1
|
Zhang H, Zhang Y, Xu K, Wang L, Zhou X, Yang M, Xie J, Li H. Inhibition of FLT1 Attenuates Neurodevelopmental Abnormalities and Cognitive Impairment in Offspring Caused by Maternal Prenatal Stress. Appl Biochem Biotechnol 2024; 196:4900-4913. [PMID: 37979086 DOI: 10.1007/s12010-023-04774-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
Fms-like tyrosine kinase 1 (FLT1) has been shown to regulate processes such as angiogenesis, neurogenesis, and cognitive impairment. However, the role of FLT1 in prenatal stress (PS) is unclear. The purpose of this study was to investigate the role of FLT1 in PS mothers and their offspring. Wire mesh restrainers were used to construct PS rat model. The levels of FLT1, IL-1β, IL-6, and ROS in clinical samples and rat samples were detected by qRT-PCR, ELisa kit, and DCFH-DA fluorescence kit. Morris water maze assay and forced swimming assay were used to test the cognitive function of offspring young rats. The apoptosis level of hippocampal neurons and the expression of NMDARs were detected by MTT assay, TUNEL assay, and Western blot. The results showed that FLT1 was upregulated in PS mothers and positively correlated with PS degree. The level of FLT1 was elevated in PS model rats. Knockdown of FLT1 reduced maternal ROS and MDA levels and increased SOD levels in PS rats. Knockdown of FLT1 also reduced the secretion of IL-1β, IL-6, and cortisol in PS rats. Inhibition of FTL1 alleviated cognitive impairment in PS offspring pups. Inhibition of FTL1 reduced hippocampal neuronal apoptosis and increased the expression of NMDARs in PS progeny. In conclusions, we demonstrated that knockdown of FLT1 inhibits maternal oxidative stress, inflammation, and cortisol secretion in PS rats. In addition, knockdown of FLT1 also alleviated cognitive dysfunction and neurodevelopmental abnormalities in PS offspring pups.
Collapse
Affiliation(s)
- Huifang Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Division of Neonatology, NO.277, West Yanta Road, Xi'an, 710061, Shaanxi, China
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Yudan Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Division of Neonatology, NO.277, West Yanta Road, Xi'an, 710061, Shaanxi, China
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Kaixuan Xu
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Lawen Wang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Xin Zhou
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Mingge Yang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Jiangli Xie
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Hui Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Division of Neonatology, NO.277, West Yanta Road, Xi'an, 710061, Shaanxi, China.
- Division of Neonatology, The Affiliated Children Hospital of Xi'an Jiaotong University, Shaanxi, 710003, China.
| |
Collapse
|
2
|
Kashiwagi H, Mariya T, Umemoto M, Ogawa S, Hirohashi Y, Fujibe Y, Kubo T, Someya M, Baba T, Ishioka S, Torigoe T, Saito T. Pregnancy-specific beta-1-glycoprotein 6 is a potential novel diagnostic biomarker of placenta accreta spectrum. Med Mol Morphol 2024; 57:35-44. [PMID: 37831187 DOI: 10.1007/s00795-023-00371-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023]
Abstract
Early diagnosis is essential for the safer perinatal management of placenta accreta spectrum (PAS). We used transcriptome analysis to investigate diagnostic maternal serum biomarkers and the mechanisms of PAS development. We analyzed eight formalin-fixed paraffin-embedded placental specimens from two placenta increta and three placenta percreta cases who underwent cesarean hysterectomy at Sapporo Medical University Hospital between 2013 and 2019. Invaded placental regions were isolated from the uterine myometrium and RNA was extracted. The transcriptome difference between normal placenta and PAS was analyzed by microarray analysis. The PAS group showed markedly decreased expression of placenta-specific genes such as LGALS13 and the pregnancy-specific beta-1-glycoprotein (PSG) family. Term enrichment analysis revealed changes in genes related to cellular protein catabolic process, female pregnancy, autophagy, and metabolism of lipids. From the highly dysregulated genes in the PAS group, we investigated the expression of PSG family members, which are secreted into the intervillous space and can be detected in maternal serum from the early stage of pregnancy. The gene expression level of PSG6 in particular was progressively decreased from placenta increta to percreta. The PSG family, especially PSG6, is a potential biomarker for PAS diagnosis.
Collapse
Affiliation(s)
- Hazuki Kashiwagi
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Tasuku Mariya
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan.
| | - Mina Umemoto
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Shiori Ogawa
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology 1st, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuya Fujibe
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Terufumi Kubo
- Department of Pathology 1st, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masayuki Someya
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Tsuyoshi Baba
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Shinichi Ishioka
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Toshihiko Torigoe
- Department of Pathology 1st, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, South 1 West 17, Chuo-ku, Sapporo, 060-8556, Japan
| |
Collapse
|
3
|
Rezaei M, Ghasemi M, Saravani M, Ghahghayi F, Shahraki-Ghadim H, Salimi S. The possible effects of the MTOR polymorphisms on preeclampsia susceptibility, severity, and onset: a case-control study and in silico analysis. Mol Biol Rep 2024; 51:335. [PMID: 38393518 DOI: 10.1007/s11033-023-09190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 12/21/2023] [Indexed: 02/25/2024]
Abstract
BACKGROUND Preeclampsia (PE) is a gestational complication with developed hypertension and proteinuria. Evidence showed the role of mTOR in various cellular processes. Therefore, this study aimed to evaluate the effects of MTOR polymorphisms on susceptibility, severity, and onset of Preeclampsia (PE). METHODS AND RESULTS A total of 250 PE pregnant women and 258 age-matched control subjects were recruited in this study. To genotype MTOR polymorphisms, the PCR-RFLP method was used. The SpliceAid 2 and PROMO tools were used for in silico analysis. The maternal MTOR rs17036508T/C polymorphism was associated with PE risk in various genetic models. There was no relationship between rs2536T/C and rs2295080T/G polymorphisms and PE. The TTC and TGC haplotypes of rs2536/ rs2295080/ rs17036508 polymorphisms were significantly higher in PE women. Subgroup analysis revealed the association between the MTOR rs2295080 variant and an increased risk of Early-onset PE (EOPE). However, the MTOR rs17036508 was associated with a higher risk of EOPE and Late- Onset PE. In addition, the MTOR rs2295080 could increase the risk of severe PE. The results of the in silico analysis showed that rs17036508 disrupted several binding motifs in the mutant sequence. The PROMO database revealed that the T to C substitution leads to the loss of the TFII-I binding site in the mutant allele. CONCLUSION The MTOR rs17036508T/C polymorphism was associated with PE risk. There was an association between the MTOR rs2295080 variant and an increased risk of EOPE. The MTOR rs17036508T/C and rs2295080T/C variants could disrupt several binding motifs and TFII-I binding respectively.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Marzieh Ghasemi
- Department of Obstetrics and Gynecology, Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohsen Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fatemeh Ghahghayi
- Department of Obstetrics and Gynecology, Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hossein Shahraki-Ghadim
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeedeh Salimi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Pintye D, Sziva RE, Mastyugin M, Young BC, Jacas S, Török M, Salahuddin S, Jagtap P, Southan GJ, Zsengellér ZK. A Novel Dual-Function Redox Modulator Relieves Oxidative Stress and Anti-Angiogenic Response in Placental Villus Explant Exposed to Hypoxia-Relevance for Preeclampsia Therapy. BIOLOGY 2023; 12:1229. [PMID: 37759628 PMCID: PMC10525611 DOI: 10.3390/biology12091229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Preeclampsia (PE) is a severe, life-threatening complication during pregnancy (~5-7%), and no causative treatment is available. Early aberrant spiral artery remodeling is associated with placental stress and the release of oxygen radicals and other reactive oxygen species (ROS) in the placenta. This precedes the production of anti-angiogenic factors, which ultimately leads to endothelial and trophoblast damage and the key features of PE. We tested whether a novel dual-function redox modulator-AKT-1005-can effectively reduce placental oxidative stress and alleviate PE symptoms in vitro. METHOD Isolated human villous explants were exposed to hypoxia and assessed to determine whether improving cell-redox function with AKT-1005 diminished ROS production, mitochondrial stress, production of the transcription factor HIF1A, and downstream anti-angiogenic responses (i.e., sFLT1, sEng production). MitoTEMPO was used as a reference antioxidant. RESULTS In our villous explant assays, pretreatment with AKT-1005 reduced mitochondrial-derived ROS production, reduced HIF-1A, sFLT1, and sEng protein expression, while increasing VEGF in hypoxia-exposed villous trophoblast cells, with better efficiency than MitoTEMPO. In addition, AKT-1005 improved mitochondrial electron chain enzyme activity in the stressed explant culture. CONCLUSIONS The redox modulator AKT-1005 has the potential to intervene with oxidative stress and can be efficacious for PE therapy. Future studies are underway to assess the in vivo efficacy of HMP.
