Our studies suggest that trophoblast cells exposed to low levels of urban PM respond with reduced growth, oxidative stress, inflammation and endoplasmic reticulum stress after taking up the particles by endocytosis. Many of the dysfunctional cellular processes ascribed to the differentially expressed proteins in this study, are similar to those described in PE, suggesting that low levels of urban PM may disrupt cellular processes in trophoblast cells. Many of the differentially expressed proteins identified in this study are involved in inflammation and may be potential biomarkers for PE.
Collapse
|
122
|
Balaguer N, Moreno I, Herrero M, Gonzáléz-Monfort M, Vilella F, Simón C. MicroRNA-30d deficiency during preconception affects endometrial receptivity by decreasing implantation rates and impairing fetal growth. Am J Obstet Gynecol 2019; 221:46.e1-46.e16. [PMID: 30826341 DOI: 10.1016/j.ajog.2019.02.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/30/2019] [Accepted: 02/22/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Maternal-embryonic crosstalk between the endometrium and the preimplantation embryo is required for normal pregnancy. Our previous results demonstrated that maternal microRNAs secreted into the endometrial fluid, specifically miR-30d, act as a transcriptomic regulator of the preimplantation embryo by the maternal intrauterine environment. OBJECTIVE To investigate the reproductive and fetal effects of murine miR-30d deficiency at the maternal-embryonic interface according to the origin of its maternal or embryonic default. STUDY DESIGN A miR-30d knockout murine model was used as the animal model to investigate the impact of maternal and/or embryonic origin of miR-30d deficiency on embryonic implantation and fetal development. Wild-type and miR-30d knockout pseudopregnant mice were used to study the effect of miR-30d deficiency on the receptivity markers by means of real-time quantitative polymerase chain reaction, immunofluorescence, and western blotting. We assessed receptivity markers and implantation rates in 6 different transfer conditions in which embryos obtained from wild-type, knockout, and knockout embryos pretreated with a miR-30d analog were transferred into either wild-type or knockout pseudopregnant females. The impact of miR-30d deficiency on fetal development was evaluated by analyzing the implantation sites and resorbing sites under physiological conditions at days 5, 6, 8, and 12 of pregnancy. Fetal growth was evaluated by analyzing fetuses and placentas at days 12 and 16 of pregnancy. RESULTS Maternal miR-30d deficiency induced a significant downregulation of endometrial receptivity markers. In wild-type recipients, miR-30d knockout embryos had poorer implantation rates than wild-type embryos (48.86 ± 14.33% vs 75.00 ± 10.47%, respectively, P = .0061). In miR-30d knockout recipients, the lowest implantation rate was observed when knockout embryos were transferred compared to wild-type embryos (26.04 ± 7.15% and 49.71 ± 8.59%, respectively, P = .0059). A positive correlation (r = 0.9978) was observed for maternal leukemia inhibitor factor expression with implantation rates. Further, the course of gestation was compromised in miR-30d knockout mothers, which had smaller implantation sites, greater rates of resorption, and fetuses with smaller crown-rump length and fetal/placental weight ratio. CONCLUSION Our results demonstrate that maternal and/or embryonic miR-30d deficiency impairs embryonic implantation and fetal development in the animal model. This finding adds a novel miRNA dimension to the understanding of pregnancy and fetal growth restriction in humans.
Collapse
Affiliation(s)
- Nuria Balaguer
- Department of Pediatrics, Obstetrics, and Gynecology, Universidad de Valencia, Valencia, Spain
| | | | - María Herrero
- R&D Department, Igenomix Foundation, Valencia, Spain
| | | | - Felipe Vilella
- R&D Department, Igenomix Foundation, Valencia, Spain; Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain.
| | - Carlos Simón
- Igenomix S.L., Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, INCLIVA, Valencia, Spain; Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA
| |
Collapse
|
123
|
de Brito Alves JL, de Oliveira Y, Carvalho NNC, Cavalcante RGS, Pereira Lira MM, Nascimento LCPD, Magnani M, Vidal H, Braga VDA, de Souza EL. Gut microbiota and probiotic intervention as a promising therapeutic for pregnant women with cardiometabolic disorders: Present and future directions. Pharmacol Res 2019; 145:104252. [PMID: 31054952 DOI: 10.1016/j.phrs.2019.104252] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/10/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022]
Abstract
Maternal cardiometabolic disorders, such as gestational diabetes mellitus, pre-eclampsia, obesity, and dyslipidemia, are the most common conditions that predispose offspring to risk for future cardiometabolic diseases, needing appropriate therapeutic approach. The implications of microbiota in the pathophysiology of maternal cardiometabolic disorders are progressively emerging and probiotics may be a simple and safe therapeutic strategy for maternal cardiometabolic management. In this review, we argue the importance of cardiometabolic dysfunction during pregnancy and/or lactation on the offspring risk for cardiometabolic disease in later life. In addition, we comprehensively discuss the microbial diversity observed in maternal cardiometabolic disorders and we present the main findings on probiotic intervention as a potential strategy for management of maternal cardiometabolic disorders. Current data reveal that gut microbiota may be transmitted from mother to offspring. Whether targeting microbiota with probiotic intervention during the periconceptional period prevents or delays the onset of cardiometabolic disorders in adult offspring should be tested in future clinical trials.
Collapse
Affiliation(s)
- José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil.
| | - Yohanna de Oliveira
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil
| | | | | | | | | | - Marciane Magnani
- Department of Food Engineering, Technology Center, Federal University of Paraiba, Joao Pessoa, Brazil
| | - Hubert Vidal
- Univ-Lyon, CarMeN(Cardio, Metabolism,Diabetes and Nutrition) Laboratory, INSERM U1060, INRA U1397, Université Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - Valdir de Andrade Braga
- Department of Biotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil
| |
Collapse
|
124
|
Nandi AA, Wadhwani NS, Joshi SR. Maternal vitamin D deficiency increases the thromboxane/prostacyclin ratio through alterations in the one-carbon cycle in Wistar rats. Biofactors 2019; 45:548-555. [PMID: 30985971 DOI: 10.1002/biof.1510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/23/2019] [Indexed: 01/03/2023]
Abstract
This study aims to test the hypothesis that vitamin D deficiency can influence long-chain polyunsaturated fatty acid metabolism through alterations in the one-carbon cycle. Wistar rats (n = 8 per group) were given either a control (1,000 IU D3/kg diet) or a vitamin D deficient (VDD) (0 IU D3/kg diet) diet from pre-pregnancy to delivery. On day 20 of gestation, pregnant female rats were delivered by C-section to collect placenta and blood. VDD group demonstrated high serum parathyroid hormone, low serum phosphate, low plasma folate, higher plasma homocysteine, and higher plasma malondialdehyde levels (P < 0.05 for all) as compared to control. Lower protein levels of placental cystathionine-β-synthase enzyme (P < 0.05) were observed in the VDD group as compared to control. VDD group demonstrated higher placental mRNA levels of the enzymes phospholipase A2 and cyclooxygenase-2 (P < 0.05 for both) as compared to control. Protein levels of the enzymes phospholipase A2 and cyclooxygenase-2 were lower (P < 0.05 for both) in the VDD group as compared to the control group. The ratio of thromboxane B2 and 6-keto prostaglandin F1α in serum was higher (P < 0.05) in the VDD group as compared to control; although the serum levels of 6-keto prostaglandin F1α and thromboxane B2 were similar in both the groups. Our findings suggest that increased oxidative stress due to maternal vitamin D deficiency results in the imbalance between the vasoconstrictor (thromboxane B2 ) and vasodilator (6-keto prostaglandin F1α ) eicosanoids, which may lead to endothelial dysfunction and poor pregnancy outcome. © 2019 BioFactors, 45 (4):548-555, 2019.
