1
|
Peng X, Chinwe Oluchi-Amaka I, Kwak-Kim J, Yang X. A comprehensive review of the roles of T-cell immunity in preeclampsia. Front Immunol 2025; 16:1476123. [PMID: 39981257 PMCID: PMC11841438 DOI: 10.3389/fimmu.2025.1476123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Preeclampsia (PE) is an obstetrical disorder that occurs after the 20th week of gestation. It is recognized as one of the "Great Obstetrical Syndromes" and principally contributes to maternal morbidity and mortality. PE has been associated with a range of immune disorders, including a preponderance of T helper (Th) 1 over Th2 cells and imbalanced levels of Th17 and T regulatory cells (Tregs). During pregnancy, T cells safeguard the placenta against immune rejection and aid embryo implantation while involved in pregnancy complications, such as PE. Promoting alloantigen-specific Treg cells is a potential preventive and therapeutic strategy for PE. However, ensuring the safety of mothers and infants is of the utmost importance since the risk-benefit ratio of reproductive and obstetric conditions differs significantly from that of immune diseases that pose a life-threatening risk. In this review, we systematically summarize the roles of T-cell immunity in the peripheral blood, reproductive tissues, and at the maternal-fetal interface of PE patients. Furthermore, the recent therapeutic approaches centered on targeting T cell immunity in PE are critically appraised.
Collapse
Affiliation(s)
- Xu Peng
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| | | | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Clinical Immunology Laboratory, Foundational Sciences and Humanities, Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Wiley KS, Martínez LE, Kwon D, Knorr DA, Epeldegui M, Fox MM. Regulatory B-Cells Are Associated Negatively With Regulatory T-Cells and Positively With Cytokines in Peripheral Blood of Pregnant Women. Am J Reprod Immunol 2025; 93:e70027. [PMID: 39854121 PMCID: PMC12001748 DOI: 10.1111/aji.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/27/2024] [Accepted: 11/27/2024] [Indexed: 01/26/2025] Open
Abstract
PROBLEM Regulatory B-cells (Bregs, CD19+CD24hiCD38hi) are a specialized B-cell subset that suppresses immune responses and potentially contribute to the maintenance of an immune-privileged environment for fetal development during pregnancy. However, little is known about the surrounding immunological environment of Bregs in gestational physiology. The relationship of regulatory T-cells (Tregs, CD4+CD25hiCD127loFoxP3+) to Bregs in coordinating immunoregulation during pregnancy is unknown. We aimed to determine whether peripheral concentrations of Bregs and/or PD-L1-expressing Bregs correlated with Tregs and cytokines during pregnancy. METHOD Peripheral blood samples were obtained from 29 pregnant women at mean 12 weeks' gestation. Participants were age ≥ 18, self-identified as Latina/Hispanic, and N = 12 primigravid. Peripheral blood mononuclear cells were isolated, stained, and analyzed by flow cytometry to determine percentages of Tregs from CD4+ T-cells and five Treg subsets defined by immune checkpoint markers, and Bregs and PD-L1+ Bregs from total B-cells. Levels of 13 cytokines were measured on a Meso Scale Discovery multiplex platform. RESULTS Bregs positively correlated with pro-inflammatory cytokine interleukin (IL)-6. PD-L1+ Bregs positively correlated with T-cell suppressive cytokine IL-10. PD-L1+ Bregs negatively correlated with Tregs and Helios+, CTLA-4+, PD-1+, TIGIT+, and TIM3+ Tregs. For primigravida, PD-L1+ Bregs correlated positively with IL-10 and negatively with Helios+ and TIGIT+ Tregs. For multigravida, PD-L1+ Bregs correlated positively with IL-8 and negatively with Helios+, CTLA-4+, PD-1+, and TIGIT+ Tregs. CONCLUSIONS This study provides insight into the immunosuppressive role of Bregs and PD-L1+ Bregs during human pregnancy. Our results suggest that PD-L1+ Bregs can employ suppressive mechanisms to limit pro-inflammatory responses in primigravida.
Collapse
Affiliation(s)
- Kyle S. Wiley
- Department of Anthropology, University of California, Los Angeles, California, USA
| | - Laura E. Martínez
- UCLA AIDS Institute and David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Dayoon Kwon
- Department of Epidemiology, UCLA Fielding School of Public Health, University of California, Los Angeles, California, USA
| | - Delaney A. Knorr
- Department of Anthropology, University of California, Los Angeles, California, USA
| | - Marta Epeldegui
- UCLA AIDS Institute and David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Molly M. Fox
- Department of Anthropology, University of California, Los Angeles, California, USA
- Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
3
|
Allerkamp HH, Bondarenko AI, Tawfik I, Kamali-Simsek N, Horvat Mercnik M, Madreiter-Sokolowski CT, Wadsack C. In vitro examination of Piezo1-TRPV4 dynamics: implications for placental endothelial function in normal and preeclamptic pregnancies. Am J Physiol Cell Physiol 2025; 328:C227-C244. [PMID: 39652778 DOI: 10.1152/ajpcell.00794.2024] [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: 10/23/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025]
Abstract
Mechanosensation is essential for endothelial cell (EC) function, which is compromised in early-onset preeclampsia (EPE), impacting offspring health. The ion channels Piezo-type mechanosensitive ion channel component 1 (Piezo1) and transient receptor potential cation channel subfamily V member 4 (TRPV4) are coregulated mechanosensors in ECs. Current evidence suggests that both channels could mediate aberrant placental endothelial function in EPE. Using isolated fetoplacental ECs (fpECs) from early control (EC) and EPE pregnancies, we show functional coexpression of both channels and that Ca2+ influx and membrane depolarization in response to chemical channel activation is reduced in EPE fpECs. Downstream of channel activation, Piezo1 alone can induce phosphorylation of endothelial nitric oxide synthase (eNOS) in fpECs, while combined activation of Piezo1 and TRPV4 only affects eNOS phosphorylation in EPE fpECs. Additionally, combined activation reduces the barrier integrity of fpECs and has a stronger effect on EPE fpECs. This implies altered Piezo1-TRPV4 coregulation in EPE. Mechanistically, we suggest this to be driven by changes in the arachidonic acid metabolism in EPE fpECs as identified by RNA sequencing. Targeting of Piezo1 and TRPV4 might hold potential for EPE treatment options in the future.NEW & NOTEWORTHY This study shows Piezo-type mechanosensitive ion channel component 1 (Piezo1) and transient receptor potential cation channel subfamily V member 4 (TRPV4) coexpression and functionality within primary human fetoplacental endothelial cells (fpECs), mediating nitric oxide (NO) production and barrier integrity. In early-onset preeclampsia (EPE), fpEC channel functionality and coregulation are impaired, affecting Ca2+ signaling and endothelial barrier function. Combined channel activation significantly reduces endothelial barrier integrity and increases NO production in EPE. Changes in arachidonic acid metabolism are suggested as a key underlying factor mediating impaired channel functionality in EPE fpECs.
Collapse
Affiliation(s)
- Hanna H Allerkamp
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | | - Ines Tawfik
- Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | | | | | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
4
|
Schliefsteiner C, Wadsack C, Allerkamp HH. Exploring the Lifeline: Unpacking the Complexities of Placental Vascular Function in Normal and Preeclamptic Pregnancies. Compr Physiol 2024; 14:5763-5787. [PMID: 39699084 DOI: 10.1002/cphy.c230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The proper development and function of the placenta are essential for the success of pregnancy and the well-being of both the fetus and the mother. Placental vascular function facilitates efficient fetal development during pregnancy by ensuring adequate gas exchange with low vascular resistance. This review focuses on how placental vascular function can be compromised in the pregnancy pathology preeclampsia, and conversely, how placental vascular dysfunction might contribute to this condition. While the maternal endothelium is widely recognized as a key focus in preeclampsia research, this review emphasizes the importance of understanding how this condition affects the development and function of the fetal placental vasculature. The placental vascular bed, consisting of microvasculature and macrovasculature, is discussed in detail, as well as structural and functional changes associated with preeclampsia. The complexity of placental vascular reactivity and function, its mediators, its impact on placental exchange and blood distribution, and how these factors are most affected in early-onset preeclampsia are further explored. These factors include foremost lipoproteins and their cargo, oxygen levels and oxidative stress, biomechanics, and shear stress. Challenges in studying placental pathophysiology are discussed, highlighting the necessity of innovative research methodologies, including ex vivo experiments, in vivo imaging tools, and computational modeling. Finally, an outlook on the potential of drug interventions targeting the placental endothelium to improve placental vascular function in preeclampsia is provided. Overall, this review highlights the need for further research and the development of models and tools to better understand and address the challenges posed by preeclampsia and its effects on placental vascular function to improve short- and long-term outcomes for the offspring of preeclamptic pregnancies. © 2024 American Physiological Society. Compr Physiol 14:5763-5787, 2024.