Collapse
Affiliation(s)
- Diana Pintye
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
| | - Réka E. Sziva
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary
| | - Maxim Mastyugin
- Department of Chemistry, University of Massachusetts, Boston, MA 02125, USA; (M.M.); (M.T.)
| | - Brett C. Young
- Department of Obstetrics and Gynecology, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (B.C.Y.); (S.S.)
| | - Sonako Jacas
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
| | - Marianna Török
- Department of Chemistry, University of Massachusetts, Boston, MA 02125, USA; (M.M.); (M.T.)
| | - Saira Salahuddin
- Department of Obstetrics and Gynecology, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (B.C.Y.); (S.S.)
| | | | | | - Zsuzsanna K. Zsengellér
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
| |
Collapse
|
5
|
Karpova NS, Dmitrenko OP, Budykina TS. Literature Review: The sFlt1/PlGF Ratio and Pregestational Maternal Comorbidities: New Risk Factors to Predict Pre-Eclampsia. Int J Mol Sci 2023; 24:ijms24076744. [PMID: 37047717 PMCID: PMC10095124 DOI: 10.3390/ijms24076744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
One of the main causes of maternal and neonatal morbidity and mortality is pre-eclampsia. It is characterized by a high sFlt1/PlGF ratio, according to prior research. Pregestational diseases in mothers may increase the risk of developing pre-eclampsia. Only a few studies have looked at the connection between maternal comorbidities before conception and the sFlt1/PlGF ratio. The most recent information regarding the association between maternal pregestational diseases and the ratio of sFlt1/PlGF is described in this review. The paper also examines current research suggesting that changes in pregnancy hormones and metabolites are related to a high sFlt1/PlGF ratio. Certain maternal disorders have been found to dramatically raise sFlt-1 and sFlt1/PlGF levels, according to an analysis of the literature. There is still debate about the data on the association between the sFlt1/PlGF ratio and maternal disorders such as HIV, acute coronary syndromes, cardiovascular function in the mother between 19 and 23 weeks of pregnancy, thyroid hormones, diabetes, and cancer. Additional research is needed to confirm these findings.
Collapse
Affiliation(s)
- Nataliia Sergeevna Karpova
- Federal State Budgetary Institution “Research Institute of Pathology and Pathophysiology”, St. Baltiyskaya, House 8, Moscow 125315, Russia
| | - Olga Pavlovna Dmitrenko
- Federal State Budgetary Institution “Research Institute of Pathology and Pathophysiology”, St. Baltiyskaya, House 8, Moscow 125315, Russia
| | - Tatyana Sergeevna Budykina
- State Budgetary Health Institution of the Moscow Region “Moscow Regional Research Institute of Obstetrics and Gynecology”, St. Pokrovka, d.22a, Moscow 101000, Russia
| |
Collapse
|
6
|
Aberdeen GW, Babischkin JS, Lindner JR, Pepe GJ, Albrecht ED. Placental sFlt-1 Gene Delivery in Early Primate Pregnancy Suppresses Uterine Spiral Artery Remodeling. Endocrinology 2022; 163:bqac012. [PMID: 35134145 PMCID: PMC8896163 DOI: 10.1210/endocr/bqac012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Indexed: 02/04/2023]
Abstract
Uterine spiral artery remodeling (SAR) is essential for promoting placental perfusion and fetal development. A defect in SAR results in placental ischemia and increase in placental expression and serum levels of the soluble fms-like tyrosine kinase-1 (sFlt-1) receptor that binds to and suppresses vascular endothelial growth factor (VEGF) bioavailability, thereby leading to maternal vascular dysfunction. We have established a nonhuman primate model of impaired SAR and maternal vascular dysfunction by prematurely elevating estradiol levels in early baboon pregnancy. However, it is unknown whether this primate model of defective SAR involves an increase in placental expression of sFlt-1, which may suppress VEGF bioavailability and thus SAR in the first trimester. Therefore, to establish the role of sFlt-1 in early pregnancy, SAR was quantified in baboons treated on days 25 through 59 of gestation (term = 184 days) with estradiol or with the sFlt-1 gene targeted selectively to the placental basal plate by ultrasound-mediated/microbubble-facilitated gene delivery technology. Placental basal plate sFlt-1 protein expression was 2-fold higher (P < 0.038) and the level of SAR for vessels > 25 µm in diameter was 72% and 63% lower (P < 0.01), respectively, in estradiol-treated and sFlt-1 gene-treated baboons than in untreated animals. In summary, prematurely elevating estradiol levels or sFlt-1 gene delivery increased placental basal plate sFlt-1 protein expression and suppressed SAR in early baboon pregnancy. This study makes the novel discovery that in elevated levels sFlt-1 has a role both in suppressing SAR in early primate pregnancy and maternal vascular endothelial function in late gestation.
Collapse
Affiliation(s)
- Graham W Aberdeen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffery S Babischkin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Eugene D Albrecht
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
7
|
Placental lesions and differential expression of pro-and anti-angiogenic growth mediators and oxidative DNA damage marker in placentae of Ghanaian suboptimal and optimal health status pregnant women who later developed preeclampsia. PLoS One 2022; 17:e0265717. [PMID: 35312727 PMCID: PMC8936490 DOI: 10.1371/journal.pone.0265717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 03/07/2022] [Indexed: 11/18/2022] Open
Abstract
Background Angiogenic growth mediators (AGMs) and oxidative stress (OS) both play essential roles in normal placental vascular development and as such, placental alterations in these factors contribute to pre-eclampsia (PE). Suboptimal health status (SHS), an intermediate between health and disease, has been associated with imbalanced AGMs and OS biomarkers. Thus, SHS pregnant women may be at increased risk of developing PE and may present abnormal placental alteration and expression of AGMs and OS compared to optimal health status (OHS) pregnant women. We examined the histopathological morphology, immunohistochemical expression of AGMs antibodies and oxidative DNA damage marker in the placentae of SHS and OHS pregnant women who developed early-onset PE (EO-PE) and late-onset (LO-PE) compared to normotensive pregnancy (NTN-P). Methods This nested case-control study recruited 593 singleton normotensive pregnant women at baseline (10–20 weeks gestation) from the Ghanaian Suboptimal Health Status Cohort Study (GHOACS) undertaken at the Komfo Anokye Teaching Hospital, Ghana. Socio-demographic, clinical and obstetrics data were collected, and a validated SHS questionnaire-25 (SHSQ-25) was used in classifying participants into SHS (n = 297) and OHS (n = 296). Participants were followed until the time of PE diagnosis and delivery (32–42 weeks gestation). Blood samples were collected at the two-time points and were assayed for AGMs; soluble fms-like tyrosine kinase-1 (sFlt-1), placental growth factor (PIGF), vascular endothelial growth factor-A (VEGF-A), and soluble endoglin (sEng), and OS biomarkers; 8-hydroxydeoxyguanosine (8-OHdG), 8-epiprostaglandinF2-alpha (8- epi-PGF2α) and total antioxidant capacity (TAC) using ELISA. Placental samples were collected for histopathological and immunohistochemical analysis. Results Of the 593 pregnant women, 498 comprising 248 SHS and 250 OHS women returned for delivery and were included in the final analysis. Of the 248 SHS women, 56, 97 and 95 developed EO-PE, LO-PE and NTN-P, respectively, whereas 14, 30 and 206 of the 250 OHS mothers developed EO-PE, LO-PE and NTN-P, respectively. At baseline, SHS_NTN pregnant women had a significant imbalance in AGMs and OS biomarkers compared to OHS_NTN pregnant women (p<0.0001). At the time of PE diagnosis, SHS_NTN-P women who developed EO-PE, LO-PE, and NTN-P had lower serum levels of P1GF, VEGF-A and TAC and correspondingly higher levels of sEng, sFlt-1, 8-epiPGF2α, and 8-OHdG than OHS-NTN-P women who developed EO-PE and LO-PE, NTN-P (p<0.0001). A reduced placental size, increased foetal/placental weight ratio, and a significantly higher proportion of fibrinoid necrosis, infarction, villous fibrin, syncytial knots, calcification, chorangiosis, tunica media/vascular wall hypertrophy and chorioamnionitis was associated with the SHS group who developed PE (EO-PE>LO-PE) more than OHS groups who developed PE (EO-PE>LO-PE) when all were compared to NTN-P (p<0.0001). The intensity of antibody expression of PIGF and VEGF-A were significantly reduced, whereas Flt-1, Eng and 8-OHdG were significantly increased in placentae from SHS-pregnant women who developed EO-PE>LO-PE more than OHS- pregnant women who developed EO-PE>LO-PE when all were compared to NTN-P (p<0.0001). Conclusion Increased lesions, oxidative DNA damage, and imbalanced expression between pro-and anti-AGMs are associated more with SHS-embodied PE placentae rather than OHS-embodied PE subtypes, thus potentially allowing differential evaluation of PE.
Collapse
|
8
|
Pitz Jacobsen D, Fjeldstad HE, Johnsen GM, Fosheim IK, Moe K, Alnæs-Katjavivi P, Dechend R, Sugulle M, Staff AC. Acute Atherosis Lesions at the Fetal-Maternal Border: Current Knowledge and Implications for Maternal Cardiovascular Health. Front Immunol 2021; 12:791606. [PMID: 34970270 PMCID: PMC8712939 DOI: 10.3389/fimmu.2021.791606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
Decidua basalis, the endometrium of pregnancy, is an important interface between maternal and fetal tissues, made up of both maternal and fetal cells. Acute atherosis is a uteroplacental spiral artery lesion. These patchy arterial wall lesions containing foam cells are predominantly found in the decidua basalis, at the tips of the maternal arteries, where they feed into the placental intervillous space. Acute atherosis is prevalent in preeclampsia and other obstetric syndromes such as fetal growth restriction. Causal factors and effects of acute atherosis remain uncertain. This is in part because decidua basalis is challenging to sample systematically and in large amounts following delivery. We summarize our decidua basalis vacuum suction method, which facilitates tissue-based studies of acute atherosis. We also describe our evidence-based research definition of acute atherosis. Here, we comprehensively review the existing literature on acute atherosis, its underlying mechanisms and possible short- and long-term effects. We propose that multiple pathways leading to decidual vascular inflammation may promote acute atherosis formation, with or without poor spiral artery remodeling and/or preeclampsia. These include maternal alloreactivity, ischemia-reperfusion injury, preexisting systemic inflammation, and microbial infection. The concept of acute atherosis as an inflammatory lesion is not novel. The lesions themselves have an inflammatory phenotype and resemble other arterial lesions of more extensively studied etiology. We discuss findings of concurrently dysregulated proteins involved in immune regulation and cardiovascular function in women with acute atherosis. We also propose a novel hypothesis linking cellular fetal microchimerism, which is prevalent in women with preeclampsia, with acute atherosis in pregnancy and future cardiovascular and neurovascular disease. Finally, women with a history of preeclampsia have an increased risk of premature cardiovascular disease. We review whether presence of acute atherosis may identify women at especially high risk for premature cardiovascular disease.