Collapse
Affiliation(s)
- Anindita A Nandi
- Department of Nutritional Medicine, Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth, Pune Satara Road, Pune, 411043, Maharashtra, India
| | - Nisha S Wadhwani
- Department of Nutritional Medicine, Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth, Pune Satara Road, Pune, 411043, Maharashtra, India
| | - Sadhana R Joshi
- Department of Nutritional Medicine, Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth, Pune Satara Road, Pune, 411043, Maharashtra, India
| |
Collapse
|
125
|
Profiles of gene expression in maternal blood predict offspring birth weight in normal pregnancy. J Dev Orig Health Dis 2019; 10:676-682. [PMID: 31204630 DOI: 10.1017/s2040174419000175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The association between lower birth weight and increased disease risk in adulthood has drawn attention to the physiological processes that shape the gestational environment. We implement genome-wide transcriptional profiling of maternal blood samples to identify subsets of genes and associated transcription control pathways that predict offspring birth weight. Female participants (N = 178, mean = 27.0 years) in a prospective observational birth cohort study were contacted between 2009 and 2014 to identify new pregnancies. An in-home interview was scheduled for early in the third trimester (mean = 30.3 weeks) to collect pregnancy-related information and a blood sample, and birth weight was measured shortly after delivery. Transcriptional activity in white blood cells was determined with a whole-genome gene expression direct hybridization assay. Fifty transcripts were differentially expressed in association with offspring birth weight, with 18 up-regulated in relation to lower birth weight, and 32 down-regulated. Examination of transcription control pathways identified increased activity of NF-κB, AP-1, EGR1, EGR4, and Gfi families, and reduced the activity of CEBP, in association with lower birth weight. Transcript origin analyses identified non-classical CD16+ monocytes, CD1c+ myeloid dendritic cells, and neutrophils as the primary cellular mediators of differential gene expression. These results point toward a systematic regulatory shift in maternal white blood cell activity in association with lower offspring birth weight, and they suggest that analyses of gene expression during gestation may provide insight into regulatory and cellular mechanisms that influence birth outcomes.
Collapse
|
126
|
Babischkin JS, Aberdeen GW, Lindner JR, Bonagura TW, Pepe GJ, Albrecht ED. Vascular Endothelial Growth Factor Delivery to Placental Basal Plate Promotes Uterine Artery Remodeling in the Primate. Endocrinology 2019; 160:1492-1505. [PMID: 31002314 PMCID: PMC6542484 DOI: 10.1210/en.2019-00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Extravillous trophoblast (EVT) uterine artery remodeling (UAR) promotes placental blood flow, but UAR regulation is unproven. Elevating estradiol (E2) in early baboon pregnancy suppressed UAR and EVT vascular endothelial growth factor (VEGF) expression, but this did not prove that VEGF mediated this process. Therefore, our primate model of prematurely elevating E2 and contrast-enhanced ultrasound cavitation of microbubble (MB) carriers was used to deliver VEGF DNA to the placental basal plate (PBP) to establish the role of VEGF in UAR. Baboons were treated on days 25 to 59 of gestation (term, 184 days) with E2 alone or with E2 plus VEGF DNA-conjugated MBs briefly infused via a maternal peripheral vein on days 25, 35, 45, and 55. At each of these times an ultrasound beam was directed to the PBP to collapse the MBs and release VEGF DNA. VEGF DNA-labeled MBs per contrast agent was localized in the PBP but not the fetus. Remodeling of uterine arteries >25 µm in diameter on day 60 was 75% lower (P < 0.001) in E2-treated (7% ± 2%) than in untreated baboons (30% ± 4%) and was restored to normal by E2/VEGF. VEGF protein levels (signals/nuclear area) within the PBP were twofold lower (P < 0.01) in E2-treated (4.2 ± 0.9) than in untreated (9.8 ± 2.8) baboons and restored to normal by E2/VEGF (11.9 ± 1.6), substantiating VEGF transfection. Thus, VEGF gene delivery selectively to the PBP prevented the decrease in UAR elicited by prematurely elevating E2 levels, establishing the role of VEGF in regulating UAR in vivo during primate pregnancy.
Collapse
Affiliation(s)
- Jeffery S Babischkin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Graham W Aberdeen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon
| | | | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Eugene D Albrecht
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
- Correspondence: Eugene D. Albrecht, PhD, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Bressler Research Laboratories 11-019, 655 West Baltimore Street, Baltimore, Maryland 21201. E-mail:
| |
Collapse
|
127
|
Wang H, She G, Zhou W, Liu K, Miao J, Yu B. Expression profile of circular RNAs in placentas of women with gestational diabetes mellitus. Endocr J 2019; 66:431-441. [PMID: 30814439 DOI: 10.1507/endocrj.ej18-0291] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Forty-five pregnant women who underwent cesarean section, including 30 cases of gestational diabetes mellitus (GDM) and 15 normal pregnant women, were enrolled in this study to examine the differential expression of circular RNAs (circRNAs) in the placentas of women with GDM by RNA sequencing (RNA-seq) analysis. The differentially expressed circRNAs were analyzed bioinformatically using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and circRNA-microRNA (miRNA) interaction prediction. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to verify the results. A total of 8,321 circRNAs were identified in the human placenta, among which 46 were differentially expressed (fold change ≥2 and p < 0.05), including three that were upregulated and 43 that were downregulated. According to the GO and KEGG enrichment results, these circRNAs may be associated with vital biological processes, cellular components, molecular functions, and signaling pathways. In particular, KEGG analysis shown they may be involved in advanced glycation end products-receptor for advanced glycation end products (AGE-RAGE) signaling pathway in diabetic complications, indicating that these circRNAs might participate in the occurrence and pathogenesis of GDM. qRT-PCR verified that the expression of circ_5824, circ_3636, and circ_0395 was consistent with RNA-seq analysis; their expression levels were significantly lower in the GDM group than in the control group. The circRNA-miRNA interaction was analyzed according to the molecular sponge mechanism, and its potential function is discussed. These results shed light on future functional studies of circRNAs related to GDM.