Collapse
Affiliation(s)
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Hanna H Allerkamp
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| |
Collapse
|
5
|
Timmer-Murillo SC, Mowrer A, Wang AZ, Jazinski-Chambers K, Piña I, Rundell MR, Bennett JM, Wagner AJ, deRoon-Cassini TA. Examining emotion regulation and inflammation as predictors of maternal mental health after fetal anomaly diagnosis. Brain Behav Immun 2024; 122:1-8. [PMID: 39106938 DOI: 10.1016/j.bbi.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/09/2024] [Accepted: 08/01/2024] [Indexed: 08/09/2024] Open
Abstract
OBJECTIVE Fetal anomalies occur in approximately 3% of pregnancies and receiving the diagnosis may be a potentially traumatic experience for families. The mental health of mothers receiving diagnoses and what predicts resilience or poor mental health is understudied. Emotion regulation is an important, modifiable, transdiagnostic factor of mental health, and may be protective post-diagnosis. Evaluating biomarkers of stress, including IL-6 and Allostatic Load (AL), can also serve as early indicators of risk, indicative of early intervention. This study assessed whether reappraisal, suppression, IL-6, and AL was associated with mental health outcomes and resilience in women after receiving a fetal anomaly diagnosis. METHODS Pregnant women (N=108) presenting to a fetal concerns clinic for initial consultation completed measures of emotion regulation (i.e., reappraisal and suppression), depression, anxiety, posttraumatic stress symptoms, and resilience between 2019-2022. A blood draw was used to assess IL-6 and create composite allostatic load measure including: IL-6, blood pressure, heart rate, glucose, cortisol, and body mass index. RESULTS Linear regressions controlling for age, gestational age, and perceived fetal diagnosis severity, demonstrated that IL-6 was negatively associated with resilience and positively associated with depression. Reappraisal was positively associated to resilience and negatively associated with depression, anxiety, and PTSD, whereas state insurance status was positively associated to anxiety and PTS symptoms. Suppression and allostatic load were not significant. CONCLUSIONS Women experiencing fetal anomaly diagnosis represent an understudied population with unaddressed mental health needs. Reappraisal serves as not only a protective factor, but one that can be enhanced to promote maternal resilience and mental health. Furthermore, elevated IL-6 may be a critical early indicator of potential intervention needs among women who are pregnant, to mitigate negative psychological states and enhance resilience.
Collapse
Affiliation(s)
- Sydney C Timmer-Murillo
- Medical College of Wisconsin, Division of Trauma and Acute Care Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | - Alyssa Mowrer
- Medical College of Wisconsin, Division of Pediatric Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | - Amy Z Wang
- University of Wisconsin, Milwaukee, Department of Psychology, 2441 E. Hartford Ave. Milwaukee, WI 53211, USA.
| | - Kelley Jazinski-Chambers
- Medical College of Wisconsin, Division of Trauma and Acute Care Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | - Isela Piña
- Medical College of Wisconsin, Division of Trauma and Acute Care Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | - Maddie R Rundell
- Medical College of Wisconsin, Division of Pediatric Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | - Jeanette M Bennett
- UNC Charlotte, Department of Psychological Science, 9201 University City Blvd Charlotte, NC 28223, USA.
| | - Amy J Wagner
- Medical College of Wisconsin, Division of Pediatric Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | - Terri A deRoon-Cassini
- Medical College of Wisconsin, Division of Trauma and Acute Care Surgery, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA; Comprehensive Injury Center, Division of Data Surveillance & Informatics, 10000 Innovation Dr. Milwaukee, WI 53226, USA.
| |
Collapse
|
6
|
Ng KW, Chaturvedi N, Coté GL, Fisher SA, Mabbott S. Biomarkers and point of care screening approaches for the management of preeclampsia. COMMUNICATIONS MEDICINE 2024; 4:208. [PMID: 39433973 PMCID: PMC11493996 DOI: 10.1038/s43856-024-00642-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Preeclampsia is a multi-organ pregnancy complication, that is primarily detected when pregnant people have high blood pressure, and is confirmed by testing for the presence of protein in the urine. While more specific and accurate diagnostic and imaging tests are becoming available, they are still in the process of undergoing widespread regulatory adoption, and so are not yet the standard of care. Since biochemical processes are a precursor to the systemic progression of disease, we review some established, emerging, and promising biomarkers that are proposed to be associated with preeclampsia, and newly developed approaches for screening them at the point of care, to reduce the burden of the disease.
Collapse
Affiliation(s)
- Ka Wai Ng
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA
| | - Nandita Chaturvedi
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA
| | - Gerard L Coté
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA
| | - Stephanie A Fisher
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Samuel Mabbott
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA.
| |
Collapse
|
7
|
Jordan MM, Amabebe E, Khanipov K, Taylor BD. Scoping Review of Microbiota Dysbiosis and Risk of Preeclampsia. Am J Reprod Immunol 2024; 92:e70003. [PMID: 39440917 PMCID: PMC11501047 DOI: 10.1111/aji.70003] [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: 04/23/2024] [Revised: 08/20/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Limited studies have investigated the role of the microbiota in hypertensive disorders of pregnancy (HDP), particularly preeclampsia, which often results in preterm birth. We evaluated 23 studies that explored the relationship between gut, vaginal, oral, or placental microbiotas and HDP. Scopus, ProQuest Health Research Premium Collection, ProQuest Nursing & Allied Health Database, EBSCO, and Ovid were searched for relevant literature. Majority (18) of studies focused on the gut microbiota, and far fewer examined the oral cavity (3), vagina (3), and placenta (1). One study examined the gut, oral, and vaginal microbiotas. The consensus highlights a potential role for microbiota dysbiosis in preeclampsia and HDP. Especially in the third trimester, preeclampsia is associated with gut dysbiosis-deficient in beneficial species of Akkermansia, Bifidobacterium, and Coprococcus but enriched with pathogenic Campylobacterota and Candidatus Saccharibacteria, with low community α-diversity. Similarly, the preeclamptic vaginal and oral microbiotas are enriched with bacterial vaginosis and periodontal disease-associated species, respectively. The trend is also observed in the placenta, which is colonized by gastrointestinal, respiratory tract, and periodontitis-related pathogens. Consequently, a chronic proinflammatory state that adversely impacts placentation is implicated. These observations however require more mechanistic studies to establish the timing of the preceding immune dysfunction and any causality.
Collapse
Affiliation(s)
- Madeleine M. Jordan
- Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
| | - Emmanuel Amabebe
- Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
| | - Kamil Khanipov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Brandie DePaoli Taylor
- Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
- Department of Population Health and Health Disparities, School of Public and Population Health, Galveston, TX, USA
| |
Collapse
|
8
|
González-Rojas A, Valencia-Narbona M. Neurodevelopmental Disruptions in Children of Preeclamptic Mothers: Pathophysiological Mechanisms and Consequences. Int J Mol Sci 2024; 25:3632. [PMID: 38612445 PMCID: PMC11012011 DOI: 10.3390/ijms25073632] [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: 01/24/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Preeclampsia (PE) is a multisystem disorder characterized by elevated blood pressure in the mother, typically occurring after 20 weeks of gestation and posing risks to both maternal and fetal health. PE causes placental changes that can affect the fetus, particularly neurodevelopment. Its key pathophysiological mechanisms encompass hypoxia, vascular and angiogenic dysregulation, inflammation, neuronal and glial alterations, and disruptions in neuronal signaling. Animal models indicate that PE is correlated with neurodevelopmental alterations and cognitive dysfunctions in offspring and in humans, an association between PE and conditions such as cerebral palsy, autism spectrum disorder, attention deficit hyperactivity disorder, and sexual dimorphism has been observed. Considering the relevance for mothers and children, we conducted a narrative literature review to describe the relationships between the pathophysiological mechanisms behind neurodevelopmental alterations in the offspring of PE mothers, along with their potential consequences. Furthermore, we emphasize aspects pertinent to the prevention/treatment of PE in pregnant mothers and alterations observed in their offspring. The present narrative review offers a current, complete, and exhaustive analysis of (i) the pathophysiological mechanisms that can affect neurodevelopment in the children of PE mothers, (ii) the relationship between PE and neurological alterations in offspring, and (iii) the prevention/treatment of PE.
Collapse
Affiliation(s)
- Andrea González-Rojas
- Laboratorio de Neurociencias Aplicadas, Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Avenida Brasil 2950, Valparaíso 2340025, Chile;
| | | |
Collapse
|
9
|
Flores-Pliego A, Espejel-Nuñez A, Borboa-Olivares H, Parra-Hernández SB, Montoya-Estrada A, González-Márquez H, González-Camarena R, Estrada-Gutierrez G. Regulation of MMP-2 by IL-8 in Vascular Endothelial Cells: Probable Mechanism for Endothelial Dysfunction in Women with Preeclampsia. Int J Mol Sci 2023; 25:122. [PMID: 38203296 PMCID: PMC10778620 DOI: 10.3390/ijms25010122] [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: 11/09/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Endothelial dysfunction (ED) in preeclampsia (PE) results from the convergence of oxidative stress, inflammation, and alterations in extracellular matrix components, affecting vascular tone and permeability. The molecular network leading to ED includes IL-8 and MMP-2. In vitro, IL-8 regulates the concentration and activity of MMP-2 in the trophoblast; this interaction has not been studied in endothelial cells during PE. We isolated human umbilical vein endothelial cells (HUVECs) from women with healthy pregnancies (NP, n = 15) and PE (n = 15). We quantified the intracellular concentration of nitric oxide and reactive oxygen species with colorimetric assays, IL-8 with ELISA, and MMP-2 with zymography and using an ELISA-type system. An IL-8 inhibition assay was used to study the influence of this cytokine on MMP-2 concentration and activity. HUVECs from women with PE showed significantly higher oxidative stress than NP. IL-8 and MMP-2 were found to be significantly elevated in PE HUVECs compared to NP. Inhibition of IL-8 in HUVECs from women with PE significantly decreased the concentration of MMP-2. We demonstrate that IL-8 is involved in the mechanisms of MMP-2 expression in HUVECs from women with PE. Our findings provide new insights into the molecular mechanisms regulating the ED distinctive of PE.
Collapse
Affiliation(s)
- Arturo Flores-Pliego
- Department of Immunobiochemistry, Instituto Nacional de Perinatología, Mexico City 11000, Mexico or (A.F.-P.); (A.E.-N.); (S.B.P.-H.)