Collapse
Affiliation(s)
| | - Heidi Elisabeth Fjeldstad
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Guro Mørk Johnsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Ingrid Knutsdotter Fosheim
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjartan Moe
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Obstetrics and Gynaecology, Bærum Hospital, Vestre Viken HF, Bærum, Norway
| | | | - Ralf Dechend
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max-Delbruck Center for Molecular Medicine, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS-Klinikum, Berlin, Germany
| | - Meryam Sugulle
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne Cathrine Staff
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Genest DS, Dal Soglio D, Girard S, Rey E. Association between proteinuria and placental pathology in preeclampsia: A retrospective study. SAGE Open Med 2021; 9:20503121211058053. [PMID: 34925835 PMCID: PMC8673865 DOI: 10.1177/20503121211058053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction: Preeclampsia develops due to placental insufficiency and systemic proinflammatory and antiangiogenic mediator release, with ensuing systemic endothelial dysfunction. Nephrotic-range proteinuria appears to be associated with worse pregnancy outcomes. The relationship between differing degrees of proteinuria and the severity of placental alterations has not been studied. Methods: This is a single-centre retrospective comparison of 150 singleton pregnancies complicated by preeclampsia and varying degrees of proteinuria. Maternal demographic, obstetrical and fetal outcome data were obtained from chart review. The placental histologic evaluations were performed by a placental pathologist blinded to all other clinical information. Results: Preeclamptic women with massive proteinuria had evidence of more severe maternal vascular malperfusion lesions. The severity of the lesions was progressive through mild, moderate and massive proteinuria. Women with massive proteinuria had a higher incidence of renal dysfunction and severe hypertension, and had earlier preterm deliveries compared to preeclamptic women with mild and moderate proteinuria (p < 0.05). Conclusion: Preeclampsia with more severe proteinuria is associated with a higher prevalence of placental maternal vascular malperfusion.
Collapse
Affiliation(s)
| | - Dorothée Dal Soglio
- Department of Pathology and Clinical Laboratory Medicine, CHU Sainte-Justine, Montreal, QC, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Université de Montréal, Centre de Recherche, CHU Sainte-Justine, Montreal, QC, Canada
| | - Evelyne Rey
- Department of Obstetrics and Gynecology, Université de Montréal, Centre de Recherche, CHU Sainte-Justine, Montreal, QC, Canada
- Division of Obstetric Medicine, Department of Medicine, University of Montreal, CHU Sainte-Justine, Montreal, QC, Canada
| |
Collapse
|
10
|
Deer E, Jones J, Cornelius DC, Comley K, Herrock O, Campbell N, Fitzgerald S, Ibrahim T, LaMarca B, Amaral LM. Progesterone Induced Blocking Factor Reduces Hypertension and Placental Mitochondrial Dysfunction in Response to sFlt-1 during Pregnancy. Cells 2021; 10:2817. [PMID: 34831040 PMCID: PMC8616090 DOI: 10.3390/cells10112817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia (PE) is characterized by new onset hypertension in association with placental ischemia, reduced fetal weight, elevated soluble fms-like tyrosine kinase-1 (sFlt-1), and placental mitochondrial (mt) dysfunction and oxidative stress (ROS). Progesterone induced blocking factor (PIBF) is a product of progesterone signaling that blocks inflammatory processes and we have previously shown PIBF to lower mean arterial blood pressure (MAP) and sFlt-1 in a rat model of PE. Infusion of sFlt-1 causes hypertension and many characteristics of PE in pregnant rodents, however, its role in causing mt dysfunction is unknown. Therefore, we hypothesize that PIBF will improve mt function and MAP in response to elevated sFlt-1 during pregnancy. We tested our hypothesis by infusing sFlt-1 via miniosmotic pumps in normal pregnant (NP) Sprague-Dawley rats (3.7 μg·kg-1·day-1) on gestation days (GD) 13-19 in the presence or absence of PIBF (2.0 µg/mL) injected intraperitoneally on GD 15 and examined mean arterial blood pressure (MAP) and placental mt ROS on GD 19. sFlt-1 increased MAP to 112 + 2 (n = 11) compared to NP rats (98 + 2 mmHg, n = 15, p < 0.05), which was lowered in the presence of sFlt-1 (100 + 1 mmHg, n = 5, p < 0.05). Placental mtATP was reduced in sFlt-1 infused rats versus NP controls, but was improved with PIBF. Placental mtROS was elevated with sFlt-1 compared to NP controls, but was reduced with PIBF. Sera from NP + sFlt-1 increased endothelial cell mtROS, which was attenuated with PIBF. These data demonstrate sFlt-1 induced HTN during pregnancy reduces placental mt function. Importantly, PIBF improved placental mt function and HTN, indicating the efficacy of improved progesterone signaling as potential therapeutics for PE.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Jalisa Jones
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Denise C. Cornelius
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS 39126, USA;
| | - Kyleigh Comley
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Owen Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lorena M. Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| |
Collapse
|
11
|
Chappell LC, Cluver CA, Kingdom J, Tong S. Pre-eclampsia. Lancet 2021; 398:341-354. [PMID: 34051884 DOI: 10.1016/s0140-6736(20)32335-7] [Citation(s) in RCA: 413] [Impact Index Per Article: 137.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/20/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022]
Abstract
Pre-eclampsia is a multisystem pregnancy disorder characterised by variable degrees of placental malperfusion, with release of soluble factors into the circulation. These factors cause maternal vascular endothelial injury, which leads to hypertension and multi-organ injury. The placental disease can cause fetal growth restriction and stillbirth. Pre-eclampsia is a major cause of maternal and perinatal mortality and morbidity, especially in low-income and middle-income countries. Prophylactic low-dose aspirin can reduce the risk of preterm pre-eclampsia, but once pre-eclampsia has been diagnosed there are no curative treatments except for delivery, and no drugs have been shown to influence disease progression. Timing of delivery is planned to optimise fetal and maternal outcomes. Clinical trials have reported diagnostic and prognostic strategies that could improve fetal and maternal outcomes and have evaluated the optimal timing of birth in women with late preterm pre-eclampsia. Ongoing studies are evaluating the efficacy, dose, and timing of aspirin and calcium to prevent pre-eclampsia and are evaluating other drugs to control hypertension or ameliorate disease progression.
Collapse
Affiliation(s)
- Lucy C Chappell
- Department of Women and Children's Health, School of Life Course Sciences, Kings' College London, London, UK.
| | - Catherine A Cluver
- Department of Obstetrics and Gynaecology, Stellenbosch University, Stellenbosch, South Africa; Tygerberg Hospital, Cape Town, South Africa
| | - John Kingdom
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Stephen Tong
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia; Mercy Hospital for Women, Heidelberg, VIC, Australia
| |
Collapse
|
12
|
Fillion A, Guerby P, Menzies D, Bujold E. The association between maternal placental growth factor and placental maternal vascular malperfusion lesions. J Gynecol Obstet Hum Reprod 2021; 50:102179. [PMID: 34107359 DOI: 10.1016/j.jogoh.2021.102179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/22/2021] [Accepted: 06/02/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Placental Growth Factor (PlGF) is used for the prediction of preeclampsia (PE), a syndrome associated with maternal vascular malperfusion (MVM). Our goal is to determine the correlation between PlGF and MVM. MATERIAL AND METHODS We performed a secondary analysis of the PEARL study that included nulliparous women with PE (cases), and low-risk nulliparous women recruited in early pregnancy (controls). All participants provided blood samples at diagnosis of PE (cases), or between 26 and 34 weeks (controls) for measurement of PlGF (B·R·A·H·M·S plus KRYPTOR automated assays), that was transformed into multiple of median (MoM). Placental examination was performed for the diagnosis of MVM based on the Amsterdam Placental Workshop Group Consensus Statement. Nonparametric tests and receiver operating characteristic (ROC) curves were used to compare PlGF in pregnancies with, and without PE, stratified by the presence of MVM. RESULTS Third trimester PlGF was lower in PE cases with MVM (N = 20; median: 0.04 MoM; interquartile: 0.03-0.09; p<0.0001), and in controls with MVM (N = 4; 0.30MoM; 0.07-0.52; p = 0.002) compared to controls without MVM (N = 29; 0.99 MoM; 0.67-1.52). PlGF in PE cases without MVM (N = 5; 0.18 MoM; 0.17-1.64) was not significantly different than in controls without MVM but the sample size was small. ROC curve demonstrated a greater predictability of PlGF for PE with MVM than PE without MVM (AUC: 0.99 vs. 0.38; p<0.0001). DISCUSSION Third trimester PlGF is a better predictor of PE associated with MVM than a predictor of PE without MVM. We hypothesize that PlGF is a stronger marker of MVM than PE.