Collapse
Affiliation(s)
- Huiyan Wang
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Guangtong She
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Wenbai Zhou
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Kezhuo Liu
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Jun Miao
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Bin Yu
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213003, China
| |
Collapse
|
128
|
Cho KHT, Xu B, Blenkiron C, Fraser M. Emerging Roles of miRNAs in Brain Development and Perinatal Brain Injury. Front Physiol 2019; 10:227. [PMID: 30984006 PMCID: PMC6447777 DOI: 10.3389/fphys.2019.00227] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
In human beings the immature brain is highly plastic and depending on the stage of gestation is particularly vulnerable to a range of insults that if sufficiently severe, can result in long-term motor, cognitive and behavioral impairment. With improved neonatal care, the incidence of major motor deficits such as cerebral palsy has declined with prematurity. Unfortunately, however, milder forms of injury characterized by diffuse non-cystic white matter lesions within the periventricular region and surrounding white matter, involving loss of oligodendrocyte progenitors and subsequent axonal hypomyelination as the brain matures have not. Existing therapeutic options for treatment of preterm infants have proved inadequate, partly owing to an incomplete understanding of underlying post-injury cellular and molecular changes that lead to poor neurodevelopmental outcomes. This has reinforced the need to improve our understanding of brain plasticity, explore novel solutions for the development of protective strategies, and identify biomarkers. Compelling evidence exists supporting the involvement of microRNAs (miRNAs), a class of small non-coding RNAs, as important post-transcriptional regulators of gene expression with functions including cell fate specification and plasticity of synaptic connections. Importantly, miRNAs are differentially expressed following brain injury, and can be packaged within exosomes/extracellular vesicles, which play a pivotal role in assuring their intercellular communication and passage across the blood-brain barrier. Indeed, an increasing number of investigations have examined the roles of specific miRNAs following injury and regeneration and it is apparent that this field of research could potentially identify protective therapeutic strategies to ameliorate perinatal brain injury. In this review, we discuss the most recent findings of some important miRNAs in relation to the development of the brain, their dysregulation, functions and regulatory roles following brain injury, and discuss how these can be targeted either as biomarkers of injury or neuroprotective agents.
Collapse
Affiliation(s)
- Kenta Hyeon Tae Cho
- Department of Physiology, Faculty of Medical Health and Sciences, University of Auckland, Auckland, New Zealand
| | - Bing Xu
- Department of Physiology, Faculty of Medical Health and Sciences, University of Auckland, Auckland, New Zealand
| | - Cherie Blenkiron
- Departments of Molecular Medicine and Pathology, Faculty of Medical Health and Sciences, University of Auckland, Auckland, New Zealand
| | - Mhoyra Fraser
- Department of Physiology, Faculty of Medical Health and Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
129
|
Hobel CJ, Dolan SM, Hindoyan NA, Zhong N, Menon R. History of the establishment of the Preterm Birth international collaborative (PREBIC). Placenta 2019; 79:3-20. [PMID: 31047707 DOI: 10.1016/j.placenta.2019.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/16/2019] [Accepted: 03/20/2019] [Indexed: 01/25/2023]
Abstract
INTRODUCTION The primary aim of PREBIC is to assess the underlying mechanisms and developing strategies for preterm birth (PTB) prevention. MATERIALS AND METHODS We used concept mapping and logic models to track goals. This paper reviews our progress over 13 years using working group activities, research developments, guest speakers, and publications. RESULTS Using interactions between genetics, environment, and behaviors we identified complex interactions between biological systems. PREBIC determined that epidemiology and biomarkers should be an initial focus. In 2005, we initiated presentations by young investigators, yearly satellite meetings, working groups including nutrition and inflammation, assessment of clinical trials, and accepted an invitation by the WHO to begin yearly meetings in Geneva. DISCUSSION PREBIC used epidemiology to identify PTB factors and complex pathways. Candidate genes are associated with the environment, behavior (stress), obesity, inflammation and insulin resistance. Epigenetic changes and production of proteins can be used as biomarkers to define risk. Subsequently, we found risk factors for PTB that were also associated with the risk of cardiovascular disease (CVD) of the mother. Tanz et al. (2017) found that a history of PTB is independently predictive of CVD later in life and suggested that a modest proportion of PTB-CVD association was accounted by CVD risk factors, many of which have been identified in this paper. CONCLUSION Our findings support a relationship between genes, environment, behaviors and risk of CVD in women. The next several years must assess which factors are modifiable early in life and before pregnancy to prevent PTB.
Collapse
Affiliation(s)
- Calvin J Hobel
- Departments of OB/GYN & Pediatrics, Cedars-Sinai Medical Center, 8635 West 3rd St. Suite 160W, Los Angeles, CA, 90048, USA; David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, 90095-1740, USA.
| | - Siobhan M Dolan
- Department of Obstetrics & Gynecology and Women's Health, Montefiore Medical Center/Albert Einstein College of Medicine, 1695 Eastchester Road Suite 301, Bronx, NY, 10461, USA.
| | - Niree A Hindoyan
- Department of Medicine, Cedars-Sinai Medical Center, 8730 Alden Drive Room W215, Los Angeles, CA, 90048, USA.
| | - Nanbert Zhong
- Developmental Genetics Laboratory, Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY, 10314, USA.
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Maternal-Fetal Medicine, Perinatal Research Division, University of Texas Medical Branch MRB 11.138, 301 University Blvd, Galveston, TX, 7755-1062, USA.
| |
Collapse
|
130
|
Biochemical Markers for Prediction of Hypertensive Disorders of Pregnancy. J Med Biochem 2019; 38:71-82. [PMID: 30820186 PMCID: PMC6298456 DOI: 10.2478/jomb-2018-0001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/27/2018] [Indexed: 12/01/2022] Open
Abstract
Background Gestational hypertension (GH) and pre eclampsia (PE) are the most common gestational complications. Several placental biochemical markers are used to predict GH/PE, but with conflicting results. Methods The study aim was to estimate the biochemical markers’ ability to predict hypertensive disorders in pregnancy. On the first ultrasonographic examination, 104 healthy pregnant women were recruited. At the regular pregnancy check-ups, BMI, blood pressure, occurrence of gestational hypertension (early or late onset), preeclampsia, eclampsia and other complications were recorded. Serum concentrations (in multiples of median – MoM) of human chorionic gonadotropin (HCG) and pregnancyassociated plasma protein A (PAPPA) were measured from the 11th to 14th gestational week, while HCG, alpha feto protein (AFP), estriol and inhibin were determined between the 16th and 19th gestational week. Results Hypertensive disorders throughout pregnancy were diagnosed in 20.2% women. Early-onset GH was registered in 7 and PE in 6 patients, while 14 had late-onset GH and 10 additional women PE. There were no significant differences (p≥0.05) in biochemical markers concentrations between women with and without GH/PE. PAPPA levels in the first and HCG in the second trimester correlated with early and late GH/PE. Moreover, higher AFP concentrations were registered in women with preeclampsia signs/symptoms. According to ROC analysis, AFP>1.05 MoM properly identified 80% of GH/PE cases. Obtained models imply that HCG, PAPPA and AFP should be used for GH/PE prediction. Conclusions Biochemical markers HCG, PAPPA and AFP could be useful in predicting gestational hypertension and preeclampsia. However, different markers should be used for early and late onset GH/PE.
Collapse
|
131
|
Trophoblast-Specific Expression of Hif-1α Results in Preeclampsia-Like Symptoms and Fetal Growth Restriction. Sci Rep 2019; 9:2742. [PMID: 30808910 PMCID: PMC6391498 DOI: 10.1038/s41598-019-39426-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/17/2018] [Indexed: 01/02/2023] Open
Abstract
The placenta is an essential organ that is formed during pregnancy and its proper development is critical for embryonic survival. While several animal models have been shown to exhibit some of the pathological effects present in human preeclampsia, these models often do not represent the physiological aspects that have been identified. Hypoxia-inducible factor 1 alpha (Hif-1α) is a necessary component of the cellular oxygen-sensing machinery and has been implicated as a major regulator of trophoblast differentiation. Elevated levels of Hif-1α in the human placenta have been linked to the development of pregnancy-associated disorders, such as preeclampsia and fetal growth restriction. As oxygen regulation is a critical determinant for placentogenesis, we determined the effects of constitutively active Hif-1α, specifically in trophoblasts, on mouse placental development in vivo. Our research indicates that prolonged expression of trophoblast-specific Hif-1α leads to a significant decrease in fetal birth weight. In addition, we noted significant physiological alterations in placental differentiation that included reduced branching morphogenesis, alterations in maternal and fetal blood spaces, and failure to remodel the maternal spiral arteries. These placental alterations resulted in subsequent maternal hypertension with parturitional resolution and maternal kidney glomeruloendotheliosis with accompanying proteinuria, classic hallmarks of preeclampsia. Our findings identify Hif-1α as a critical molecular mediator of placental development and indicate that prolonged expression of Hif-1α, explicitly in placental trophoblasts causes maternal pathology and establishes a mouse model that significantly recapitulates the physiological and pathophysiological characteristics of preeclampsia with fetal growth restriction.