- Postgraduate in Experimental Biology, Division of Biological and Health Sciences, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico
| | - Aurora Espejel-Nuñez
- Department of Immunobiochemistry, Instituto Nacional de Perinatología, Mexico City 11000, Mexico or (A.F.-P.); (A.E.-N.); (S.B.P.-H.)
| | - Hector Borboa-Olivares
- Community Interventions Research Branch, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; @inper.gob.mx
| | - Sandra Berenice Parra-Hernández
- Department of Immunobiochemistry, Instituto Nacional de Perinatología, Mexico City 11000, Mexico or (A.F.-P.); (A.E.-N.); (S.B.P.-H.)
| | - Araceli Montoya-Estrada
- Coordination of Gynecological and Perinatal Endocrinology, Instituto Nacional de Perinatología, Mexico City 11000, Mexico;
| | - Humberto González-Márquez
- Health Science Department, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico; (H.G.-M.); (R.G.-C.)
| | - Ramón González-Camarena
- Health Science Department, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico; (H.G.-M.); (R.G.-C.)
| | | |
Collapse
|
10
|
Ullah A, Zhao J, Singla RK, Shen B. Pathophysiological impact of CXC and CX3CL1 chemokines in preeclampsia and gestational diabetes mellitus. Front Cell Dev Biol 2023; 11:1272536. [PMID: 37928902 PMCID: PMC10620730 DOI: 10.3389/fcell.2023.1272536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Diabetes-related pathophysiological alterations and various female reproductive difficulties were common in pregnant women with gestational diabetes mellitus (GDM), who had 21.1 million live births. Preeclampsia (PE), which increases maternal and fetal morbidity and mortality, affects approximately 3%-5% of pregnancies worldwide. Nevertheless, it is unclear what triggers PE and GDM to develop. Therefore, the development of novel moderator therapy approaches is a crucial advancement. Chemokines regulate physiological defenses and maternal-fetal interaction during healthy and disturbed pregnancies. Chemokines regulate immunity, stem cell trafficking, anti-angiogenesis, and cell attraction. CXC chemokines are usually inflammatory and contribute to numerous reproductive disorders. Fractalkine (CX3CL1) may be membrane-bound or soluble. CX3CL1 aids cell survival during homeostasis and inflammation. Evidence reveals that CXC and CX3CL1 chemokines and their receptors have been the focus of therapeutic discoveries for clinical intervention due to their considerable participation in numerous biological processes. This review aims to give an overview of the functions of CXC and CX3CL1 chemokines and their receptors in the pathophysiology of PE and GDM. Finally, we examined stimulus specificity for CXC and CX3CL1 chemokine expression and synthesis in PE and GDM and preclinical and clinical trials of CXC-based PE and GDM therapies.
Collapse
Affiliation(s)
- Amin Ullah
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zhao
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Kosińska-Kaczyńska K, Rebizant B, Czeszko-Paprocka H, Bojdo A, Przybylski M, Chaberek K, Lewandowska A, Szymusik I, Brawura-Biskupski-Samaha R. Maternal SARS-CoV-2 Infection at Delivery Increases IL-6 Concentration in Umbilical Cord Blood. J Clin Med 2023; 12:5672. [PMID: 37685739 PMCID: PMC10488671 DOI: 10.3390/jcm12175672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infection in pregnant women may induce inflammation within the amniotic cavity and/or an increase in proinflammatory cytokines in fetal circulation. The aim was to investigate levels of IL-6 in maternal blood, umbilical cord blood, and amniotic fluid in pregnant women infected with SARS-CoV-2 at delivery. METHODS A single-center prospective observational case-control study of pregnant women diagnosed with SARS-CoV-2 infection at delivery was conducted. A total of 48 infected and 42 healthy women had IL-6 concentrations measured in their blood, amniotic fluid, and umbilical cord blood. RESULTS The concentrations of IL-6 in maternal blood and amniotic fluid were similar in the study and control groups, while umbilical cord blood concentrations were significantly higher in SARS-CoV-2-positive women. The umbilical cord blood IL-6 concentration was related to composite neonatal morbidity. CONCLUSIONS Maternal SARS-CoV-2 infection in pregnant women at delivery increases umbilical cord blood IL-6 concentration. The correlation between maternal and umbilical blood concentrations indicates a possibility of passage of IL-6 through the placenta. Perinatal alterations resulting from maternal SARS-CoV-2 infection at delivery carry a risk of impacting the health of infants even in asymptomatic course of infection.
Collapse
Affiliation(s)
- Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| | - Beata Rebizant
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| | - Hanna Czeszko-Paprocka
- Central Analytical Laboratory, Warsaw Infectious Diseases Hospital, 01-201 Warsaw, Poland;
| | - Agata Bojdo
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| | - Maciej Przybylski
- Department of Medical Microbiology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Katarzyna Chaberek
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| | - Agnieszka Lewandowska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| | - Robert Brawura-Biskupski-Samaha
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (K.K.-K.); (A.B.); (K.C.); (A.L.); (I.S.); (R.B.-B.-S.)
| |
Collapse
|
12
|
Jha A, Baumann N, Shadid I, Shah J, Chen YCS, Lee-Sarwar KA, Zeiger RS, O'Connor GT, Bacharier LB, Carey VJ, Laranjo N, Fichorova RN, Litonjua AA, Weiss ST, Mirzakhani H. The relationship of fetal sex and maternal race and ethnicity with early and late pregnancy C-reactive protein and interleukin-8. Am J Reprod Immunol 2023; 90:e13746. [PMID: 37491932 DOI: 10.1111/aji.13746] [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: 09/05/2022] [Revised: 03/11/2023] [Accepted: 06/22/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Promotion of a healthy pregnancy is dependent on a coordinated immune response that minimizes inflammation at the maternal-fetal interface. Few studies investigated the effect of fetal sex on proinflammatory biomarkers during pregnancy and whether maternal race could impact this association. We aimed to examine whether fetal sex could, independently of maternal race/ethnicity and the condition of pregnancy (normal vs. complicated), impact inflammatory markers (C-reactive protein [CRP] and interleukin-8 [IL-8] levels) in early and late pregnancy. METHODS OF STUDY This study was a cohort analysis using prospectively collected data from pregnant women who participated in the Vitamin Antenatal Asthma Reduction Trial (VDAART, N = 816). Maternal serum CRP and IL-8 levels were measured in early and late pregnancy (10-18 and 32-38 weeks of gestation, respectively). Five hundred and twenty-eight out of 816 pregnant women who participated in the trial had available CRP and IL-8 measurements at both study time points. We examined the association of fetal sex with early and late CRP and IL-8 levels and their paired sample difference. We further investigated whether maternal race/ethnicity, pregnancy complications (i.e., preeclampsia and gestational diabetes), and early pregnancy body mass index (BMI) could affect the association between these two biomarkers and fetal sex adjusting for potential confounders. For this purpose, we used generalized linear and logistic regression models on log-normalized early and late CRP and IL-8 levels as well as their split at median to form high and low groups. RESULTS Women pregnant with male fetuses (266/528 = 56.5%) had higher CRP levels in early to mid-pregnancy (β = .18: 95% confidence interval [CI]: CI = 0.03-0.32; p = .02). Twenty-seven percent (143/528) of the study subjects were Hispanic. Hispanic African American [AA] women and women of races other than White and AA had higher levels of CRP at early to mid-pregnancy compared with White women (β = .57; 95% CI: 0.17-0.97; p < .01 and β = .27; 95% CI: 0.05-0.48; p = .02, respectively). IL-8 levels were not associated with fetal sex in early and late pregnancy (p's > .05). Other factors such as gestational diabetes and early pregnancy BMI were associated with higher CRP levels and higher CRP and IL-8 levels, respectively. Dichotomizing log-normalized cytokine levels at the median in a sensitivity analysis, women with male fetuses had lower odds of high (above-median) IL-8 levels at early pregnancy. Also, women with races other than AA and White carrying male fetuses had higher odds of having high (above-median) late-pregnancy CRP and early-pregnancy IL-8 levels (adjusted odds ratio [aOR] = 3.80, 95% CI: 0.24-1.23; p = .02 and aOR = 3.57; 95% CI: 0.23-1.03; p = .02, respectively). Of the pregnancy complications, women with gestational diabetes mellitus had a higher paired difference of early and late pregnancy CRP levels (β = .38; 95% CI: 0.09-0.68; p = .01), but no difference in IL-8 levels (p's > .05). No associations between the inflammatory markers and preeclampsia were found. CONCLUSION Fetal sex is associated with CRP in early pregnancy and an association with IL-8 in early pregnancy is implied. Our study further indicates that maternal race/ethnicity could be a contributing factor in the relationship between fetal sex and inflammatory responses during pregnancy. However, the specificity and level of the contribution might vary by type of cytokine, pregnancy stage, and other confounding factors such as BMI that may impact these associations.