Collapse
Affiliation(s)
- Alexandre Fillion
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, Quebec City, Canada
| | - Paul Guerby
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, Quebec City, Canada; Department of Obstetrics and Gynecology, Paule de Viguier Hospital, CHU de Toulouse, Toulouse, Inserm U-1048, Université de Toulouse, France
| | - Didier Menzies
- Department of Fetopathology CHRU de Nancy, Nancy, France
| | - Emmanuel Bujold
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, Quebec City, Canada; Department of Obstetrics and Gynecology, Faculty of Medicine, CHU de Québec-Université Laval, Quebec City, Canada.
| |
Collapse
|
13
|
Johnson KM, Smith L, Modest AM, Salahuddin S, Karumanchi SA, Rana S, Young BC. Angiogenic factors and prediction for ischemic placental disease in future pregnancies. Pregnancy Hypertens 2021; 25:12-17. [PMID: 34020330 DOI: 10.1016/j.preghy.2021.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Ischemic placental disease (IPD), including preeclampsia, abruption, and fetal growth restriction, often recurs in subsequent pregnancies. Angiogenic factors of placental origin have been implicated in the pathogenesis of preeclampsia, but have not been studied as predictors of IPD in subsequent pregnancies. We hypothesized that elevated angiogenic factors in an index pregnancy would be associated with recurrence of IPD. STUDY DESIGN We conducted a retrospective cohort study of patients undergoing evaluation for preeclampsia who had angiogenic factors measured in an index pregnancy and experienced a subsequent pregnancy at the same institution. Patients with IPD in the index pregnancy were included. A high ratio of soluble fms-like tyrosine kinase 1 (sFlt1) and placental growth factor (PlGF) was defined as greater than or equal to 85. MAIN OUTCOME MEASURES The primary outcome was IPD in a subsequent pregnancy. RESULTS We included 109 patients in the analysis. The sFlt1/PlGF ratio was elevated in 30% of participants. Those with an elevated ratio were more likely to be nulliparous in the index pregnancy, and less likely to have chronic hypertension. The recurrence of IPD in the study was 27%, with a non-significant difference in risk based on a high sFlt-1/P1GF ratio RR 0.58 (95% CI 0.21 - 1.6) compared to a low ratio. CONCLUSIONS A high sFlt1/P1GF ratio in an index pregnancy is not associated with a higher risk of IPD in a subsequent pregnancy. These data suggest placental angiogenic biomarkers are specific to the pregnancy and not a reflection of maternal predisposition to IPD.
Collapse
Affiliation(s)
- Katherine M Johnson
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
| | - Laura Smith
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Anna M Modest
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Saira Salahuddin
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center/Harvard Medical School, 99 Brookline Avenue, RN 359, Boston, MA 02215, USA
| | - S A Karumanchi
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center/Harvard Medical School, 99 Brookline Avenue, RN 359, Boston, MA 02215, USA; Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Sarosh Rana
- Department of Obstetrics and Gynecology, University of Chicago, 5741 S. Maryland Ave., Chicago, IL 60637, USA
| | - Brett C Young
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| |
Collapse
|
14
|
Cottrell JN, Witcher AC, Comley K, Cunningham MW, Ibrahim T, Cornelius DC, LaMarca B, Amaral LM. Progesterone-induced blocking factor improves blood pressure, inflammation, and pup weight in response to reduced uterine perfusion pressure (RUPP). Am J Physiol Regul Integr Comp Physiol 2021; 320:R719-R727. [PMID: 33533305 DOI: 10.1152/ajpregu.00152.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Preeclampsia (PE) is characterized by new-onset hypertension in association with elevated natural killer (NK) cells and inflammatory cytokines, which are likely culprits for decreased fetal weight during PE pregnancies. As progesterone increases during normal pregnancy, it stimulates progesterone-induced blocking factor (PIBF). PIBF has been shown to decrease inflammation and cytolytic NK cells, both of which are increased during PE. We hypothesized that PIBF reduces inflammation as a mechanism to improve hypertension in the preclinical reduced uterine perfusion pressure (RUPP) rat model of PE. PIBF (2.0 µg/mL) was administered intraperitoneally on gestational day 15 to either RUPP or normal pregnant (NP) rats. On day 18, carotid catheters were inserted. Mean arterial blood pressure (MAP) and samples were collected on day 19. MAP in NP rats (n = 11) was 100 ± 2 mmHg and 105 ± 3 mmHg in NP + PIBF rats (n = 8) and 122 ± 1 mmHg in RUPP rats (n = 10), which improved to 110 ± 2 mmHg in RUPP + PIBF rats (n = 11), P < 0.05. Pup weight was 2.4 ± 0.1 g in NP, 2.5 ± 0.1 g in NP + PIBF, 1.9 ± 0.1 g in RUPP, and improved to 2.1 ± 0.1 g in RUPP + PIBF rats. Circulating and placental cytolytic NK cells, IL-17, and IL-6 were significantly reduced while IL-4 and T helper (TH) 2 cells were significantly increased in RUPP rats after PIBF administration. Importantly, vasoactive pathways preproendothelin-1, nitric oxide, and soluble fms-Like tyrosine Kinase-1 (sFlt-1) were normalized in RUPP + PIBF rats compared with RUPP rats, P < 0.05. Our findings suggest that PIBF normalized IL-4/TH2 cells, which was associated with improved inflammation, fetal growth restriction, and blood pressure in the RUPP rat model of PE.
Collapse
Affiliation(s)
- Jesse N Cottrell
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alexis C Witcher
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kyleigh Comley
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mark W Cunningham
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tarek Ibrahim
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Babbette LaMarca
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorena M Amaral
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
15
|
Nuzzo AM, Giuffrida D, Moretti L, Re P, Grassi G, Menato G, Rolfo A. Placental and maternal sFlt1/PlGF expression in gestational diabetes mellitus. Sci Rep 2021; 11:2312. [PMID: 33504861 PMCID: PMC7840991 DOI: 10.1038/s41598-021-81785-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Gestational diabetes mellitus (GDM) and preeclampsia (PE) are both characterized by endothelial dysfunction and GDM women have higher incidence of PE. The placenta plays a key role in PE pathogenesis but its contribution to PE during GDM remains unclear. Herein, we compared placental and maternal blood anti-angiogenic soluble fms-like tyrosine kinase-1 (sFlt1) and pro-angiogenic Placental Growth Factor (PlGF) expressions in GDM and GDM-PE pregnancies compared to controls (CTRL) and PE cases. Electrochemiluminescence immunoassays showed a significantly higher maternal blood sFlt1/PlGF values in GDM-PE relative to CTRL and GDM pregnancies. We reported that placental PlGF gene expression was significantly decreased in GDM, PE and GDM-PE relative to CTRL. However, PlGF protein levels were significantly increased in GDM and GDM-PE relative to CTRL and PE placentae. Finally, sFlt1 gene expression was significantly increased in PE relative to CTRL, GDM and GDM-PE placentae. In contrast, sFlt1 protein expression was significantly decreased in GDM-PE relative to CTRL, GDM and PE placentae. Finally, higher sFlt1/PlGF ratio in GDM-PE maternal blood suggest that sFlt1 overproduction is related to PE onset also in GDM pregnancies even though characterized by a less severe endothelial dysfunction in terms of angiogenic biomarkers.
Collapse
Affiliation(s)
- Anna Maria Nuzzo
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Domenica Giuffrida
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Laura Moretti
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Paola Re
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Giorgio Grassi
- Department of Endocrinology, Diabetology, and Metabolism, Città della Salute e della Scienza University Hospital, Corso Bramante 88, 10126, Turin, Italy
| | - Guido Menato
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy.
| |
Collapse
|
16
|
Fillion A, Guerby P, Menzies D, Lachance C, Comeau MP, Bussières MC, Doucet-Gingras FA, Zérounian S, Bujold E. Pathological investigation of placentas in preeclampsia (the PEARL study). Hypertens Pregnancy 2020; 40:56-62. [PMID: 33373265 DOI: 10.1080/10641955.2020.1866008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION: Preeclampsia (PE), but mainly preterm PE, is associated with deep placentation disorders. We aimed to compare placental pathologies in pregnancies affected by term and preterm PE compared to normal pregnancies. METHODS: We performed a prospective case-cohort study. Low-risk nulliparous women were recruited in the first trimester and women who developed PE were recruited at diagnosis. Placental pathologies were reported according to the Amsterdam Placental Workshop Group Consensus Statement and were compared between cases and controls. PE cases stratified as term (≥37 weeks) and preterm PE (<37 weeks). Our primary outcome was maternal vascular malperfusion (MVM). RESULTS: Twenty-four women who developed preterm PE were compared to 10 women who developed term PE and 41 women without PE. Preterm PE (92%) was associated with more MVM than term PE (10%, p < 0.01) and controls (4%, p < 0.01), but the rate of MVM was similar between term PE and controls (p = 0.56). Preterm PE was also associated with more placental infarcts (65% vs. 20% vs. 15%); advanced villous maturation (91% vs. 30% vs. 1%); and hypoplastic villous maturation (70% vs. 10% vs. 3%); and moderate to severe decidual vasculopathy (56% vs. 10% vs. 3%) than term PE and controls (all p < 0.05). CONCLUSION: Most cases of preterm PE are associated with MVM, placental infarcts, advanced and/or hypoplastic villous maturation, and moderate to severe decidual vasculopathy, while it is infrequent in term PE and pregnancies without PE. Preterm and term preeclampsia have a different pathologic process that should be considered for their prevention and clinical management.