Collapse
|
132
|
Wu H, Wu S, Zhu Y, Ye M, Shen J, Liu Y, Zhang Y, Bu S. Hsa_circRNA_0054633 is highly expressed in gestational diabetes mellitus and closely related to glycosylation index. Clin Epigenetics 2019; 11:22. [PMID: 30736847 PMCID: PMC6368772 DOI: 10.1186/s13148-019-0610-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background Circular RNA (circRNA) is involved in the pathological processes of various diseases. CircRNA is more stable than linear RNAs and is expressed in high levels in tissues, making it a better biomarker candidate than linear RNAs. In this study, we aimed to identify potential circRNA biomarkers of gestational diabetes mellitus (GDM). Methods A retrospective case–control study was conducted using data and samples from women treated at a hospital in China between July 10, 2017, and February 15, 2018. We collected serum samples from 40 healthy pregnant women (controls) and 40 women with GDM (cases) during the second trimester as well as 65 controls and 65 cases during the third trimester of pregnancy. Placenta tissues and neonatal cord blood were each from another 20 cases and 20 controls. We selected six circRNAs (hsa_circRNA_0054633, hsa_circRNA_103410, hsa_circRNA_063981, hsa_circRNA_102682, hsa_circRNA_0018508, and hsa_circRNA_406918) as candidate biomarkers and used quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) to measure their concentrations in the serum and placental tissues. The Pearson correlation test was used to assess the correlation between various circRNAs and between circRNA and clinical variables. The area under the receiver operating characteristic (ROC) curve was used to assess the diagnostic value of circRNAs for GDM at each stage. Results Hsa_circRNA_0054633 was highly expressed in the blood during the second and third trimesters; its expression was also high in the placenta but low in the cord blood (P < 0.05). Hsa_cirRNA_0054633 was highly correlated with GHBA1 and GHBA1c levels in maternal blood samples at various stages of the GDM group (including placental tissue and umbilical cord blood) (P < 0.05). Hsa_circRNA_063981, hsa_circRNA_102682, and hsa_circRNA_103410 were also differentially expressed between the case and control groups at different stages (P < 0.05). There was a strong correlation between hsa_circRNA_0054633 and hsa_circRNA_103410 levels in third-trimester maternal blood (P = 0.000, r = 0.554) and in neonatal umbilical cord blood (P = 0.000, r = 0.866). Hsa_circRNA_0054633 showed a significant diagnostic value in the second and third trimesters of pregnancy, placenta, and cord blood (AUC = 0.793, 0.664, 0.747, and 0.783, respectively, P < 0.001). Conclusion This study suggests that hsa_cirRNA_0054633 is abnormally expressed in GDM patients and may play a potential role in the development of GDM. The possibility of using circRNAs for the diagnosis of GDM requires additional investigation in future studies. Electronic supplementary material The online version of this article (10.1186/s13148-019-0610-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hangyu Wu
- Diabetes Research Center, Medical School, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China
| | - Siyang Wu
- Diabetes Research Center, Medical School, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China
| | - Yingchao Zhu
- Diabetes Research Center, Medical School, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China
| | - Mei Ye
- Department of Gynaecology and Obstetrics, Ningbo University Medical Center Lihuili Eastern Hospital, Taipei Medical University, 1111 Jiangnan Road, Ningbo, 315048, Zhejiang, China
| | - Jun Shen
- Department of Gynaecology and Obstetrics, Ningbo University Medical Center Lihuili Eastern Hospital, Taipei Medical University, 1111 Jiangnan Road, Ningbo, 315048, Zhejiang, China
| | - Yan Liu
- Department of Gynaecology and Obstetrics, Ningbo University Medical Center Lihuili Eastern Hospital, Taipei Medical University, 1111 Jiangnan Road, Ningbo, 315048, Zhejiang, China
| | - Yisheng Zhang
- Department of Gynaecology and Obstetrics, Ningbo University Medical Center Lihuili Eastern Hospital, Taipei Medical University, 1111 Jiangnan Road, Ningbo, 315048, Zhejiang, China.
| | - Shizhong Bu
- Diabetes Research Center, Medical School, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
133
|
Tadalafil treatment in mice for preeclampsia with fetal growth restriction has neuro-benefic effects in offspring through modulating prenatal hypoxic conditions. Sci Rep 2019; 9:234. [PMID: 30659198 PMCID: PMC6338749 DOI: 10.1038/s41598-018-36084-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023] Open
Abstract
We have demonstrated that tadalafil facilitates fetal growth in mice with L-NG-nitroarginine methyl ester (L-NAME)-induced preeclampsia (PE) with fetal growth restriction (FGR). Tadalafil is a selective phosphodiesterase 5 inhibitor that dilates the maternal blood sinuses in the placenta, thereby facilitating the growth of the fetus. The purpose of this study was to investigate the effects of tadalafil treatment for PE and FGR on the developing brain in FGR offspring using an L-NAME-induced mouse model of PE with FGR. A control group of dams received carboxymethylcellulose (CMC). L-NAME-treated groups received L-NAME dissolved in CMC from 11 days post coitum (d.p.c.). The L-NAME-treated dams were divided into two subgroups 14 d.p.c. One subgroup continued to receive L-NAME. The other subgroup received L-NAME with tadalafil suspended in CMC. Tadalafil treatment for PE with FGR reduced the expression of hypoxia-inducible factor-2α in the placenta and in the brain of the FGR fetus. Moreover, tadalafil treatment in utero shows improved synaptogenesis and myelination in FGR offspring on postnatal day 15 (P15) and P30. These results suggest that tadalafil treatment for PE with FGR not only facilitates fetal growth, but also has neuroprotective effects on the developing brain of FGR offspring through modulating prenatal hypoxic conditions.