Collapse
Affiliation(s)
- Anjali Jha
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Noah Baumann
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Iskander Shadid
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jhill Shah
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Pulmonary Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yih-Chieh S Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen A Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert S Zeiger
- Department of Clinical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California, USA
| | - George T O'Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Leonard B Bacharier
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Vincent J Carey
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Nancy Laranjo
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital at Strong, University of Rochester Medical Center, Rochester, New York, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hooman Mirzakhani
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Zhou C, Freel C, Mills O, Yang XR, Yan Q, Zheng J. MicroRNA-29 differentially mediates preeclampsia-dysregulated cellular responses to cytokines in female and male fetal endothelial cells. J Physiol 2023; 601:3631-3645. [PMID: 37401732 PMCID: PMC10807859 DOI: 10.1113/jp284746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023] Open
Abstract
Preeclampsia (PE) differentially impairs female and male fetal endothelial cell function, which is associated with an increased risk of adult-onset cardiovascular disorders in children born to mothers with PE. However, the underlying mechanisms are poorly defined. We hypothesize that dysregulation of microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in PE disturbs gene expression and cellular responses to cytokines in fetal endothelial cells in a fetal sex-dependent manner. RT-qPCR analysis of miR-29a/c-3p was performed on female and male unpassaged (P0) human umbilical vein endothelial cells (HUVECs) from normotensive (NT) pregnancies and PE. Bioinformatic analysis of an RNA-seq dataset was performed to identify PE-dysregulated miR-29a/c-3p target genes in female and male P0-HUVECs. Gain- and loss-of-function assays were conducted to determine the effects of miR-29a/c-3p on endothelial monolayer integrity and proliferation in response to transforming growth factor-β1 (TGFβ1) and tumour necrosis factor-α (TNFα) in NT and PE HUVECs at passage 1. We observed that PE downregulated miR-29a/c-3p in male and female P0-HUVECs. PE dysregulated significantly more miR-29a/c-3p target genes in female vs. male P0-HUVECs. Many of these PE-differentially dysregulated miR-29a/c-3p target genes are associated with critical cardiovascular diseases and endothelial function. We further demonstrated that miR-29a/c-3p knockdown specifically recovered the PE-abolished TGFβ1-induced strengthening of endothelial monolayer integrity in female HUVECs, while miR-29a/c-3p overexpression specifically enhanced the TNFα-promoted cell proliferation in male PE HUVECs. In conclusion, PE downregulates miR-29a/c-3p expression and differentially dysregulates miR-29a/c-3p target genes associated with cardiovascular diseases and endothelial function in female and male fetal endothelial cells, possibly contributing to the fetal sex-specific endothelial dysfunction observed in PE. KEY POINTS: Preeclampsia differentially impairs female and male fetal endothelial cell function in responses to cytokines. Pro-inflammatory cytokines are elevated in maternal circulation during pregnancy in preeclampsia. MicroRNAs are critical regulators of endothelial cell function during pregnancy. We have previously reported that preeclampsia downregulated microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in primary fetal endothelial cells. However, it is unknown if PE differentially dysregulates the expression of miR-29a/c-3p in female and male fetal endothelial cells. We show that preeclampsia downregulates miR-29a/c-3p in male and female HUVECs and preeclampsia dysregulates cardiovascular disease- and endothelial function-associated miR-29a/c-3p target genes in HUVECs in a fetal sex-specific manner. MiR-29a/c-3p differentially mediate cell responses to cytokines in female and male fetal endothelial cells from preeclampsia. We have revealed fetal sex-specific dysregulation of miR-29a/c-3p target genes in fetal endothelial cells from preeclampsia. This differential dysregulation may contribute to fetal sex-specific endothelial dysfunction in offspring born to preeclamptic mothers.
Collapse
Affiliation(s)
- Chi Zhou
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
- Department of Obstetrics and Gynecology, the University of Arizona, Tucson, AZ, United States
| | - Colman Freel
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Current Institution: University of Nebraska Medical Center, Omaha, NE, United States
| | - Olivia Mills
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
| | - Xin-Ran Yang
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
| | - Qin Yan
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
14
|
Mukherjee I, Singh S, Karmakar A, Kashyap N, Mridha AR, Sharma JB, Luthra K, Sharma RS, Biswas S, Dhar R, Karmakar S. New immune horizons in therapeutics and diagnostic approaches to Preeclampsia. Am J Reprod Immunol 2023; 89:e13670. [PMID: 36565013 DOI: 10.1111/aji.13670] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/02/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are one of the commonest maladies, affecting 5%-10% of pregnancies worldwide. The American College of Obstetricians and Gynecologists (ACOG) identifies four categories of HDP, namely gestational hypertension (GH), Preeclampsia (PE), chronic hypertension (CH), and CH with superimposed PE. PE is a multisystem, heterogeneous disorder that encompasses 2%-8% of all pregnancy-related complications, contributing to about 9% to 26% of maternal deaths in low-income countries and 16% in high-income countries. These translate to 50 000 maternal deaths and over 500 000 fetal deaths worldwide, therefore demanding high priority in understanding clinical presentation, screening, diagnostic criteria, and effective management. PE is accompanied by uteroplacental insufficiency leading to vascular and metabolic changes, vasoconstriction, and end-organ ischemia. PE is diagnosed after 20 weeks of pregnancy in women who were previously normotensive or hypertensive. Besides shallow trophoblast invasion and inadequate remodeling of uterine arteries, dysregulation of the nonimmune system has been the focal point in PE. This results from aberrant immune system activation and imbalanced differentiation of T cells. Further, a failure of tolerance toward the semi-allogenic fetus results due to altered distribution of Tregs such as CD4+FoxP3+ or CD4+CD25+CD127(low) FoxP3+ cells, thereby creating a cytotoxic environment by suboptimal production of immunosuppressive cytokines like IL-10, IL-4, and IL-13. Also, intracellular production of complement protein C5a may result in decreased FoxP3+ regulatory T cells. With immune system dysfunction as a major driver in PE pathogenesis, it is logical that therapeutic targeting of components of the immune system with pharmacologic agents like anti-inflammatory and immune-modulating molecules are either being used or under clinical trial. Cholesterol synthesis inhibitors like Pravastatin may improve placental perfusion in PE, while Eculizumab (monoclonal antibody inhibiting C5) and small molecular inhibitor of C5a, Zilucoplan are under investigation. Monoclonal antibody against IL-17(Secukinumab) has been proposed to alter the Th imbalance in PE. Autologous Treg therapy and immune checkpoint inhibitors like anti-CTLA-4 are emerging as new candidates in immune horizons for PE management in the future.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.,Amity Institute of Biotechnology (AIB), Amity University, Noida, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Abhibrato Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neha Kashyap
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Asit Ranjan Mridha
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Radhey Shyam Sharma
- Ex-Head and Scientist G, Indian Council of Medical Research, New Delhi, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Noida, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
15
|
Barron A, Manna S, McElwain CJ, Musumeci A, McCarthy FP, O’Keeffe GW, McCarthy CM. Maternal pre-eclampsia serum increases neurite growth and mitochondrial function through a potential IL-6-dependent mechanism in differentiated SH-SY5Y cells. Front Physiol 2023; 13:1043481. [PMID: 36714304 PMCID: PMC9877349 DOI: 10.3389/fphys.2022.1043481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction: Pre-eclampsia (PE) is a common and serious hypertensive disorder of pregnancy, which affects 3%-5% of first-time pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Prenatal exposure to PE is associated with an increased risk of neurodevelopmental disorders in affected offspring, although the cellular and molecular basis of this increased risk is largely unknown. Methods: Here, we examined the effects of exposure to maternal serum from women with PE or a healthy uncomplicated pregnancy on the survival, neurite growth and mitochondrial function of neuronally differentiated human SH-SY5Y neuroblastoma cells, which are commonly used to study neurite growth. Neurite growth and mitochondrial function are two strongly linked neurodevelopmental parameters in which alterations have been implicated in neurodevelopmental disorders. Following this, we investigated the pleiotropic cytokine interleukin-6 (IL-6) levels as a potential mechanism. Results: Cells exposed to 3% (v/v) PE serum for 72 h exhibited increased neurite growth (p < 0.05), which was validated in the human neural progenitor cell line, ReNcell® VM (p < 0.01), and mitochondrial respiration (elevated oxygen consumption rate (p < 0.05), basal mitochondrial respiration, proton leak, ATP synthesis, and non-mitochondrial respiration) compared to control serum-treated cells. ELISA analysis showed elevations in maternal IL-6 in PE sera (p < 0.05) and placental explants (p < 0.05). In support of this, SH-SY5Y cells exposed to 3% (v/v) PE serum for 24 h had increased phospho-STAT3 levels, which is a key intracellular mediator of IL-6 signalling (p < 0.05). Furthermore, treatment with anti-IL-6 neutralizing antibody blocked the effects of PE serum on neurite growth (p < 0.05), and exposure to IL-6 promoted neurite growth in SH-SY5Y cells (p < 0.01). Discussion: Collectively these data show elevated serum levels of maternal IL-6 in PE, which increases neurite growth and mitochondrial function in SH-SY5Y cells. This rationalizes the further study of IL-6 as a potential mediator between PE exposure and neurodevelopmental outcome in the offspring.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Samprikta Manna
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Colm J. McElwain
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Andrea Musumeci
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Gerard W. O’Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Cork Neuroscience Centre, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| |
Collapse
|
16
|
Adenekan MA, Oluwole AA, Olorunfemi G, Sekumade AI, Ajepe AA, Okunade KS. Maternal tumour necrosis factor-alpha levels in preeclamptic pregnancies in Lagos, South-West Nigeria. Pregnancy Hypertens 2022; 30:198-203. [PMID: 36323062 PMCID: PMC9839453 DOI: 10.1016/j.preghy.2022.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This study aimed to assess the association between maternal serum levels of TNF-α and preeclampsia. METHODS An analytical cross-sectional study involving 45 women diagnosed with preeclampsia and 45 healthy normotensive pregnant women matched for age, and gestational age at enrolment. Venous samples were collected from each participant after informed consent was obtained. Serum TNF-α level was determined using the human TNF-α competitive enzyme-linked immunosorbent assay (ELISA) technique with ELISA Kit. Hypothesis testing was done using the Chi-square test for categorical variables, the independent samples t-test and the Kruskal-Wallis test for numerical variables. All significances were reported at P < 0.05. RESULTS The median concentrations of TNF-α in women with preeclampsia of varying severity were significantly higher than those with normotensive pregnancies (P = 0.001). The median level of TNF-α was also significantly higher in patients with severe features of preeclampsia than in those without. The estimated cut-off levels of serum TNF-α were 15.6 ng/mL and 26.4 ng/mL respectively for the development and severity of preeclampsia. Maternal serum TNF-α level in preeclamptic patients is strongly correlated with systolic and diastolic BP, serum uric acid and alkaline phosphatase levels, proteinuria, and platelet count (P < 0.05). CONCLUSION We can infer from this study that increased maternal serum levels of TNF-α may play a significant role in the pathogenesis of preeclampsia. We recommend further validation of these findings with a more robust longitudinal characterization of maternal serum TNF-α profiles in pregnancy through a well-designed prospective cohort study.