Collapse
Affiliation(s)
- Alexandre Fillion
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Social and Preventive Medicine, Université Laval , Quebec City, Canada
| | - Paul Guerby
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Obstetrics and Gynecology, Paule De Viguier Hospital, Centre Hospitalier Universitaire De Toulouse, Toulouse, Inserm U-1048, Université De Toulouse , France
| | - Didier Menzies
- Department of Fetopathology CHRU De Nancy, Nancy France.,Department of Pathology, Laboratoire National De Santé (LNS) , Luxembourg
| | - Caroline Lachance
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval , Quebec City, Canada
| | - Marie-Pier Comeau
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval , Quebec City, Canada
| | - Marie-Claude Bussières
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval , Quebec City, Canada
| | - Félicia-Allysson Doucet-Gingras
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval , Quebec City, Canada
| | - Sophie Zérounian
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval , Quebec City, Canada
| | - Emmanuel Bujold
- Reproduction, Mother and Child Health Unit, CHU De Québec - Université Laval Research Center, Université Laval , Quebec City, Canada.,Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval , Quebec City, Canada
| |
Collapse
|
17
|
de Moreuil C, Herry E, Lacut K, Chauvet J, Moineau MP, Lede F, Tremouilhac C, Merviel P, Petesch BP, Moigne EL, Marcorelles P. Correlation of biological parameters with placental parameters and pregnancy outcomes in pre-eclamptic women. Pregnancy Hypertens 2019; 19:61-66. [PMID: 31911371 DOI: 10.1016/j.preghy.2019.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/09/2019] [Accepted: 12/23/2019] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Pre-eclampsia is characterized by maternal vascular malperfusion and chronic inflammation in placenta. Our purpose was to investigate the potential correlation of biological parameters with placental parameters and pregnancy outcomes in pre-eclamptic women. METHODS Pre-eclamptic women were identified by interrogation of the Medical Registry Department in six French maternities between April 2013 and June 2018. Histological parameters in placentas (weight, macroscopic and microscopic lesions), baseline maternal characteristics and pregnancy outcomes (course of pregnancy, newborns' characteristics) were collected. Biological parameters were blood cell ratios (Platelet-to-Lymphocyte Ratio (PLR), Neutrophil-to-Lymphocyte Ratio (NLR)) collected at delivery and Placental growth factor (PlGF) measured in women with an available first trimester serum sample. Correlations of blood cell ratios and PlGF levels with placental parameters and pregnancy outcomes were assessed by Pearson's correlation test for quantitative parameters and by logistic regression analysis for qualitative parameters. RESULTS 202 pregnancies were included, among which 68 had a first trimester PlGF quantification. No correlation was found between biological parameters and placental lesions. Low PLR was correlated with low placental weight (r = 0.156, p = 0.036) and with low birth weight (r = 0.179, p = 0.015). Low PlGF was correlated with long time from pre-eclampsia diagnosis to delivery (r = -0.250, p = 0.048). CONCLUSIONS There is no correlation between biological parameters and placental lesions in pre-eclamptic women. Yet, low PLR at delivery is correlated with low placental and birth weights. Moreover, low first trimester PlGF is correlated with long time from pre-eclampsia diagnosis to delivery.
Collapse
Affiliation(s)
- Claire de Moreuil
- Département de médecine vasculaire, médecine interne et pneumologie, CHU de Brest, Hôpital La Cavale Blanche, Brest Cedex, France; EA 3878, GETBO, Université de Bretagne Occidentale, Brest Cedex, France.
| | - Emma Herry
- Service de gynécologie et d'obstétrique, CHU de Brest, Hôpital Morvan, Brest Cedex, France
| | - Karine Lacut
- Département de médecine vasculaire, médecine interne et pneumologie, CHU de Brest, Hôpital La Cavale Blanche, Brest Cedex, France; EA 3878, GETBO, Université de Bretagne Occidentale, Brest Cedex, France
| | - Juliette Chauvet
- Département de biochimie, CHU de Brest, Hôpital La Cavale Blanche, Brest Cedex, France
| | - Marie-Pierre Moineau
- Département de biochimie, CHU de Brest, Hôpital La Cavale Blanche, Brest Cedex, France
| | - Françoise Lede
- Service d'anatomopathologie, CHU de Brest, Hôpital Morvan, Brest Cedex, France
| | - Christophe Tremouilhac
- EA 3878, GETBO, Université de Bretagne Occidentale, Brest Cedex, France; Service de gynécologie et d'obstétrique, CHU de Brest, Hôpital Morvan, Brest Cedex, France
| | - Philippe Merviel
- EA 3878, GETBO, Université de Bretagne Occidentale, Brest Cedex, France; Service de gynécologie et d'obstétrique, CHU de Brest, Hôpital Morvan, Brest Cedex, France
| | - Brigitte Pan Petesch
- EA 3878, GETBO, Université de Bretagne Occidentale, Brest Cedex, France; Fédération de cancérologie et d'hématologie, CHU de Brest, Hôpital Morvan, Brest Cedex, France
| | - Emmanuelle Le Moigne
- Département de médecine vasculaire, médecine interne et pneumologie, CHU de Brest, Hôpital La Cavale Blanche, Brest Cedex, France; EA 3878, GETBO, Université de Bretagne Occidentale, Brest Cedex, France
| | - Pascale Marcorelles
- Service d'anatomopathologie, CHU de Brest, Hôpital Morvan, Brest Cedex, France; EA 4685 LIEN, Université de Bretagne Occidentale, Brest Cedex, France
| |
Collapse
|
18
|
Abstract
Cardiovascular disease remains the leading killer of women, with sex-specific manifestation, mechanisms, and morbidity. Preeclampsia, fetal growth restriction, and a subset of preterm births demonstrate aberrancies in the maternal vessels supplying the placenta and damage to the placental parenchyma consistent with hypoxic/ischemic or oxidative injury. This constellation of findings, maternal vascular malperfusion (MVM) lesions, may hold the key to understanding and identifying the elevated risk for early cardiovascular disease in women who experience adverse pregnancy outcomes. This intriguing possibility has only begun to be examined, but accumulating evidence is compelling and is reviewed here.
Collapse
|
19
|
Austdal M, Silva GB, Bowe S, Thomsen LCV, Tangerås LH, Bjørge L, Bathen TF, Iversen AC. Metabolomics Identifies Placental Dysfunction and Confirms Flt-1 (FMS-Like Tyrosine Kinase Receptor 1) Biomarker Specificity. Hypertension 2019; 74:1136-1143. [PMID: 31495279 DOI: 10.1161/hypertensionaha.119.13184] [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] [Indexed: 12/22/2022]
Abstract
Clinical end-stage parameters define the pregnancy disorders preeclampsia and fetal growth restriction while classification of the underlying placental dysfunction is missing and urgently needed. Flt-1 (FMS-like tyrosine kinase receptor 1) is the most promising placenta-derived predictive biomarker for preeclampsia. We aimed to classify placental dysfunction in preeclampsia and fetal growth restriction at delivery by metabolic profiling and authenticate the biomarker Flt-1 for placental dysfunction. We studied 143 pregnancies with or without preeclampsia and/or fetal growth restriction delivered by cesarean section. Metabolic placenta profiles were created by high-resolution magic angle spinning nuclear magnetic resonance spectroscopy and the resulting placental phenotypes obtained by hierarchical clustering. Placental Flt-1 expression (membrane-bound and soluble isoforms combined) and maternal serum Flt-1 expression (soluble isoforms) were analyzed by immunohistochemistry and ELISA, respectively. We identified 3 distinct placenta groups by 21 metabolites and diagnostic outcome parameters; normal placentas, moderate placental dysfunction, and severe placental dysfunction. Increased placental Flt-1 was associated with severe placental dysfunction, and increased serum Flt-1 was associated with moderate and severe placental dysfunction. The preeclamptic pregnancies with and without placental dysfunction could be distinguished by 5 metabolites and placental Flt-1. Placental Flt-1 alone could separate normal pregnancies with and without placental dysfunction. In conclusion, metabolomics could classify placental dysfunction and provide information not identified by traditional diagnostics and metabolites with biomarker potential were identified. Flt-1 was confirmed as precision biomarker for placental dysfunction, substantiating its usefulness for identification of high-risk pregnancies for preeclampsia and fetal growth restriction with placental involvement.
Collapse
Affiliation(s)
- Marie Austdal
- From the Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU) (M.A., G.B.S., L.C.V.T., L.H.T., A.-C.I.), Trondheim University Hospital, Norway.,Department of Circulation and Medical Imaging, NTNU (M.A., T.F.B.), Trondheim University Hospital, Norway.,Department of Research, Stavanger University Hospital, Norway (M.A.)
| | - Gabriela Brettas Silva
- From the Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU) (M.A., G.B.S., L.C.V.T., L.H.T., A.-C.I.), Trondheim University Hospital, Norway.,Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, Norway (G.B.S., S.B., L.H.T.)
| | - Sophie Bowe
- Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, Norway (G.B.S., S.B., L.H.T.)
| | - Liv Cecilie Vestrheim Thomsen
- From the Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU) (M.A., G.B.S., L.C.V.T., L.H.T., A.-C.I.), Trondheim University Hospital, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital and Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway (L.C.V.T., L.B.)
| | - Line Haugstad Tangerås
- From the Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU) (M.A., G.B.S., L.C.V.T., L.H.T., A.-C.I.), Trondheim University Hospital, Norway.,Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, Norway (G.B.S., S.B., L.H.T.)
| | - Line Bjørge
- Department of Gynecology and Obstetrics, Haukeland University Hospital and Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway (L.C.V.T., L.B.)