Collapse
|
134
|
Xu C, Chen X, Reece EA, Lu W, Yang P. The increased activity of a transcription factor inhibits autophagy in diabetic embryopathy. Am J Obstet Gynecol 2019; 220:108.e1-108.e12. [PMID: 30312583 DOI: 10.1016/j.ajog.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Maternal diabetes induces neural tube defects and stimulates the activity of the forkhead box O3 (Fox)O3a in the embryonic neuroepithelium. We previously demonstrated that deleting the FOXO3a gene ameliorates maternal diabetes-induced neural tube defects. Macroautophagy (hereafter referred to as "autophagy") is essential for neurulation. Rescuing autophagy suppressed by maternal diabetes in the developing neuroepithelium inhibits neural tube defect formation in diabetic pregnancy. This evidence suggests a possible link between FoxO3a and impaired autophagy in diabetic embryopathy. OBJECTIVE We aimed to determine whether maternal diabetes suppresses autophagy through FoxO3a, and if the transcriptional activity of FoxO3a is required for the induction of diabetic embryopathy. STUDY DESIGN We used a well-established type 1 diabetic embryopathy mouse model, in which diabetes was induced by streptozotocin, for our in vivo studies. To determine if FoxO3a mediates the inhibitory effect of maternal diabetes on autophagy in the developing neuroepithelium, we induced diabetic embryopathy in FOXO3a gene knockout mice and FoxO3a dominant negative transgenic mice. Embryos were harvested at embryonic day 8.5 to determine FoxO3a and autophagy activity and at embryonic day 10.5 for the presence of neural tube defects. We also examined the expression of autophagy-related genes. C17.2 neural stem cells were used for in vitro examination of the potential effects of FoxO3a on autophagy. RESULTS Deletion of the FOXO3a gene restored the autophagy markers, lipidation of microtubule-associated protein 1A/1B-light chain 3I to light chain 3II, in neurulation stage embryos. Maternal diabetes decreased light chain 3I-positive puncta number in the neuroepithelium, which was restored by deleting FoxO3a. Maternal diabetes also decreased the expression of positive regulators of autophagy (Unc-51 like autophagy activating kinase 1, Coiled-coil myosin-like BCL2-interacting protein, and autophagy-related gene 5) and the negative regulator of autophagy, p62. FOXO3a gene deletion abrogated the dysregulation of autophagy genes. In vitro data showed that the constitutively active form of FoxO3a mimicked high glucose in repressing autophagy. In cells cultured under high-glucose conditions, overexpression of the dominant negative FoxO3a mutant blocked autophagy impairment. Dominant negative FoxO3a overexpression in the developing neuroepithelium restored autophagy and significantly reduced maternal diabetes-induced apoptosis and neural tube defects. CONCLUSION Our study revealed that diabetes-induced FoxO3a activation inhibited autophagy in the embryonic neuroepithelium. We also observed that FoxO3a transcriptional activity mediated the teratogenic effect of maternal diabetes because dominant negative FoxO3a prevents maternal diabetes-induced autophagy impairment and neural tube defect formation. Our findings suggest that autophagy activators could be therapeutically effective in treating maternal diabetes-induced neural tube defects.
Collapse
|
135
|
Kuna M, Waller JP, Logue OC, Bidwell GL. Polymer size affects biodistribution and placental accumulation of the drug delivery biopolymer elastin-like polypeptide in a rodent pregnancy model. Placenta 2018; 72-73:20-27. [PMID: 30501877 PMCID: PMC6287274 DOI: 10.1016/j.placenta.2018.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/19/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Fusion of therapeutic agents to Elastin-like Polypeptide (ELP) is a novel drug delivery strategy for prevention of placental drug transfer. Previous studies have used a 60 kDa ELP tag for this purpose. However, placental transfer of ELP may be size dependent. The goal of this study was to measure the effects of ELP polymer size on pharmacokinetics, biodistribution, and placental transfer of ELP. METHODS Three ELPs ranging from 25 to 86 kDa (4.1-6.8 nm hydrodynamic radius) were fluorescently labeled and administered by i.v. bolus to pregnant Sprague Dawley rats on gestational day 14. Plasma levels were monitored for 4 h, organ levels and placental transfer determined by ex vivo fluorescence imaging, and placental localization determined by confocal microscopy. RESULTS Increasing ELP size resulted in slower plasma clearance and increased deposition in all major maternal organs, except in the kidneys where an opposite effect was observed. Placental levels increased with an increase in size, while in the pups, little to no ELP was detected. DISCUSSION Pharmacokinetics and biodistribution of ELPs during pregnancy are size dependent, but all ELPs tested were too large to traverse the placental barrier. These studies verify that ELP fusion is a powerful method of modulating half-life and preventing placental transfer of cargo molecules. The tunable nature of the ELP sequence makes it ideal for drug delivery applications during pregnancy, where it can be used to target drugs to the mother while preventing fetal drug exposure.
Collapse
Affiliation(s)
- Marija Kuna
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jamarius P Waller
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Omar C Logue
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gene L Bidwell
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
136
|
Guo D, Jiang H, Chen Y, Yang J, Fu Z, Li J, Han X, Wu X, Xia Y, Wang X, Chen L, Tang Q, Wu W. Elevated microRNA-141-3p in placenta of non-diabetic macrosomia regulate trophoblast proliferation. EBioMedicine 2018; 38:154-161. [PMID: 30420300 PMCID: PMC6306401 DOI: 10.1016/j.ebiom.2018.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/21/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Several studies have reported microRNAs (miRNAs) could regulate the placental development, though the role and mechanism of miRNAs in the development of non-diabetic macrosomia (NDFMS) remains unclear. METHODS To identify the aberrantly expressed key miRNAs in placenta of NDFMS, we employed a strategy consisting of initial screening with miRNA microarray and further validation with quantitative RT-PCR assay (qRT-PCR). In vitro cellular model and a mouse pregnancy model were used to delineate the functional effects of key miRNA on proliferation, invasion, and migration. FINDINGS miR-141-3p was identified as the key miRNA with expression level significantly higher in placentas of NDFMS compared with those from normal controls. Overexpressed miR-141-3p in HTR-8/SVneo cells contributed to increased cell proliferation, invasion, and migration. miR-141-3p inhibition in HTR-8/SVneo cells resulted in decreased cell proliferation and invasion. Significantly increased infant birth weight was observed in late pregnancy of C57BL/6J mice treated with miR-141-3p agomir. However, no significant difference was found in early pregnancy of C57BL/6J mice treated with miR-141-3p agomir. INTERPRETATION miR-141-3p could stimulate placental cell proliferation to participate in the occurrence and development of NDFMS.
Collapse
Affiliation(s)
- Dan Guo
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Preventive Health Branch, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| | - Hua Jiang
- Department of Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yiqiu Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jing Yang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ziqiang Fu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jing Li
- Department of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xian Wu
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Liping Chen
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Nantong University, Nantong, China.
| | - Qiuqin Tang
- Department of Obstetrics, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China; National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA.
| |
Collapse
|
137
|
Loch-Caruso R, Hassan I, Harris SM, Kumar A, Bjork F, Lash LH. Trichloroethylene exposure in mid-pregnancy decreased fetal weight and increased placental markers of oxidative stress in rats. Reprod Toxicol 2018; 83:38-45. [PMID: 30468822 DOI: 10.1016/j.reprotox.2018.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 12/29/2022]
Abstract
Although epidemiology studies have associated maternal trichloroethylene (TCE) exposure with decreased birth weight and preterm birth, mechanistic explanations for these associations are currently lacking. We hypothesized that TCE targets the placenta with adverse consequences for pregnancy outcomes. Pregnant Wistar rats were exposed orally to vehicle or 480 mg TCE/kg body weight from gestational days (gd) 6-16, and tissues were collected on gd 16. Exposure to TCE significantly decreased average fetal weight without reducing maternal weight. In placenta, TCE significantly increased 8-hydroxy-deoxyguanosine, global 5-hydroxymethylcytosine, and mRNA expression of Tet3, which codes for an enzyme involved in 5-hydroxymethylcytosine formation. Furthermore, glutathione S-transferase activity and immunohistochemical staining were increased in placentas of TCE-exposed rats. The present study provides the first evidence that TCE increases markers of oxidative stress in placenta in a fetal growth restriction rat model, providing new insight into the placenta as a potentially relevant target for TCE-induced adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Rita Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109-2029, USA.