Collapse
Affiliation(s)
- Muisi A Adenekan
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Ayodeji A Oluwole
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Lagos, Nigeria; Department of Obstetrics and Gynaecology, College of Medicine, University of Lagos, Nigeria
| | - Gbenga Olorunfemi
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Lagos, Nigeria; Division of Epidemiology and Biostatistics, School of Public Health, University of Witwatersrand, Johannesburg, South Africa
| | - Adebayo I Sekumade
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Adegbenga A Ajepe
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Kehinde S Okunade
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Lagos, Nigeria; Department of Obstetrics and Gynaecology, College of Medicine, University of Lagos, Nigeria.
| |
Collapse
|
17
|
Katoh Y, Iriyama T, Yano E, Sayama S, Seyama T, Kotajima-Murakami H, Sato A, Sakuma H, Iguchi Y, Yoshikawa M, Inaoka N, Ichinose M, Toshimitsu M, Sone K, Kumasawa K, Nagamatsu T, Ikeda K, Osuga Y. Increased production of inflammatory cytokines and activation of microglia in the fetal brain of preeclamptic mice induced by angiotensin II. J Reprod Immunol 2022; 154:103752. [PMID: 36202022 DOI: 10.1016/j.jri.2022.103752] [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: 06/27/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/14/2022]
Abstract
Preeclampsia (PE) is a hypertensive obstetric disorder with poor prognosis for both the mother and offspring. Infants born to mothers with PE are known to be at increased risk of developing higher brain dysfunction, such as autism. However, how maternal PE can affect the environment in the fetal brain has not been fully elucidated. Here, we examined the impact of PE on the fetal brain in a mouse model of PE induced by angiotensin II (Ang II), focusing on changes in the inflammatory condition. We confirmed that pregnant mice which were continuously administered Ang II exhibited PE phenotypes, including high blood pressure, proteinuria, and fetal growth restriction. Quantitative RT-PCR analysis demonstrated that the brain of fetuses on embryonic day 17.5 (E17.5) in the Ang II-administered pregnant mice showed increased expression of cytokines, interleukin (IL)- 6, IL-17a, tumor necrosis factor-α, interferon-γ, IL-12, IL-4, and IL-10. Immunohistochemical analysis over a wide area, from the tip of the frontal lobe to the posterior cerebral end, on E17.5 revealed that the microglia in the fetal brain of the Ang II-administered group displayed higher solidity and circularity than those of the control group, indicating that the microglia had transformed to an amoeboid morphology and were activated. Our findings suggest that maternal PE may cause altered inflammatory conditions in the fetal brain, which might be associated with the pathological mechanism connecting maternal PE and brain dysfunction in the offspring.
Collapse
Affiliation(s)
- Yoshihisa Katoh
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan; Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan.
| | - Eriko Yano
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Seisuke Sayama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Takahiro Seyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | | | - Atsushi Sato
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan
| | - Hiroshi Sakuma
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoshinobu Iguchi
- Technology Research Division, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Midori Yoshikawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Naoko Inaoka
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Mari Ichinose
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Masatake Toshimitsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Keiichi Kumasawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Vilotić A, Nacka-Aleksić M, Pirković A, Bojić-Trbojević Ž, Dekanski D, Jovanović Krivokuća M. IL-6 and IL-8: An Overview of Their Roles in Healthy and Pathological Pregnancies. Int J Mol Sci 2022; 23:ijms232314574. [PMID: 36498901 PMCID: PMC9738067 DOI: 10.3390/ijms232314574] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Interleukin-6 (IL-6) is an acknowledged inflammatory cytokine with a pleiotropic action, mediating innate and adaptive immunity and multiple physiological processes, including protective and regenerative ones. IL-8 is a pro-inflammatory CXC chemokine with a primary function in attracting and activating neutrophils, but also implicated in a variety of other cellular processes. These two ILs are abundantly expressed at the feto-maternal interface over the course of a pregnancy and have been shown to participate in numerous pregnancy-related events. In this review, we summarize the literature data regarding their role in healthy and pathological pregnancies. The general information related to IL-6 and IL-8 functions is followed by an overview of their overall expression in cycling endometrium and at the feto-maternal interface. Further, we provide an overview of their involvement in pregnancy establishment and parturition. Finally, the implication of IL-6 and IL-8 in pregnancy-associated pathological conditions, such as pregnancy loss, preeclampsia, gestational diabetes mellitus and infection/inflammation is discussed.
Collapse
|
19
|
Saadat N, Zhang L, Hyer S, Padmanabhan V, Woo J, Engeland CG, Misra DP, Giurgescu C. Psychosocial and behavioral factors affecting inflammation among pregnant African American women. Brain Behav Immun Health 2022; 22:100452. [PMID: 35403067 PMCID: PMC8990056 DOI: 10.1016/j.bbih.2022.100452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/12/2022] [Indexed: 01/08/2023] Open
Abstract
African American women are reported to have greater inflammation compared with women from other racial groups. Higher inflammation during pregnancy has been associated with increased risk of adverse perinatal outcomes. We hypothesized that maternal inflammation is related to depressive symptoms and social and behavioral risk factors among pregnant African American women. Pregnant African American women (n = 187) were recruited at prenatal clinics in the Midwest. Women completed questionnaires and had blood drawn at a prenatal visit. Plasma levels of cytokines (interferon gamma [IFN]-γ, interleukin [IL]-6, IL-8, IL-10, tumor necrosis factor [TNF]-α) and C-reactive protein (CRP) were measured by multiplex assays. Women had a mean age of 26.58±5.42 years and a mean gestational age at data collection of 16.35±5.95 weeks. Twenty-six percent of women had Center for Epidemiological Studies-Depression (CES-D) scores ≥23 (scores that have been correlated with clinical diagnosis of depression), 15.5% smoked cigarettes, 16.6% used marijuana, and 5.3% reported experiencing intimate partner violence (IPV). Higher CES-D scores were correlated with higher plasma CRP levels (r = 0.16, p = 0.046). Women who reported any experiences of IPV during pregnancy had higher levels of IL-8 (p = 0.018) and lower levels of IFN-γ (p = 0.012) compared with women who did not report IPV. Cigarette smoking during pregnancy was associated with lower levels of the anti-inflammatory cytokine IL-10 (p = 0.003). These findings suggest that depressive symptoms, IPV, and cigarette smoking during pregnancy relate to select inflammatory markers in pregnant African American women. The relationships of inflammation with these factors should be further investigated to better understand the mechanisms which influence maternal and fetal health outcomes. Depressive symptoms are related to higher levels of CRP in pregnant African American Women. Intimate partner violence in pregnancy is related to higher interleukin 8 levels. Smoking during pregnancy is associated with lower interleukin 10 levels. Inflammation is related to depressive symptoms, intimate partner violence and smoking in pregnant African American women.
Collapse
|
20
|
Ozmen A, Guzeloglu-Kayisli O, Tabak S, Guo X, Semerci N, Nwabuobi C, Larsen K, Wells A, Uyar A, Arlier S, Wickramage I, Alhasan H, Totary-Jain H, Schatz F, Odibo AO, Lockwood CJ, Kayisli UA. Preeclampsia is Associated With Reduced ISG15 Levels Impairing Extravillous Trophoblast Invasion. Front Cell Dev Biol 2022; 10:898088. [PMID: 35837332 PMCID: PMC9274133 DOI: 10.3389/fcell.2022.898088] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2023] Open
Abstract
Among several interleukin (IL)-6 family members, only IL-6 and IL-11 require a gp130 protein homodimer for intracellular signaling due to lack of intracellular signaling domain in the IL-6 receptor (IL-6R) and IL-11R. We previously reported enhanced decidual IL-6 and IL-11 levels at the maternal-fetal interface with significantly higher peri-membranous IL-6 immunostaining in adjacent interstitial trophoblasts in preeclampsia (PE) vs. gestational age (GA)-matched controls. This led us to hypothesize that competitive binding of these cytokines to the gp130 impairs extravillous trophoblast (EVT) differentiation, proliferation and/or invasion. Using global microarray analysis, the current study identified inhibition of interferon-stimulated gene 15 (ISG15) as the only gene affected by both IL-6 plus IL-11 vs. control or IL-6 or IL-11 treatment of primary human cytotrophoblast cultures. ISG15 immunostaining was specific to EVTs among other trophoblast types in the first and third trimester placental specimens, and significantly lower ISG15 levels were observed in EVT from PE vs. GA-matched control placentae (p = 0.006). Induction of primary trophoblastic stem cell cultures toward EVT linage increased ISG15 mRNA levels by 7.8-fold (p = 0.004). ISG15 silencing in HTR8/SVneo cultures, a first trimester EVT cell line, inhibited invasion, proliferation, expression of ITGB1 (a cell migration receptor) and filamentous actin while increasing expression of ITGB4 (a receptor for hemi-desmosomal adhesion). Moreover, ISG15 silencing further enhanced levels of IL-1β-induced pro-inflammatory cytokines (CXCL8, IL-6 and CCL2) in HTR8/SVneo cells. Collectively, these results indicate that ISG15 acts as a critical regulator of EVT morphology and function and that diminished ISG15 expression is associated with PE, potentially mediating reduced interstitial trophoblast invasion and enhancing local inflammation at the maternal-fetal interface. Thus, agents inducing ISG15 expression may provide a novel therapeutic approach in PE.