| | - Tone Frost Bathen
- Department of Circulation and Medical Imaging, NTNU (M.A., T.F.B.), Trondheim University Hospital, Norway
| | - Ann-Charlotte Iversen
- From the Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU) (M.A., G.B.S., L.C.V.T., L.H.T., A.-C.I.), Trondheim University Hospital, Norway
| |
Collapse
|
20
|
Muthyala T, Rathore A, Shahnaz G, Kumar D, Singh P, Bhasin S. Does morbidly adherent placenta mask hypertension in pregnancy - a case report and review of literature. J OBSTET GYNAECOL 2019; 39:1019-1021. [PMID: 31210086 DOI: 10.1080/01443615.2019.1588866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Tanuja Muthyala
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College , New Delhi , India
| | - Asmita Rathore
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College , New Delhi , India
| | - Gazala Shahnaz
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College , New Delhi , India
| | - Devender Kumar
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College , New Delhi , India
| | - Preeti Singh
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College , New Delhi , India
| | - Sangeeta Bhasin
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College , New Delhi , India
| |
Collapse
|
21
|
Johnsen G, Størvold G, Alnaes-Katjavivi P, Roald B, Golic M, Dechend R, Redman C, Staff A. Lymphocyte characterization of decidua basalis spiral arteries with acute atherosis in preeclamptic and normotensive pregnancies. J Reprod Immunol 2019; 132:42-48. [DOI: 10.1016/j.jri.2019.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/13/2019] [Accepted: 03/21/2019] [Indexed: 01/21/2023]
|
22
|
[Potential value of placental angiogenic factors as biomarkers in preeclampsia for clinical physicians]. Nephrol Ther 2019; 15:413-429. [PMID: 30935786 DOI: 10.1016/j.nephro.2018.10.005] [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] [Received: 04/09/2018] [Revised: 10/10/2018] [Accepted: 10/14/2018] [Indexed: 12/20/2022]
Abstract
The role of angiogenic factors in the onset of clinical manifestations of preeclampsia was demonstrated in 2003 by the implication of sFlt-1, PlGF and VEGF, and in 2006 by the implication of soluble endoglin. Placental ischemia and inflammation observed in preeclampsia alter both the production and progression of angiogenic factors during pregnancy. During the first trimester, the combination of PlGF with clinical, biophysical and biological factors results in a better test than the conventional one. However, the clinical value of this method remains to be confirmed. During the second and third trimesters, the sFlt-1/PlGF ratio may be used, with or without pre-existing renal disease, for short-term prediction, diagnosis, and prognosis, and to evaluate the effectiveness of preeclampsia treatment. While a sFlt-1/PlGF ratio<38 and≤33, respectively, rules out the short-term onset and diagnosis of preeclampsia, a sFlt-1/PlGF ratio≥85 between 20 and 34 weeks of pregnancy and≥110 beyond 34 weeks of pregnancy confirms a diagnosis of preeclampsia. Angiogenic and non-angiogenic preeclampsia are identified by a sFlt-1PlGF≥85 and<85, respectively, with the risk of maternal and fetal complications at two weeks differing between the two. Similarly, a sFlt-1/PlGF ratio>665 and>205, respectively, is a good short-term predictor of adverse outcomes of early and late-onset preeclampsia. These values could be incorporated into future guidelines for better clinical management of preeclampsia.
Collapse
|
23
|
Placenta Accreta Spectrum: A Review of Pathology, Molecular Biology, and Biomarkers. DISEASE MARKERS 2018; 2018:1507674. [PMID: 30057649 PMCID: PMC6051104 DOI: 10.1155/2018/1507674] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/10/2018] [Indexed: 12/14/2022]
Abstract
Background. Placenta accreta spectrum (PAS) is a condition of abnormal placental invasion encompassing placenta accreta, increta, and percreta and is a major cause of severe maternal morbidity and mortality. The diagnosis of a PAS is made on the basis of histopathologic examination and characterised by an absence of decidua and chorionic villi are seen to directly adjacent to myometrial fibres. The underlying molecular biology of PAS is a complex process that requires further research; for ease, we have divided these processes into angiogenesis, proliferation, and inflammation/invasion. A number of diagnostic serum biomarkers have been investigated in PAS, including human chorionic gonadotropin (HCG), pregnancy-associated plasma protein-A (PAPP-A), and alpha-fetoprotein (AFP). They have shown variable reliability and variability of measurement depending on gestational age at sampling. At present, a sensitive serum biomarker for invasive placentation remains elusive. In summary, there are a limited number of studies that have contributed to our understanding of the molecular biology of PAS, and additional biomarkers are needed to aid diagnosis and disease stratification.
Collapse
|
24
|
Weiner E, Feldstein O, Tamayev L, Grinstein E, Barber E, Bar J, Schreiber L, Kovo M. Placental histopathological lesions in correlation with neonatal outcome in preeclampsia with and without severe features. Pregnancy Hypertens 2018; 12:6-10. [PMID: 29674201 DOI: 10.1016/j.preghy.2018.02.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/19/2018] [Accepted: 02/06/2018] [Indexed: 12/09/2022]
Abstract
OBJECTIVE We aimed to compare pregnancy outcome and placental histopathology in women with preeclampsia (PE) with and without severe features. METHODS The medical records and placental pathology reports of all pregnancies complicated by PE during 2008-2016, were reviewed. Results were compared between those with and without severe features (severe PE vs. mild PE groups), according to current ACOG guidelines. Placental lesions were classified to maternal/fetal vascular supply lesions, and maternal/fetal inflammatory responses. Small for gestational age (SGA) was defined as neonatal birth-weight ≤10th%. Composite adverse neonatal outcome was defined as one or more of the following: sepsis, transfusion, phototherapy, respiratory morbidity, cerebral morbidity, NEC, or death. RESULTS The severe PE group (n = 284) was characterized by lower gestational age at delivery (p < 0.001), and higher rates of antenatal corticosteroid use (p = 0.003), and cesarean deliveries (p < 0.001) as compared to the mild PE group (n = 151). More placentas <10th% and more composite maternal vascular malperfusion (MVM) lesions were observed in the severe PE group as compared to the mild PE group (p < 0.001 for both). In multivariate analysis, composite placental MVM lesions were independently associated with severe PE (aOR = 1.75, 95%CI 1.4-4.9). Higher rates of SGA (p = 0.016), and composite adverse neonatal outcome (p = 0.002) characterized the severe PE group. In multivariate analysis, adverse neonatal outcome was independently associated with gestational age (aOR = 0.54, 95%CI 0.49-0.68), SGA (aOR = 1.75, 95%CI = 1.15-3.59), severe PE (aOR = 1.8, 95%CI = 1.13-3.54) and placental MVM lesions (aOR = 2.13, 95%CI = 1.05-4.39). CONCLUSION More pronounced placental pathology and higher rate of adverse neonatal outcome characterize preeclampsia with severe features as compared with the milder form of the disease.
Collapse
Affiliation(s)
- Eran Weiner
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Ohad Feldstein
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liliya Tamayev
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ehud Grinstein
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elad Barber
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Bar
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Letizia Schreiber
- Department of Pathology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Kovo
- Department of Obstetrics & Gynecology, The Edith Wolfson Medical Center, Holon, Israel affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
25
|
Scioscia M. D-chiro inositol phosphoglycans in preeclampsia: Where are we, where are we going? J Reprod Immunol 2017; 124:1-7. [DOI: 10.1016/j.jri.2017.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/15/2017] [Accepted: 09/27/2017] [Indexed: 12/24/2022]
|
26
|
Abstract
PURPOSE To confirm reduced expression of soluble fms-like tyrosine kinase 1 (sFlt-1) in accreta/increta. METHODS Formalin-fixed tissue sections from 11 peripartum hysterectomies with invasive placentation and 5 controls were stained for sFlt-1. Stain intensity was scored in selected 100× microscopic fields. We compared sFlt-1 expression in invasive areas among cases, non-invasive areas among cases and areas from control placentas. RESULTS Chorionic villi displayed significantly decreased sFlt-1 expression in invasive areas of cases compared to control placentas (p = 0.003), as well as in non-invasive areas of cases compared to control placentas (p = 0.01). There was no difference in sFlt-1 expression between invasive and non-invasive areas among cases. CONCLUSIONS Expression of sFlt-1 is diminished in villous trophoblasts from patients with placenta increta or percreta. Local depth of invasion was not associated with sFlt-1 expression, suggesting a more global abnormality across the implantation site rather than localized to areas of histologic invasion.