| | - Iman Hassan
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109-2029, USA.
| | - Sean M Harris
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109-2029, USA.
| | - Anjana Kumar
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109-2029, USA.
| | - Faith Bjork
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109-2029, USA.
| | - Lawrence H Lash
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA.
| |
Collapse
|
138
|
Tsur A, Kalish F, Burgess J, Nayak NR, Zhao H, Casey KM, Druzin ML, Wong RJ, Stevenson DK. Pravastatin improves fetal survival in mice with a partial deficiency of heme oxygenase-1. Placenta 2018; 75:1-8. [PMID: 30712660 DOI: 10.1016/j.placenta.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/21/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Statins induce heme oxygenase-1 (HO-1) expression in vitro and in vivo. Low HO-1 expression is associated with pregnancy complications, e.g. preeclampsia and recurrent miscarriages. Here, we investigated the effects of pravastatin on HO-1 expression, placental development, and fetal survival in mice with a partial HO-1 deficiency. METHODS At E14.5, untreated pregnant wild-type (WT, n=13-18), untreated HO-1+/- (Het, n=6-9), and Het mice treated with pravastatin (Het+Pravastatin, n=12-14) were sacrificed. Numbers of viable fetuses/resorbed concepti were recorded. Maternal livers and placentas were harvested for HO activity. Hematoxylin and eosin (H&E) and CD31 immunohistochemical staining were performed on whole placentas. RESULTS Compared with WT, HO activity in Het livers (65±18%, P<0.001) and placentas (74±7%, P<0.001) were significantly decreased. Number of viable fetuses per dam was significantly lower in Untreated Het dams (6.0±2.2) compared with WT (9.1±1.4, P<0.01), accompanied by a higher relative risk (RR) for concepti resorption (17.1, 95% CI 4.0-73.2). In Hets treated with pravastatin, maternal liver and placental HO activity increased, approaching levels of WT controls (to 83±7% and 87±14%, respectively). The number of viable fetuses per dam increased to 7.7±2.5 with a decreased RR for concepti resorption (2.7, 95% CI 1.2-5.9). In some surviving Untreated Het placentas, there were focal losses of cellular architecture and changes suggestive of reduced blood flow in the labyrinth. These findings were absent in Het+Pravastatin placentas. DISCUSSION Pravastatin induces maternal liver and placental HO activity, may affect placental function and improve fetal survival in the context of a partial deficiency of HO-1.
Collapse
Affiliation(s)
- Abraham Tsur
- Dept of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Flora Kalish
- Dept of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jordan Burgess
- Dept of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nihar R Nayak
- Dept of Obstetrics & Gynecology, Wayne State University, Detroit, MI, USA
| | - Hui Zhao
- Dept of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kerriann M Casey
- Dept of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Maurice L Druzin
- Dept of Obstetrics & Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald J Wong
- Dept of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - David K Stevenson
- Dept of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
139
|
Hwuang E, Vidorreta M, Schwartz N, Moon BF, Kochar K, Tisdall MD, Detre JA, Witschey WRT. Assessment of uterine artery geometry and hemodynamics in human pregnancy with 4d flow mri and its correlation with doppler ultrasound. J Magn Reson Imaging 2018; 49:59-68. [PMID: 30390347 DOI: 10.1002/jmri.26229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/31/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Uterine artery (UtA) hemodynamics might be used to predict risk of hypertensive pregnancy disorders, including preeclampsia and intrauterine growth restriction. PURPOSE OR HYPOTHESIS To determine the feasibility of 4D flow MRI in pregnant subjects by characterizing UtA anatomy, computing UtA flow, and comparing UtA velocity, and pulsatility and resistivity indices (PI, RI) with transabdominal Doppler ultrasound (US). STUDY TYPE Prospective cross-sectional study from June 6, 2016, to May 2, 2018. POPULATION OR SUBJECTS OR PHANTOM OR SPECIMEN OR ANIMAL MODEL Forty-one singleton pregnant subjects (age [range] = 27.0 ± 5.9 [18-41] years) in their second or third trimester. We additionally scanned three subjects who had prepregnancy diabetes or chronic hypertension. FIELD STRENGTH/SEQUENCE The subjects underwent UtA and placenta MRI using noncontrast angiography and 4D flow at 1.5T. ASSESSMENT UtA anatomy was described based on 4D flow-derived noncontrast angiography, while UtA flow properties were characterized by net flow, systolic/mean/diastolic velocity, PI and RI through examination of 4D flow data. PI and RI are standard hemodynamic parameters routinely reported on Doppler US. STATISTICAL TESTS Spearman's rank correlation, Wilcoxon signed rank tests, and Bland-Altman plots were used to preliminarily investigate the relationships between flow parameters, gestational age, and Doppler US. or RESULTS: 4D flow MRI and UtA flow quantification was feasible in all subjects. There was considerable heterogeneity in UtA geometry in each subject between left and right UtAs and between subjects. Mean 4D flow-based parameters were: mean bilateral flow rate = 605.6 ± 220.5 mL/min, PI = 0.72 ± 0.2, and RI = 0.47 ± 0.1. Bilateral flow did not change with gestational age. We found that MRI differed from US in terms of lower PI (mean difference -0.1) and RI (mean difference < -0.1) with Wilcoxon signed rank test P = 0.05 and P = 0.13, respectively. DATA CONCLUSION 4D flow MRI is a feasible approach for describing UtA anatomy and flow in pregnant subjects. LEVEL OF EVIDENCE Technical Efficacy: Stage 1 J. Magn. Reson. Imaging 2019;49:59-68.