Collapse
Affiliation(s)
- Asli Ozmen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ozlem Guzeloglu-Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Selcuk Tabak
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Xiaofang Guo
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Chinedu Nwabuobi
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Kellie Larsen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ali Wells
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Asli Uyar
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States
| | - Sefa Arlier
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ishani Wickramage
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Hasan Alhasan
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Hana Totary-Jain
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Frederick Schatz
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Anthony O. Odibo
- Divisions of Maternal-Fetal Medicine and Ultrasound, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, United States
| | - Charles J. Lockwood
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Umit A. Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States,*Correspondence: Umit A. Kayisli,
| |
Collapse
|
21
|
Gomez-Lopez N, Romero R, Tao L, Gershater M, Leng Y, Zou C, Farias-Jofre M, Galaz J, Miller D, Tarca AL, Arenas-Hernandez M, Bhatti G, Garcia-Flores V, Liu Z, Para R, Kanninen T, Hadaya O, Paredes C, Xu Y. Distinct Cellular Immune Responses to SARS-CoV-2 in Pregnant Women. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1857-1872. [PMID: 35379748 PMCID: PMC9180665 DOI: 10.4049/jimmunol.2101123] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/08/2022] [Indexed: 12/11/2022]
Abstract
Pregnant women are at increased risk of adverse outcomes, including preeclampsia and preterm birth, that may result from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Pregnancy imprints specific maternal immune responses that can modulate host susceptibility to microbial infection; therefore, recent studies have focused on the humoral response against SARS-CoV-2 in pregnant women. However, the pregnancy-specific cellular immune responses triggered by SARS-CoV-2 infection are poorly understood. In this study, we undertook an extensive in vitro investigation to determine the cellular immune responses to SARS-CoV-2 particles and proteins/peptides in pregnant women. First, we show that SARS-CoV-2 particles do not alter the pregnancy-specific oxidative burst of neutrophils and monocytes. Yet, SARS-CoV-2 particles/proteins shift monocyte activation from the classical to intermediate states in pregnant, but not in nonpregnant, women. Furthermore, SARS-CoV-2 proteins, but not particles or peptide pools, mildly enhance T cell activation during pregnancy. As expected, B cell phenotypes are heavily modulated by SARS-CoV-2 particles in all women; yet, pregnancy itself further modified such responses in these adaptive immune cells. Lastly, we report that pregnancy itself governs cytokine responses in the maternal circulation, of which IFN-β and IL-8 were diminished upon SARS-CoV-2 challenge. Collectively, these findings highlight the differential in vitro responses to SARS-CoV-2 in pregnant and nonpregnant women and shed light on the immune mechanisms implicated in coronavirus disease 2019 during pregnancy.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI;
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI;
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
- Detroit Medical Center, Detroit, MI; and
| | - Li Tao
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Meyer Gershater
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Chengrui Zou
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Marcelo Farias-Jofre
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Zhenjie Liu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tomi Kanninen
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Ola Hadaya
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Carmen Paredes
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
22
|
Negi R, Haritha V, Aziz N, Siddiqui AH. Biochemical markers in the pathogenesis of preeclampsia: novel link between placental growth factor and interleukin-6. Mol Cell Biochem 2022; 477:1765-1774. [PMID: 35292877 DOI: 10.1007/s11010-022-04403-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Abstract
Preeclampsia (PE) is a multisystem disorder of pregnancy characterized by sudden onset of hypertension and proteinuria. The appearance and diagnosis of the disease remain elusive and the only treatment is the termination of pregnancy. The onset of the disease is preceded by a shift in the balance of the angiogenic and antiangiogenic proteins in the maternal circulation. We surmised that the assessment of the levels of these proteins during pregnancy could lead to a proper diagnosis of the disease. In this study, we determined the levels of angiogenic and antiangiogenic proteins and IL-6 in maternal circulation during normotensive and hypertensive pregnancy, including PE. Serum isolated from pregnant women during antenatal visits was used to determine the concentrations of these proteins. The levels of antiangiogenic proteins, sFlt-1 and sEng, were higher in hypertensive disorders [gestational hypertension (GH), mild PE, and PE] of pregnancy and were significantly higher for PE than for GH. The levels of sFlt-1 and sEng were higher in PE samples compared to those in GH and NT samples. These proteins may have contributed to increased blood pressure. The levels of PlGF were decreased in pregnant women having GH, mild PE, and PE. The levels of the inflammatory intermediate, IL-6, were increased in PE samples compared to those in the GH and normotensive samples. The evaluation of the altered levels of antiangiogenic and angiogenic proteins can be useful for diagnosis of PE.
Collapse
Affiliation(s)
- Rahul Negi
- School of Medical Sciences, University of Hyderabad, Professor C. R. Rao Road, Gachibowli, Hyderabad, TS, 500046, India
| | - Vemanamanda Haritha
- Fernandez Hosp Educational & Research Foundation, Bogulkunta, Hyderabad, TS, 500001, India
| | - Nuzhat Aziz
- Fernandez Hosp Educational & Research Foundation, Bogulkunta, Hyderabad, TS, 500001, India
| | - Athar H Siddiqui
- School of Medical Sciences, University of Hyderabad, Professor C. R. Rao Road, Gachibowli, Hyderabad, TS, 500046, India.
| |
Collapse
|
23
|
Broekhuizen M, Hitzerd E, van den Bosch TPP, Dumas J, Verdijk RM, van Rijn BB, Danser AHJ, van Eijck CHJ, Reiss IKM, Mustafa DAM. The Placental Innate Immune System Is Altered in Early-Onset Preeclampsia, but Not in Late-Onset Preeclampsia. Front Immunol 2022; 12:780043. [PMID: 34992598 PMCID: PMC8724430 DOI: 10.3389/fimmu.2021.780043] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia is a severe placenta-related pregnancy disorder that is generally divided into two subtypes named early-onset preeclampsia (onset <34 weeks of gestation), and late-onset preeclampsia (onset ≥34 weeks of gestation), with distinct pathophysiological origins. Both forms of preeclampsia have been associated with maternal systemic inflammation. However, alterations in the placental immune system have been less well characterized. Here, we studied immunological alterations in early- and late-onset preeclampsia placentas using a targeted expression profile approach. RNA was extracted from snap-frozen placenta samples (healthy n=13, early-onset preeclampsia n=13, and late-onset preeclampsia n=6). The expression of 730 immune-related genes from the Pan Cancer Immune Profiling Panel was measured, and the data were analyzed in the advanced analysis module of nSolver software (NanoString Technology). The results showed that early-onset preeclampsia placentas displayed reduced expression of complement, and toll-like receptor (TLR) associated genes, specifically TLR1 and TLR4. Mast cells and M2 macrophages were also decreased in early-onset preeclampsia compared to healthy placentas. The findings were confirmed by an immunohistochemistry approach using 20 healthy, 19 early-onset preeclampsia, and 10 late-onset preeclampsia placentas. We conclude that the placental innate immune system is altered in early-onset preeclampsia compared to uncomplicated pregnancies. The absence of these alterations in late-onset preeclampsia placentas indicates dissimilar immunological profiles. The study revealed distinct pathophysiological processes in early-onset and late-onset preeclampsia placentas and imply that a tailored treatment to each subtype is desirable.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Emilie Hitzerd
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Jasper Dumas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands.,The Tumor Immuno-Pathology (TIP) Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bas B van Rijn
- Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands.,The Tumor Immuno-Pathology (TIP) Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
24
|
Amelio GS, Provitera L, Raffaeli G, Tripodi M, Amodeo I, Gulden S, Cortesi V, Manzoni F, Cervellini G, Tomaselli A, Pravatà V, Garrido F, Villamor E, Mosca F, Cavallaro G. Endothelial dysfunction in preterm infants: The hidden legacy of uteroplacental pathologies. Front Pediatr 2022; 10:1041919. [PMID: 36405831 PMCID: PMC9671930 DOI: 10.3389/fped.2022.1041919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Millions of infants are born prematurely every year worldwide. Prematurity, particularly at lower gestational ages, is associated with high mortality and morbidity and is a significant global health burden. Pregnancy complications and preterm birth syndrome strongly impact neonatal clinical phenotypes and outcomes. The vascular endothelium is a pivotal regulator of fetal growth and development. In recent years, the key role of uteroplacental pathologies impairing endothelial homeostasis is emerging. Conditions leading to very and extremely preterm birth can be classified into two main pathophysiological patterns or endotypes: infection/inflammation and dysfunctional placentation. The first is frequently related to chorioamnionitis, whereas the second is commonly associated with hypertensive disorders of pregnancy and fetal growth restriction. The nature, timing, and extent of prenatal noxa may alter fetal and neonatal endothelial phenotype and functions. Changes in the luminal surface, oxidative stress, growth factors imbalance, and dysregulation of permeability and vascular tone are the leading causes of endothelial dysfunction in preterm infants. However, the available evidence regarding endothelial physiology and damage is limited in neonates compared to adults. Herein, we discuss the current knowledge on endothelial dysfunction in the infectious/inflammatory and dysfunctional placentation endotypes of prematurity, summarizing their molecular features, available biomarkers, and clinical impact. Furthermore, knowledge gaps, shadows, and future research perspectives are highlighted.
Collapse
Affiliation(s)
- Giacomo Simeone Amelio
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Gulden
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Cortesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Francesca Manzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Gaia Cervellini
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Valentina Pravatà
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, Madrid, Spain
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), University of Maastricht, Maastricht, Netherlands
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
25
|
Li X, Liu L, Whitehead C, Li J, Thierry B, Le TD, Winter M. OUP accepted manuscript. Brief Funct Genomics 2022; 21:296-309. [PMID: 35484822 PMCID: PMC9328024 DOI: 10.1093/bfgp/elac006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia is a pregnancy-specific disease that can have serious effects on the health of both mothers and their offspring. Predicting which women will develop preeclampsia in early pregnancy with high accuracy will allow for improved management. The clinical symptoms of preeclampsia are well recognized, however, the precise molecular mechanisms leading to the disorder are poorly understood. This is compounded by the heterogeneous nature of preeclampsia onset, timing and severity. Indeed a multitude of poorly defined causes including genetic components implicates etiologic factors, such as immune maladaptation, placental ischemia and increased oxidative stress. Large datasets generated by microarray and next-generation sequencing have enabled the comprehensive study of preeclampsia at the molecular level. However, computational approaches to simultaneously analyze the preeclampsia transcriptomic and network data and identify clinically relevant information are currently limited. In this paper, we proposed a control theory method to identify potential preeclampsia-associated genes based on both transcriptomic and network data. First, we built a preeclampsia gene regulatory network and analyzed its controllability. We then defined two types of critical preeclampsia-associated genes that play important roles in the constructed preeclampsia-specific network. Benchmarking against differential expression, betweenness centrality and hub analysis we demonstrated that the proposed method may offer novel insights compared with other standard approaches. Next, we investigated subtype specific genes for early and late onset preeclampsia. This control theory approach could contribute to a further understanding of the molecular mechanisms contributing to preeclampsia.