Collapse
|
27
|
Spiel M, Salahuddin S, Pernicone E, Zsengeller Z, Wang A, Modest AM, Karumanchi SA, Hecht JL. Placental soluble fms-like tyrosine kinase expression in small for gestational age infants and risk for adverse outcomes. Placenta 2017; 52:10-16. [PMID: 28454690 DOI: 10.1016/j.placenta.2017.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 02/03/2017] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Soluble fms-like tyrosine kinase 1 (sFLT-1) is an anti-angiogenic factor implicated in the pathogenesis of preterm preeclampsia. We evaluated sFLT-1 expression and placental pathology in pregnancies complicated by small for gestational age (SGA) infants (<10th percentile), without evidence of preeclampsia. METHODS Clinical and histologic data were compared between groups with high or low sFLT-1 expression determined by immunohistochemistry on archived placentas. RESULTS Nineteen of 69 placentas showed high sFLT-1 expression. The high sFLT-1 group had higher predelivery median systolic blood pressure (BP); 140 (interquartile range (IQR) 133-152) vs. 126 (118-139) mm Hg (p = 0.003), and median diastolic BP; 87 (78-94) vs. 77.5 (71-86) mm Hg (p = 0.02). Abnormal umbilical Doppler abnormalities were more prevalent; 89.5% vs. 46% (p = 0.001). These pregnancies delivered earlier; 31.9 weeks (28.3-34.7 weeks) vs. 37.1 weeks (33.7-38.7 weeks) (p < 0.001), and infants had lower birthweight; 980 grams (520-1545 grams) vs. 2087.5 grams (1455-2340 grams) (p < 0.001). Placental-weight to fetal-weight ratios, a marker of vascular insufficiency, was increased in the high sFlt-1 group: 0.18 (0.14-0.28) vs 0.15 (0.13-0.18), p = 0.03. Placentas with high sFLT-1 showed more decidual vasculopathy; 42.1% vs. 10.0% (p = 0.005), infarction; 36.8% vs. 14.0% (p = 0.048), distal villous hypoplasia; 78.9% vs. 36.0% (p = 0.001), and fetal thrombotic vasculopathy; 47.4% vs. 16.0% (p = 0.01). DISCUSSION Placental sFLT-1 expression is upregulated in approximately 28% of non-preeclamptic pregnancies complicated by SGA infants. These pregnancies showed increased placental vascular pathology, more umbilical Doppler abnormalities, and earlier delivery with lower birthweight. A subgroup of non-preeclamptic fetal growth restriction with upregulated sFlt-1 expression may share a common pathogenic pathway with preterm preeclampsia. This subgroup is worthy of additional study.
Collapse
Affiliation(s)
- Melissa Spiel
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Saira Salahuddin
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA; Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Elizabeth Pernicone
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Zsuzsanna Zsengeller
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Alice Wang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA; Department of Medicine, Boston University School of Medicine, One Medical Center Place, Boston, MA 02118, USA
| | - Anna M Modest
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - S Ananth Karumanchi
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA; Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA.
| |
Collapse
|
28
|
Weel IC, Baergen RN, Romão-Veiga M, Borges VT, Ribeiro VR, Witkin SS, Bannwart-Castro C, Peraçoli JC, De Oliveira L, Peraçoli MT. Association between Placental Lesions, Cytokines and Angiogenic Factors in Pregnant Women with Preeclampsia. PLoS One 2016; 11:e0157584. [PMID: 27315098 PMCID: PMC4912084 DOI: 10.1371/journal.pone.0157584] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/01/2016] [Indexed: 11/29/2022] Open
Abstract
Preeclampsia (PE) is considered the leading cause of maternal and perinatal morbidity and mortality. The placenta seems to play an essential role in this disease, probably due to factors involved in its formation and development. The present study aimed to investigate the association between placental lesions, cytokines and angiogenic factors in pregnant women with preeclampsia (PE). We evaluated 20 normotensive pregnant women, 40 with early-onset PE and 80 with late-onset PE. Placental samples were analyzed for histopathology, immunohistochemistry and determination of granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-10 (IL-10), transforming growth factor-beta 1 (TGF-β1), tumor necrosis factor-alpha (TNF-α), placental growth factor (PlGF), vascular endothelial growth factor (VEGF), fms-like tyrosine-kinase-1 (Flt-1) and endoglin (Eng) levels. Higher percentages of increased syncytial knots and increased perivillous fibrin deposits, and greater levels of TNF-α, TGF-β1and Flt-1 were detected in placentas from early-onset PE. Levels of IL-10, VEGF and PlGF were decreased in PE versus normotensive placentas. Both the TNF-α/IL-10 and sFlt-1/PlGF ratios were higher in placental homogenate of early-onset PE than late-onset PE and control groups. The more severe lesions and the imbalance between TNF-α/IL-10 and PlGF/sFlt-1 in placentas from early-onset PE allows differentiation of early and late-onset PE and suggests higher placental impairment in early-onset PE.
Collapse
Affiliation(s)
- Ingrid C. Weel
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Rebecca N. Baergen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College – New York Presbyterian Hospital, New York, United States of America
| | - Mariana Romão-Veiga
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Vera T. Borges
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Vanessa R. Ribeiro
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Steven S. Witkin
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, United States of America
| | - Camila Bannwart-Castro
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Jose C. Peraçoli
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Leandro De Oliveira
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
| | - Maria T. Peraçoli
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University, 18618–970, Botucatu, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
29
|
Korzeniewski SJ, Romero R, Chaiworapongsa T, Chaemsaithong P, Kim CJ, Kim YM, Kim JS, Yoon BH, Hassan SS, Yeo L. Maternal plasma angiogenic index-1 (placental growth factor/soluble vascular endothelial growth factor receptor-1) is a biomarker for the burden of placental lesions consistent with uteroplacental underperfusion: a longitudinal case-cohort study. Am J Obstet Gynecol 2016; 214:629.e1-629.e17. [PMID: 26688491 DOI: 10.1016/j.ajog.2015.11.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/21/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Placental lesions consistent with maternal vascular underperfusion (MVU) are thought to be pathogenically linked to preeclampsia, small-for-gestational-age newborns, fetal death, and spontaneous preterm labor and delivery; yet, these lesions cannot be diagnosed antenatally. We previously reported that patients with such conditions and lesions have an abnormal profile of the angiogenic placental growth factor (PlGF) and antiangiogenic factors (eg, soluble vascular endothelial growth factor receptor [sVEGFR]-1). OBJECTIVE The objectives of this study were to: (1) examine the relationship between the maternal plasma PlGF/sVEGFR-1 concentration ratio (referred to herein as angiogenic index-1) and the burden of histologic placental features consistent with MVU; and (2) test the hypothesis that angiogenic index-1 can identify patients in the midtrimester who are destined to deliver before 34 weeks of gestation with multiple (ie, ≥3) histologic placental features consistent with MVU. STUDY DESIGN A 2-stage case-cohort sampling strategy was used to select participants from among 4006 women with singleton gestations enrolled from 2006 through 2010 in a longitudinal study. Maternal plasma angiogenic index-1 ratios were determined using enzyme-linked immunosorbent assays. Placentas underwent histologic examination according to standardized protocols by experienced pediatric pathologists who were blinded to clinical diagnoses and pregnancy outcomes. The diagnosis of lesions consistent with MVU was made using criteria proposed by the Perinatal Section of the Society for Pediatric Pathology. Weighted analyses were performed to reflect the parent cohort; "n*" is used to reflect weighted frequencies. RESULTS (1) Angiogenic index-1 (PlGF/sVEGFR-1) concentration ratios were determined in 7560 plasma samples collected from 1499 study participants; (2) the prevalence of lesions consistent with MVU was 21% (n* = 833.9/3904) and 27% (n* = 11.4/42.7) of women with ≥3 MVU lesions delivered before 34 weeks of gestation; (3) a low angiogenic index-1 (<2.5th quantile for gestational age) in maternal plasma samples obtained within 48 hours of delivery had a sensitivity of 73% (n* = 8.3/11.4; 95% confidence interval [CI], 47-98%), a specificity of 94% (n* = 3130.9/3316.2; 95% CI, 94-95%), a positive likelihood ratio of 12.2, and a negative likelihood ratio of 0.29 in the identification of patients who delivered placentas with ≥3 MVU lesions at <34 weeks; (4) prospectively, at 20-23 weeks of gestation, a maternal plasma concentration of angiogenic index-1 <2.5th quantile identified 70% (n* = 7.2/10.3; 95% CI, 42-98%) of patients who delivered placentas with ≥3 MVU lesions before 34 weeks (specificity, 97% [n* = 2831.3/2918; 95% CI, 96-98%]; positive likelihood ratio, 23; negative likelihood ratio, 0.31); and (5) among women without obstetrical complications who delivered at term, angiogenic index-1 was lower in women with than without placental lesions consistent with MVU (P < .05). CONCLUSION Maternal plasma angiogenic index-1 (PlGF/sVEGFR-1) is the first biomarker for the burden of placental lesions consistent with MVU. We propose that an accumulation of these lesions in placentas delivered before 34 weeks is a histologic counterpart of an antiangiogenic profile.
Collapse
|
30
|
Makris A, Yeung KR, Lim SM, Sunderland N, Heffernan S, Thompson JF, Iliopoulos J, Killingsworth MC, Yong J, Xu B, Ogle RF, Thadhani R, Karumanchi SA, Hennessy A. Placental Growth Factor Reduces Blood Pressure in a Uteroplacental Ischemia Model of Preeclampsia in Nonhuman Primates. Hypertension 2016; 67:1263-72. [PMID: 27091894 DOI: 10.1161/hypertensionaha.116.07286] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/04/2016] [Indexed: 01/04/2023]
Abstract
An imbalance in the angiogenesis axis during pregnancy manifests as clinical preeclampsia because of endothelial dysfunction. Circulating soluble fms-like tyrosine kinase 1 (sFLT-1) increases and placental growth factor (PlGF) reduces before and during disease. We investigated the clinical and biochemical effects of replenishing the reduced circulating PlGF with recombinant human PlGF (rhPlGF) and thus restoring the angiogenic balance. Hypertensive proteinuria was induced in a nonhuman primate (Papio hamadryas) by uterine artery ligation at 136 days gestation (of a 182-day pregnancy). Two weeks after uteroplacental ischemia, rhPlGF (rhPlGF, n=3) or normal saline (control, n=4) was administered by subcutaneous injection (100 μg/kg per day) for 5 days. Blood pressure was monitored by intra-arterial radiotelemetry and sFLT-1 and PlGF by ELISA. Uteroplacental ischemia resulted in experimental preeclampsia evidenced by increased blood pressure, proteinuria, and endotheliosis on renal biopsy and elevated sFLT-1. PlGF significantly reduced after uteroplacental ischemia. rhPlGF reduced systolic blood pressure in the treated group (-5.2±0.8 mm Hg; from 132.6±6.6 mm Hg to 124.1±7.6 mm Hg) compared with an increase in systolic blood pressure in controls (6.5±3 mm Hg; from 131.3±1.5 mm Hg to 138.6±1.5 mm Hg). Proteinuria reduced in the treated group (-72.7±55.7 mg/mmol) but increased in the control group. Circulating levels of total sFLT-1 were not affected by the administration of PlGF; however, a reduction in placental sFLT-1 mRNA expression was demonstrated. There was no significant difference between the weights or lengths of the neonates in the rhPlGF or control group; however, this study was not designed to assess fetal safety or outcomes. Increasing circulating PlGF by the administration of rhPlGF improves clinical parameters in a primate animal model of experimental preeclampsia.