Collapse
Affiliation(s)
- Eileen Hwuang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marta Vidorreta
- Siemens Healthineers, Tarrytown, New York, USA.,Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nadav Schwartz
- Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brianna F Moon
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kirpal Kochar
- Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Matthew Dylan Tisdall
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John A Detre
- Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Walter R T Witschey
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
140
|
Romero R, Kim YM, Pacora P, Kim CJ, Benshalom-Tirosh N, Jaiman S, Bhatti G, Kim JS, Qureshi F, Jacques SM, Jung EJ, Yeo L, Panaitescu B, Maymon E, Hassan SS, Hsu CD, Erez O. The frequency and type of placental histologic lesions in term pregnancies with normal outcome. J Perinat Med 2018; 46:613-630. [PMID: 30044764 PMCID: PMC6174692 DOI: 10.1515/jpm-2018-0055] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/31/2018] [Indexed: 12/22/2022]
Abstract
Objective To determine the frequency and type of histopathologic lesions in placentas delivered by women with a normal pregnancy outcome. Methods This retrospective cohort study included placental samples from 944 women with a singleton gestation who delivered at term without obstetrical complications. Placental lesions were classified into the following four categories as defined by the Society for Pediatric Pathology and by our unit: (1) acute placental inflammation, (2) chronic placental inflammation, (3) maternal vascular malperfusion and (4) fetal vascular malperfusion. Results (1) Seventy-eight percent of the placentas had lesions consistent with inflammatory or vascular lesions; (2) acute inflammatory lesions were the most prevalent, observed in 42.3% of the placentas, but only 1.0% of the lesions were severe; (3) acute inflammatory lesions were more common in the placentas of women with labor than in those without labor; (4) chronic inflammatory lesions of the placenta were present in 29.9%; and (5) maternal and fetal vascular lesions of malperfusion were detected in 35.7% and 19.7%, respectively. Two or more lesions with maternal or fetal vascular features consistent with malperfusion (high-burden lesions) were present in 7.4% and 0.7%, respectively. Conclusion Most placentas had lesions consistent with inflammatory or vascular lesions, but severe and/or high-burden lesions were infrequent. Mild placental lesions may be interpreted either as acute changes associated with parturition or as representative of a subclinical pathological process (intra-amniotic infection or sterile intra-amniotic inflammation) that did not affect the clinical course of pregnancy.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- 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
| | - Yeon Mee Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Percy Pacora
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Neta Benshalom-Tirosh
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Sunil Jaiman
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Gaurav Bhatti
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
| | - Jung-Sun Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Pathology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Faisal Qureshi
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Suzanne M. Jacques
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Eun Jung Jung
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Lami Yeo
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Eli Maymon
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
| | - Offer Erez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
141
|
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: 4.7] [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
|
142
|
Fajersztajn L, Veras MM. Hypoxia: From Placental Development to Fetal Programming. Birth Defects Res 2018; 109:1377-1385. [PMID: 29105382 DOI: 10.1002/bdr2.1142] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022]
Abstract
Hypoxia may influence normal and different pathological processes. Low oxygenation activates a variety of responses, many of them regulated by hypoxia-inducible factor 1 complex, which is mostly involved in cellular control of O2 consumption and delivery, inhibition of growth and development, and promotion of anaerobic metabolism. Hypoxia plays a significant physiological role in fetal development; it is involved in different embryonic processes, for example, placentation, angiogenesis, and hematopoiesis. More recently, fetal hypoxia has been associated directly or indirectly with fetal programming of heart, brain, and kidney function and metabolism in adulthood. In this review, the role of hypoxia in fetal development, placentation, and fetal programming is summarized. Hypoxia is a basic mechanism involved in different pregnancy disorders and fetal health developmental complications. Although there are scientific data showing that hypoxia mediates changes in the growth trajectory of the fetus, modulates gene expression by epigenetic mechanisms, and determines the health status later in adulthood, more mechanistic studies are needed. Furthermore, if we consider that intrauterine hypoxia is not a rare event, and can be a consequence of unavoidable exposures to air pollution, nutritional deficiencies, obesity, and other very common conditions (drug addiction and stress), the health of future generations may be damaged and the incidence of some diseases will markedly increase as a consequence of disturbed fetal programming. Birth Defects Research 109:1377-1385, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lais Fajersztajn
- LIM 05 Departamento de Patologia, Hospital da Clinicas, Faculdade de Medicina Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Mariana Matera Veras
- LIM 05 Departamento de Patologia, Hospital da Clinicas, Faculdade de Medicina Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| |
Collapse
|
143
|
Abstract
The spectrum of sleep-disordered breathing (SDB) ranges from mild snoring to obstructive sleep apnea, the most severe form of SDB. Current recommendations are to treat these women with continuous positive airway pressure despite limited data. SDB in early and mid-pregnancy is associated with preeclampsia and gestational diabetes. Pregnant women with a diagnosis of obstructive sleep apnea at delivery were at significantly increased risk of having cardiomyopathy, congestive heart failure, pulmonary embolism, and in-hospital death. These effects were exacerbated in the presence of obesity. Postpartum, these women are at risk for respiratory suppression and should be monitored.
Collapse
Affiliation(s)
- Jennifer E Dominguez
- Department of Anesthesiology, Obstetric Anesthesiology, Division of Women's Anesthesia, Duke University Medical Center, Mail Sort #9, DUMC Box 3094, Durham, NC 27710, USA
| | - Linda Street
- Division of Maternal Fetal Medicine, Department of OB/GYN, Medical College of Georgia, Augusta University, 1120 15th Street, BA-7410, Augusta, GA 30912, USA
| | - Judette Louis
- Division of Maternal Fetal Medicine, Department of OB/GYN, University of South Florida, 2 Tampa General Circle Suite 6050, Tampa, FL 33606, USA.
| |
Collapse
|
144
|
Dominguez JE, Habib AS, Krystal AD. A review of the associations between obstructive sleep apnea and hypertensive disorders of pregnancy and possible mechanisms of disease. Sleep Med Rev 2018; 42:37-46. [PMID: 29929840 DOI: 10.1016/j.smrv.2018.05.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022]
Abstract
Obesity is prevalent among pregnant women in the United States; 15-20% of obese pregnant women have obstructive sleep apnea. The prevalence of obstructive sleep apnea increases along with body mass index, age and in the presence of other co-morbidities. Untreated obstructive sleep apnea in women is associated with a range of cardiovascular, pulmonary and metabolic co-morbidities; recent studies suggest that women with obstructive sleep apnea in pregnancy may be at significantly greater risk of entering pregnancy with chronic hypertension and/or of developing hypertensive disorders of pregnancy: gestational hypertension; preeclampsia; or eclampsia. This has serious public health implications; hypertensive disorders of pregnancy are a major cause of maternal and neonatal morbidity and mortality and are associated with a greater lifetime risk for cardiovascular disease. The mechanisms that associated obstructive sleep apnea with hypertensive disorders of pregnancy have not been defined, but several pathways are scientifically plausible. In this review, we will present a comprehensive literature review of the following: the associations between obstructive sleep apnea and hypertensive disorders of pregnancy; the proposed mechanisms that may connect obstructive sleep apnea and hypertensive disorders of pregnancy; and the effectiveness of treatment at mitigating these adverse outcomes.
Collapse
Affiliation(s)
| | - Ashraf S Habib
- Duke Department of Anesthesiology, Durham, NC, 27710, USA
| | - Andrew D Krystal
- Duke Department of Psychiatry and Behavioral Sciences, Durham, NC 27710, USA; University of California, San Francisco Department of Psychiatry, San Francisco, CA 94143, USA
| |
Collapse
|
145
|
Xu Z, Jin X, Cai W, Zhou M, Shao P, Yang Z, Fu R, Cao J, Liu Y, Yu F, Fan R, Zhang Y, Zou S, Zhou X, Yang N, Chen X, Li Y. Proteomics Analysis Reveals Abnormal Electron Transport and Excessive Oxidative Stress Cause Mitochondrial Dysfunction in Placental Tissues of Early-Onset Preeclampsia. Proteomics Clin Appl 2018; 12:e1700165. [PMID: 29676857 DOI: 10.1002/prca.201700165] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 04/06/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Early-onset preeclampsia (EOS-PE) refers to preeclampsia that occurred before 34 gestation weeks. This study is conducted to explore the relationship between mitochondrial dysfunction and the pathogenesis of EOS-PE using proteomic strategy. EXPERIMENTAL DESIGN To identify altering expressed mitochondrial proteins between severe EOS-PE and healthy pregnancies, enrichment of mitochondria coupled with iTRAQ-based quantitative proteomic method is performed. Immunohistochemistry (IHC) and western blot are performed to detect the alteration of changing expression proteins, and confirmed the accuracy of proteomic results. RESULTS A total of 1372 proteins were quantified and 132 altering expressed proteins were screened, including 86 downregulated expression proteins and 46 upregulated expression proteins (p < 0.05). Bioinformatics analysis showed that differentially expressed proteins participated in numerous biological processes, including oxidation-reduction process, respiratory electron transport chain, and oxidative phosphorylation. Especially, mitochondria-related molecules, PRDX2, PARK7, BNIP3, BCL2, PDHA1, SUCLG1, ACADM, and NDUFV1, are involved in energy-production process in the matrix and membrane of mitochondria. CONCLUSIONS AND CLINICAL RELEVANCE Results of the experiment show that abnormal electron transport, excessive oxidative stress, and mitochondrion disassembly might be the main cause of mitochondrial dysfunction, and is related to the pathogenesis of EOS-PE.