Collapse
Affiliation(s)
- Xiaomei Li
- UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Lin Liu
- UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Clare Whitehead
- Pregnancy Research Centre, Dept of Obstetrics & Gynaecology, University of Melbourne, Royal Women’s Hospital, Melbourne, 3052, VIC, Australia
| | - Jiuyong Li
- UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Thuc D Le
- Corresponding authors: Thuc D. Le, UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail: ; M. Winter, Future Industries Institute, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail:
| | - Marnie Winter
- Corresponding authors: Thuc D. Le, UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail: ; M. Winter, Future Industries Institute, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail:
| |
Collapse
|
26
|
Fragoso MBT, Ferreira RC, Tenório MCDS, Moura FA, de Araújo ORP, Bueno NB, Goulart MOF, de Oliveira ACM. Biomarkers of Inflammation and Redox Imbalance in Umbilical Cord in Pregnancies with and without Preeclampsia and Consequent Perinatal Outcomes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9970627. [PMID: 34795845 PMCID: PMC8595010 DOI: 10.1155/2021/9970627] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/16/2021] [Accepted: 10/11/2021] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To compare redox imbalance and inflammation biomarkers in umbilical cords from pregnancies with and without preeclampsia (PE) and to analyse their relationships with perinatal outcomes. METHODS A controlled cross-sectional study was conducted in Maceió, Alagoas, Brazil, that involved pregnant women with PE and a group of women without the disease, through the application of a standardized questionnaire. After delivery, umbilical cord samples were collected to measure antioxidant defense, products from oxidative damage, and inflammation biomarkers such as myeloperoxidase (MPO), interleukin- (IL-) 6, IL-8, IL-10, and tumor necrosis factor-alpha (TNF-α). Statistical analyses were performed using Stata version 13.0 software and IBM Statistical Package for the Social Sciences (SPSS) 20.0, adopting a 95% confidence level (α = 0.05), with the chi-square test, the Wilcoxon-Mann-Whitney test, and the multinomial and Poisson regression tests. RESULTS One hundred PE pregnant women and 50 women without the disease were studied. The umbilical cords from PE pregnancies showed higher levels of reduced glutathione (GSH) (p ≤ 0.001), glutathione peroxidase (GPx) (p = 0.016), and malondialdehyde (MDA) (p = 0.028) and lower levels of IL-6 (p = 0.030) and TNF-α (p ≤ 0.001) than the other group, with some associations among these biomarkers with perinatal outcomes. CONCLUSION The higher levels of GSH and GPx, in addition to the lower levels of IL-6 and TNF-α, found in the PE umbilical cord, may result from adaptive mechanisms to maintain the oxidative and inflammatory balance; however, despite these changes, the damage to the cell membranes was not minimized, as the MDA level was higher in women with PE than in women without the disease. This implies that a redox imbalance is present, confirming that other physiological and adaptive mechanisms are being activated to preserve foetal health. Therefore, the present work unveils an important role of the umbilical cord in controlling redox imbalance and inflammation in PE pregnancies. Our results reinforce the necessity for continuous research on GSH as a protective compound for the perinatal outcome, especially in PE women.
Collapse
Affiliation(s)
- Marilene Brandão Tenório Fragoso
- Instituto de Química e Biotecnologia (IQB/UFAL), Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Raphaela Costa Ferreira
- Programa de Pós-graduação em Ciências da Saúde, ICBS, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Micaely Cristina dos Santos Tenório
- Instituto de Química e Biotecnologia (IQB/UFAL), Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Fabiana Andréa Moura
- Faculdade de Nutrição, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Orlando Roberto Pimentel de Araújo
- Instituto de Química e Biotecnologia (IQB/UFAL), Programa de Pós-graduação Em Química e Biotecnologia, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Nassib Bezerra Bueno
- Faculdade de Nutrição, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Marília Oliveira Fonseca Goulart
- Instituto de Química e Biotecnologia (IQB/UFAL), Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
- Programa de Pós-graduação em Ciências da Saúde, ICBS, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
- Instituto de Química e Biotecnologia (IQB/UFAL), Programa de Pós-graduação Em Química e Biotecnologia, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| | - Alane Cabral Menezes de Oliveira
- Faculdade de Nutrição, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km. 96.7, Tabuleiro dos Martins, CEP 57.072-970 Maceió, Alagoas, Brazil
| |
Collapse
|
27
|
Caplan M, Keenan-Devlin LS, Freedman A, Grobman W, Wadhwa PD, Buss C, Miller GE, Borders AEB. Lifetime Psychosocial Stress Exposure Associated with Hypertensive Disorders of Pregnancy. Am J Perinatol 2021; 38:1412-1419. [PMID: 32615616 DOI: 10.1055/s-0040-1713368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Hypertensive disorders of pregnancy (HDP) complicate 5 to 10% of all pregnancies and are a major cause of pregnancy-related morbidity. Exposure to psychosocial stress has been associated with systemic inflammation and adverse birth outcomes in pregnant women. Thus, it is probable that psychosocial stress and inflammation play a role in the development of HDP. The primary objective of this analysis was to determine if a woman's lifetime psychosocial stress exposure was associated with an increased risk of HDP. Additionally, we examined whether serum inflammation was an underlying biological mediator for this relationship. STUDY DESIGN A multisite prospective study was conducted in a sociodemographically diverse cohort of 647 pregnant women. At a study visit between 12 and 206/7 weeks' gestation, maternal psychosocial stress was assessed with six validated assessments and inflammation was measured via log-transformed serum concentrations of interferon-γ, interleukin (IL)-10, IL-13, IL-6, IL-8, and tumor necrosis factor-α. A composite stress score was calculated for each participant from the six stress assessments. The diagnosis of HDP was abstracted from the medical record and was defined as the presence of gestational hypertension after 20 weeks of pregnancy and/or preeclampsia. The association between composite stress and HDP was determined using binary logistic regression. Inflammation, using the six inflammatory biomarkers, was tested as a potential mediator between stress and HDP. RESULTS Participants with higher composite stress scores were more likely to develop HDP (odds ratio [OR]: 1.50, 95% confidence interval [CI]: 1.06-2.12). When adjusted for known risk modifiers, including maternal age, race/ethnicity, parity, pre-pregnancy body mass index, diabetes, chronic hypertension, and smoking during pregnancy, the risk remained unchanged (OR: 1.50, 95% CI: 1.03-2.20). No mediation effect by inflammation was observed. CONCLUSION Independent of known risk factors, women exposed to greater composite stress burden across the life course are at increased risk of developing HDP. KEY POINTS · This study was conducted to determine if women with high levels of psychosocial stress have differences in risk for hypertensive disorders of pregnancy (HDP).. · Independent of known risk factors, women with increased lifetime psychosocial burden are at higher risk for HDP.. · A model that captures multiple domains of life stress may better predict HDP than a unimodal stress assessment..
Collapse
Affiliation(s)
- Madeleine Caplan
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, School of Medicine, Duke University, Durham, North Carolina
| | - Lauren S Keenan-Devlin
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, University of Chicago Pritzker School of Medicine, Evanston, Illinois
| | - Alexa Freedman
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Institute for Policy Research, Northwestern University, University of Chicago Pritzker School of Medicine, Evanston, Illinois
| | - William Grobman
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Center for Healthcare Studies, Institute for Public Health and Medicine, Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Pathik D Wadhwa
- Department of Psychiatry and Human Behavior, University of California, School of Medicine, UC Irvine Development, Health and Disease Research Program, Irvine, California
| | - Claudia Buss
- UC Irvine Development, Health and Disease Research Program, University of California Irvine, Irvine, California.,Department of Medical Psychology, Charité, University Medicine Berlin, Berlin, Germany
| | - Gregory E Miller
- Department of Psychology, Institute for Policy Research, Northwestern University, Evanston, Illinois
| | - Ann E B Borders
- Department of Obstetrics and Gynecology, NorthShore University Health System, Center for Healthcare Studies, Institute for Public Health and Medicine, University of Chicago Pritzker School of Medicine, Northwestern University, Evanston, Illinois
| |
Collapse
|
28
|
Lean SC, Jones RL, Roberts SA, Heazell AEP. A prospective cohort study providing insights for markers of adverse pregnancy outcome in older mothers. BMC Pregnancy Childbirth 2021; 21:706. [PMID: 34670515 PMCID: PMC8527686 DOI: 10.1186/s12884-021-04178-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
Background Advanced maternal age (≥35 years) is associated with increased rates of adverse pregnancy outcome. Better understanding of underlying pathophysiological processes may improve identification of older mothers who are at greatest risk. This study aimed to investigate changes in oxidative stress and inflammation in older women and identify clinical and biochemical predictors of adverse pregnancy outcome in older women. Methods The Manchester Advanced Maternal Age Study (MAMAS) was a multicentre, observational, prospective cohort study of 528 mothers. Participants were divided into three age groups for comparison 20–30 years (n = 154), 35–39 years (n = 222) and ≥ 40 years (n = 152). Demographic and medical data were collected along with maternal blood samples at 28 and 36 weeks’ gestation. Multivariable analysis was conducted to identify variables associated with adverse outcome, defined as one or more of: small for gestational age (< 10th centile), FGR (<5th centile), stillbirth, NICU admission, preterm birth < 37 weeks’ gestation or Apgar score < 7 at 5 min. Biomarkers of inflammation, oxidative stress and placental dysfunction were quantified in maternal serum. Univariate and multivariable logistic regression was used to identify associations with adverse fetal outcome. Results Maternal smoking was associated with adverse outcome irrespective of maternal age (Adjusted Odds Ratio (AOR) 4.22, 95% Confidence Interval (95%CI) 1.83, 9.75), whereas multiparity reduced the odds (AOR 0.54, 95% CI 0.33, 0.89). In uncomplicated pregnancies in older women, lower circulating anti-inflammatory IL-10, IL-RA and increased antioxidant capacity (TAC) were seen. In older mothers with adverse outcome, TAC and oxidative stress markers were increased and levels of maternal circulating placental hormones (hPL, PlGF and sFlt-1) were reduced (p < 0.05). However, these biomarkers only had modest predictive accuracy, with the largest area under the receiver operator characteristic (AUROC) of 0.74 for placental growth factor followed by TAC (AUROC = 0.69). Conclusions This study identified alterations in circulating inflammatory and oxidative stress markers in older women with adverse outcome providing preliminary evidence of mechanistic links. Further, larger studies are required to determine if these markers can be developed into a predictive model of an individual older woman’s risk of adverse pregnancy outcome, enabling a reduction in stillbirth rates whilst minimising unnecessary intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-021-04178-6.