Collapse
Affiliation(s)
- Angela Makris
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.).
| | - Kristen R Yeung
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Shirlene M Lim
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Neroli Sunderland
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Scott Heffernan
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - John F Thompson
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Jim Iliopoulos
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Murray C Killingsworth
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Jim Yong
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Bei Xu
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Robert F Ogle
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Ravi Thadhani
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - S Ananth Karumanchi
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| | - Annemarie Hennessy
- From the Medicine Faculty, Western Sydney University and Ingham Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., J.I., M.C.K., B.X., A.H.); Medicine Faculty, University of New South Wales, Sydney, NSW, Australia (A.M., M.C.K., J.Y.); Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia (A.M., J.I.); Vascular Immunology Group, Heart Research Institute, Sydney, NSW, Australia (A.M., K.R.Y., S.M.L., B.X., A.H.); Nephrology Department (N.S., S.H.), Melanoma Unit (J.F.T.), and Obstetrics Department (R.F.O.), Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Surgery, University of Sydney, Sydney, NSW, Australia (J.F.T.); Anatomical Pathology Department (M.C.K., J.Y.) and Vascular Surgery Department (J.I.), Liverpool Hospital, Liverpool, NSW, Australia; Division of Nephrology, Massachusetts General Hospital, Boston (R.T.); and Centre for Vascular Biology, Beth Israel Deaconess Medical Centre, Boston, MA (S.A.K.)
| |
Collapse
|
31
|
Abstract
OBJECTIVES Preeclampsia is a serious pregnancy-specific hypertensive syndrome that is characterized by widespread maternal endothelial dysfunction. Previous studies have shown that increased levels of circulating cell-free fetal DNA in women with preeclampsia correspond to the degree of disease severity; however, it is unknown whether this DNA is a key signal that contributes to the development of preeclampsia. The detection of DNA is critical to appropriate innate immune responses. The interferon-inducible protein 16 (IFI16) - a member of the HIN-200 family - is an innate immune receptor for intracellular DNA, which is implicated in the control of cell growth, apoptosis, angiogenesis, and immunomodulation; however, its role in preeclampsia remains unresolved. Here, we tested the hypothesis that this DNA can activate IFI16 in the placentas of women with preeclampsia and is sufficient to induce soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sEng) production. METHODS We characterized IFI16 in severe preeclamptic placentas and assessed whether DNA increased the release of sFlt-1 and sEng from trophoblast cells and placental explants. Furthermore, we determined whether IFI16 was involved in DNA-induced sFlt-1 and sEng production. RESULTS Placental immunoreactivity and protein levels of IFI16 were significantly increased in women with preeclampsia compared to matched control women. Treatment of human trophoblasts with the IFI16 agonist poly(dA:dT) significantly increased IFI16 levels. Furthermore, poly(dA:dT) induced sFlt-1 and sEng production by human trophoblasts in an IFI16-dependent manner. CONCLUSIONS We conclude that trophoblast cells respond to cell-free fetal DNA through the IFI16 receptor, resulting in the production of the preeclampsia-related antiangiogenic factors sFlt-1 and sEng.
Collapse
|
32
|
Hecht JL, Zsengeller ZK, Spiel M, Karumanchi SA, Rosen S. Revisiting decidual vasculopathy. Placenta 2016; 42:37-43. [PMID: 27238712 DOI: 10.1016/j.placenta.2016.04.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/01/2016] [Accepted: 04/06/2016] [Indexed: 01/11/2023]
Abstract
INTRODUCTION In the setting of preeclampsia (PE), decidual vasculopathy (DV) can be seen along the free membranes. METHODS We describe DV using stains for CD31, CD34, Cd42b, CD68, desmin, fibrin and Masson's trichrome in patients with preeclampsia and fetal growth restriction. RESULTS We first examined the "membrane roll" sections from the placentas of six patients with preeclampsia. Affected vessels showed endothelial proliferation with detachment. Remodeling of the media was characterized by smooth muscle loss with variable degrees of fibrin deposition. CD31 and CD34 highlighted the prominent endothelium and showed striking particulate staining throughout the media. All of these findings infer a sequence of endothelial injury, fragmentation and repair with incorporation of endothelial components into the vascular wall. We evaluated the frequency of DV by clinical presentation; in cases with PE with and without small for gestational age (SGA) (N = 15), and SGA with and without Doppler flow abnormalities (N = 15). All groups except the SGA without Doppler abnormalities showed DV. Among placentas with DV, the most severely affected group was PE with SGA; the least affected was PE without SGA. DISCUSSION The association with SGA suggests that the DV is a subacute process of vascular injury that accelerates in the setting of PE. The majority of DV cases were not initially recognized suggesting a role for endothelial markers for DV detection. We also propose that the rampant endothelial injury seems to be a prominent finding in the decidual vessels of subjects with PE complicated by SGA and a similar process in the systemic vasculature may be responsible for the circulating endothelial microparticles reported in such patients.
Collapse
Affiliation(s)
- Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center (BIDMC), 330 Brookline Ave, Boston, MA 02215, USA.
| | - Zsuzsanna K Zsengeller
- Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), 330 Brookline Ave, Boston, MA 02215, USA.
| | - Melissa Spiel
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center (BIDMC), 330 Brookline Ave, Boston, MA 02215, USA.
| | - S Ananth Karumanchi
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center (BIDMC), 330 Brookline Ave, Boston, MA 02215, USA; Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), 330 Brookline Ave, Boston, MA 02215, USA.
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center (BIDMC), 330 Brookline Ave, Boston, MA 02215, USA.
| |
Collapse
|
33
|
Lundberg GP, Dunbar SB, Wenger NK. Guidelines for the Reduction of Cardiovascular Disease in Women. J Obstet Gynecol Neonatal Nurs 2016; 45:402-12. [PMID: 26993208 DOI: 10.1016/j.jogn.2016.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2015] [Indexed: 11/30/2022] Open
Abstract
Since 1984, more women than men have died each year from cardiovascular disease. Various organizations, such as the American Heart Association and the American College of Cardiology, have published prevention guidelines for heart disease and stroke that may improve care for women. In this article, we review these guidelines, the results of new studies on emerging risk factors, and new approaches for reducing cardiovascular disease in women.
Collapse
|
34
|
Alnaes-Katjavivi P, Lyall F, Roald B, Redman CWG, Staff AC. Acute atherosis in vacuum suction biopsies of decidua basalis: An evidence based research definition. Placenta 2015; 37:26-33. [PMID: 26608629 DOI: 10.1016/j.placenta.2015.10.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acute atherosis (AA) of the uteroplacental spiral arteries has been characterised by subendothelial lipid-laden foam cells, perivascular leukocyte infiltrates (PVI) and fibrinoid necrosis. Because precise diagnostic criteria are not available for comparative research studies we developed and tested new simplified criteria based on 237 cases. METHODS Decidual basalis samples were collected by vacuum suction at elective cesarean deliveries. Spiral arteries were evaluated in serial decidual tissue sections from women with normal pregnancy, preeclampsia, and diabetes. Features of AA were sought in parallel sections stained with H&E and immunostained for CD68, cytokeratin CK7 and desmin, and costained with Periodic Acid Schiff (PAS). RESULTS Foam cell lesions were defined as two or more adjacent, intramural, vacuolated CD68 positive cells, PVI as a focal perivascular lymphocyte accumulation, more dense than in the surrounding decidua. Increased fibrinoid (PAS positive) was identified if present in ≥75% of the arterial wall circumference. PVI and increased fibrinoid were significantly associated with preeclampsia but not specifically associated with the presence of foam cell lesions. Hence we diagnosed decidua basalis AA lesions solely by the presence of foam cell lesions, occurring in preeclampsia (37%), diabetes (10%) and healthy normotensive women (11%). The simplified criterion was reproducible by different investigators. Decidua basalis AA occurred most commonly and extensively in preeclampsia, but did not distinguish between preterm and term disease. DISCUSSION Our evidence based criterion for decidua basalis AA diagnosis in vacuum suction biopsies may not apply to myometrial or decidua parietalis arteries. In decidual basalis samples it should facilitate comparisons between research studies, to improve pathophysiological understanding of AA and preeclampsia.
Collapse
Affiliation(s)
- Patji Alnaes-Katjavivi
- Department of Obstetrics and Department of Gynecology, Oslo University Hospital, Ullevål and Faculty of Medicine, University of Oslo, Norway
| | - Fiona Lyall
- University of Glasgow, School of Medicine, Glasgow, UK
| | - Borghild Roald
- Department of Pathology, Oslo University Hospital, Ullevål and Faculty of Medicine, University of Oslo, Norway
| | | | - Anne Cathrine Staff
- Department of Obstetrics and Department of Gynecology, Oslo University Hospital, Ullevål and Faculty of Medicine, University of Oslo, Norway.
| |
Collapse
|
35
|
|