Collapse
Affiliation(s)
- Zhongwei Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China.,Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, 300162, China
| | - Xiaohan Jin
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China.,Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, 300162, China
| | - Wei Cai
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, 300162, China
| | - Maobin Zhou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Ping Shao
- Women and Children Health Care Center, Tianjin, 300070, China
| | - Zhen Yang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Rong Fu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Jin Cao
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Yan Liu
- Tianjin First Center Hospital, Tianjin, 300192, China
| | - Fang Yu
- Obstetrics and Gynecology Department, Pingjin Hospital, Tianjin, 300162, China
| | - Rong Fan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Yan Zhang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Shuang Zou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, 300162, China
| | - Ning Yang
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, 300162, China
| | - Xu Chen
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Yuming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, 300162, China
| |
Collapse
|
146
|
Abstract
PURPOSE OF REVIEW Although not fully understood, the physiopathology of preeclampsia is thought to involve an abnormal placentation, diffuse endothelial cell dysfunction and increased systemic inflammation. As micronutrients play a key role in placental endothelial function, oxidative stress and expression of angiogenic factors, periconceptional micronutrient supplementation has been proposed to reduce the risk of preeclampsia. However, recent studies reported conflicting results. RECENT FINDINGS Calcium intake (>1 g/day) may reduce the risk of preeclampsia in women with low-calcium diet. Data from recently updated Cochrane reviews did not support routine supplementation of vitamins C, E or D for either the prevention or treatment of preeclampsia. Evidences are also poor to support zinc or folic acid supplementation for preeclampsia prevention. Dark chocolate, flavonoid-rich food, and long-chain polyunsaturated fatty acids might also be candidates for prevention of preeclampsia. SUMMARY Through antioxidant, anti-inflammatory or vasoactive proprieties, micronutrients are good candidates for preeclampsia prevention. Calcium supplementation is recommended to prevent preeclampsia in women with low-calcium intake. Despite positive clinical and in-vitro data, strong evidence to support periconceptional supplementation of other micronutrients for preeclampsia risk-reduction is still lacking. Further studies are also needed to evaluate the benefit of nutritional supplementation such as chocolate and long-chain polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Najate Achamrah
- Department of Endocrinology, Diabetology, Hypertension and Nutrition, Geneva University Hospitals, Geneva, Switzerland
| | | |
Collapse
|
147
|
Rubinchik-Stern M, Shmuel M, Bar J, Kovo M, Eyal S. Adverse placental effects of valproic acid: Studies in perfused human placentas. Epilepsia 2018; 59:993-1003. [PMID: 29667177 DOI: 10.1111/epi.14078] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE In utero exposure to valproic acid (VPA) has been associated with worse pregnancy outcomes compared to all other antiepileptic drugs. We have previously shown that VPA alters the expression of placental transporters for hormones and nutrients in vitro and in pregnant mice. Here, our aim was to characterize the effects of short exposure to VPA on the expression of carriers for compounds essential for fetal development in human placentas ex vivo, under controlled conditions. METHODS Placentas were obtained from cesarean deliveries of women with no known epilepsy. Cotyledons were cannulated and perfused in the absence or the presence of VPA (42, 83, or 166 μg/mL; n = 6/group) in the maternal perfusate over 180 minutes. A customized gene panel array was used to analyze the expression of carrier genes in the perfused cotyledons. We additionally measured in the perfused placentas folic acid concentrations and histone acetylation. RESULTS VPA significantly altered the mRNA levels of major carriers for folic acid, glucose, choline, thyroid hormones, and serotonin (P < .05) and reduced placental folate concentrations by 25%-35% (P = .059). The effects were observed at therapeutic concentrations sufficient to enhance placental histone acetylation, and some were concentration-dependent. SIGNIFICANCE Our results point to the placenta as a novel target of VPA, implying potential involvement of the placenta in VPA's adverse fetal outcomes.
Collapse
Affiliation(s)
- Miriam Rubinchik-Stern
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miriam Shmuel
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jacob Bar
- Department of Obstetrics & Gynecology, Edith Wolfson Medical Center, Holon, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Kovo
- Department of Obstetrics & Gynecology, Edith Wolfson Medical Center, Holon, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Eyal
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
148
|
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: 2.8] [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
|
149
|
Circular RNA expression profiles in placental villi from women with gestational diabetes mellitus. Biochem Biophys Res Commun 2018. [DOI: 10.1016/j.bbrc.2018.03.051] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
150
|
Kubo-Kaneda M, Tanaka H, Maki S, Nii M, Umekawa T, Osato K, Kamimoto Y, Kondo E, Ikeda T. Placental growth factor as a predictor of the efficacy of tadalafil treatment for fetal growth restriction. J Matern Fetal Neonatal Med 2018; 32:2879-2882. [PMID: 29580120 DOI: 10.1080/14767058.2018.1450863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Purpose: We recently demonstrated the efficacy of tadalafil treatment for fetal growth restriction (FGR). This study aimed to evaluate the utility of serum placental growth factor (PlGF) level for predicting the efficacy of tadalafil for the treatment of FGR. Materials and methods: The correlations between serum level of PlGF and fetal growth velocity were retrospectively assessed in nine pregnant women receiving tadalafil for FGR before 30 weeks' gestation. Results: Median gestational age was 26 weeks (range 26-28 weeks), and median deviation of estimated fetal weight from standard weight was -2.1 standard deviations (SD) (-2.2 to -1.9 SD) at the beginning of tadalafil treatment. The median serum PlGF level was 227 pg/ml (40.2-427.0 pg/ml) before tadalafil treatment and 278 pg/ml (66.2-729.5 pg/ml) more than 2 weeks after initiation of tadalafil treatment (median gestational week at measurement of PlGF after treatment, 33 weeks [28-33 weeks]). The median fetal growth velocity from enrollment to birth was 17.5 g/day (12.1-20.3 g/day). Maternal serum PlGF levels were increased after tadalafil treatment in all nine cases (median increase in PlGF, 73.1 pg/ml [26.0-281.5 pg/ml]). Notably, maternal serum PlGF level before tadalafil treatment significantly correlated with fetal growth velocity (R2 = 0.63, p < .01). Conclusions: Tadalafil treatment may increase maternal serum PlGF levels. Our results suggest that maternal serum PlGF levels can be used as a predictor of the efficacy of tadalafil treatment for FGR.
Collapse
Affiliation(s)
- Michiko Kubo-Kaneda
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Hiroaki Tanaka
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Shintaro Maki
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Masafumi Nii
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Takashi Umekawa
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Kazuhiro Osato
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Yuki Kamimoto
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Eiji Kondo
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| | - Tomoaki Ikeda
- a Department of Obstetrics and Gynecology , Mie University School of Medicine , Tsu , Japan
| |
Collapse
|