Collapse
Affiliation(s)
- Samantha C Lean
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St. Mary's Hospital, 5th Floor (Research), Oxford Road, Manchester, M13 9WL, UK
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St. Mary's Hospital, 5th Floor (Research), Oxford Road, Manchester, M13 9WL, UK
| | - Stephen A Roberts
- Centre for Biostatistics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alexander E P Heazell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St. Mary's Hospital, 5th Floor (Research), Oxford Road, Manchester, M13 9WL, UK.
| |
Collapse
|
29
|
Kaneko K, Ozawa N, Murashima A. Obstetric anti-phospholipid syndrome: from pathogenesis to treatment. Immunol Med 2021; 45:79-93. [PMID: 34470570 DOI: 10.1080/25785826.2021.1969116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by clinical manifestations such as thrombosis and obstetric complications with documented persistence of antiphospholipid antibodies (aPLs). Recent studies have revealed that the cause of aPL-related obstetric complications is dysfunction of placental trophoblasts and inflammation of the maternal-fetal interface induced by aPLs, not thrombosis. Although aPLs are associated with recurrence of serious complications during pregnancy, appropriate combination therapy with heparin and low-dose aspirin can improve the course of 70-80% of subsequent pregnancies. Preconception counseling and patient-tailored treatment are fundamental to improving maternal and fetal outcomes. Non-anticoagulant treatments such as hydroxychloroquine and statins are being developed for cases refractory to conventional treatment. Risk factors for thrombosis after pregnancy complications were identified based on the analysis of large databases of obstetric APS.
Collapse
Affiliation(s)
- Kayoko Kaneko
- Division of Maternal Medicine, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Nobuaki Ozawa
- Division of Reproductive Medicine and Maternal Care/Immunology, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Atsuko Murashima
- Division of Maternal Medicine, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
30
|
Nasri F, Zare M, Hesampour F, Ahmadi M, Ali-Hassanzadeh M, Mostafaei S, Gharesi-Fard B. Are genetic variations in IL-1β and IL-6 cytokines associated with the risk of pre-eclampsia? Evidence from a systematic review and meta-analysis. J Matern Fetal Neonatal Med 2021; 35:6600-6609. [PMID: 34275427 DOI: 10.1080/14767058.2021.1918092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This systematic review and meta-analysis study was performed to assess the potential association between interleukin-1 beta (IL-1β) single nucleotide polymorphisms (SNPs) (rs1143634 and rs16944) and interleukin-6 (IL-6) SNP (rs1800795) and pre-eclampsia (PE). METHODS A comprehensive literature search was conducted in the international search engines and databases, including MEDLINE (via PubMed), Scopus, and Web of Science (ISI) up to 9 March 2021. After retrieving relevant articles, data extraction was performed by four authors independently. Pooled ORs and corresponding 95% CIs were used to evaluate the association between IL-1β and IL-6 polymorphisms and PE risk. Cochran's Q test was used to check heterogeneity, and the I2 index was calculated for measuring the heterogeneity between the estimations of included studies. RESULTS After reviewing fully published studies, 21 studies were included in this study based on the eligibility criteria. Our results showed that rs16944 and rs1143634 of IL-1β were significantly associated with the risk of PE. Regarding rs16944, the minor C allele significantly decreased the risk of PE (C vs. T: OR = 0.79, 95% CI = 0.69-0.90). In contrast, the minor T allele of rs1143634 significantly increased the risk of PE (T vs. C: OR = 1. 28, 95% CI = 1.04-1.58). There was no significant association between IL-6 rs1800795 (C vs. G: OR = 1.04, 95% CI = 0.93-1.16) polymorphism and PE risk. CONCLUSIONS In conclusion, this meta-analysis suggests rs1143634 and rs16944 polymorphisms of IL-1β are related to the risk of PE.
Collapse
Affiliation(s)
- Fatemeh Nasri
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Zare
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Hesampour
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Moslem Ahmadi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Shayan Mostafaei
- Department of Biostatistics, Faculty of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Epidemiology and Biostatistics Unit, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
31
|
Waker CA, Kaufman MR, Brown TL. Current State of Preeclampsia Mouse Models: Approaches, Relevance, and Standardization. Front Physiol 2021; 12:681632. [PMID: 34276401 PMCID: PMC8284253 DOI: 10.3389/fphys.2021.681632] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE) is a multisystemic, pregnancy-specific disorder and a leading cause of maternal and fetal death. PE is also associated with an increased risk for chronic morbidities later in life for mother and offspring. Abnormal placentation or placental function has been well-established as central to the genesis of PE; yet much remains to be determined about the factors involved in the development of this condition. Despite decades of investigation and many clinical trials, the only definitive treatment is parturition. To better understand the condition and identify potential targets preclinically, many approaches to simulate PE in mice have been developed and include mixed mouse strain crosses, genetic overexpression and knockout, exogenous agent administration, surgical manipulation, systemic adenoviral infection, and trophoblast-specific gene transfer. These models have been useful to investigate how biological perturbations identified in human PE are involved in the generation of PE-like symptoms and have improved the understanding of the molecular mechanisms underpinning the human condition. However, these approaches were characterized by a wide variety of physiological endpoints, which can make it difficult to compare effects across models and many of these approaches have aspects that lack physiological relevance to this human disorder and may interfere with therapeutic development. This report provides a comprehensive review of mouse models that exhibit PE-like symptoms and a proposed standardization of physiological characteristics for analysis in murine models of PE.
Collapse
Affiliation(s)
- Christopher A Waker
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Melissa R Kaufman
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| |
Collapse
|
32
|
Morosin SK, Lochrin AJ, Delforce SJ, Lumbers ER, Pringle KG. The (pro)renin receptor ((P)RR) and soluble (pro)renin receptor (s(P)RR) in pregnancy. Placenta 2021; 116:43-50. [PMID: 34020806 DOI: 10.1016/j.placenta.2021.04.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/31/2021] [Accepted: 04/27/2021] [Indexed: 11/25/2022]
Abstract
The (pro)renin receptor ((P)RR) is a multi-functional protein that can be proteolytically cleaved and released in a soluble form (s(P)RR). Recently, the (P)RR and s(P)RR have become of interest in pregnancy and its associated pathologies. This is because the (P)RR not only activates tissue renin angiotensin systems, but it is also an integral component of vacuolar-ATPase, activates the wingless/integrated (Wnt)/β-catenin and extracellular signal regulated kinases 1 and 2/mitogen-activated protein kinase signalling pathways, and stabilises the β subunit of pyruvate dehydrogenase. Additionally, s(P)RR is detected in plasma and urine, and maternal plasma levels are elevated in pregnancy complications including fetal growth restriction, preeclampsia and gestational diabetes mellitus. Therefore, s(P)RR has potential as a biomarker for these pregnancy pathologies. Preliminary functional findings suggest that s(P)RR may be important for regulating fluid balance, inflammation and blood pressure, all of which contribute to a successful pregnancy. The (P)RR and s(P)RR regulate pathways that are known to be important in maintaining pregnancy, however their role in the physiological context of pregnancy is poorly characterised. This review summarises the known and potential functions of the (P)RR and s(P)RR in pregnancy, and how their dysregulation may contribute to pregnancy complications. It also highlights the need for further research into the source and function of s(P)RR in pregnancy. Soluble (P)RR levels could be indicative of placental, kidney or liver dysfunction and therefore be a novel clinical biomarker, or therapeutic target, to improve the detection and treatment of pregnancy pathologies.
Collapse
Affiliation(s)
- Saije K Morosin
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Alyssa J Lochrin
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Sarah J Delforce
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia.
| |
Collapse
|
33
|
Gurm H, Hirota JA, Raha S. Cannabinoid Signalling in Immune-Reproductive Crosstalk during Human Pregnancy. Biomedicines 2021; 9:267. [PMID: 33800053 PMCID: PMC8000565 DOI: 10.3390/biomedicines9030267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the intricate involvement of the endocannabinoid system in various physiological processes, it remains one of the most under-studied biological systems of the human body. The scope of endocannabinoid signalling is widespread, ranging from modulation of immune responses in innate and adaptive immunity to gestational processes in female physiology. Cannabinoid receptors are ubiquitously distributed in reproductive tissues and are thought to play a role in regulating the immune-reproductive interactions required for successful pregnancy, specifically among uterine natural killer cells and placental extravillous trophoblasts. The use of cannabis during pregnancy, however, can perturb endocannabinoid homeostasis through effects mediated by its major constituents, Δ-9-tetrahydrocannabinol and cannabidiol. Decidualization of the endometrium, invasion, and angiogenesis may be impaired as a consequence, leading to clinical complications such as miscarriage and preeclampsia. In this review, the crosstalk between endocannabinoid signalling in uterine natural killer cells and placental extravillous trophoblasts will be examined in healthy and complicated pregnancies. This lays a foundation for discussing the potential of targeting the endocannabinoid system for therapeutic benefit, particularly with regard to the emerging field of synthetic cannabinoids.
Collapse
Affiliation(s)
- Harmeet Gurm
- Department of Pediatrics and the Graduate Program in Medical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Jeremy A. Hirota
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Program in Medical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| |
Collapse
|
34
|
Associations between Dietary Patterns and Inflammatory Markers during Pregnancy: A Systematic Review. Nutrients 2021; 13:nu13030834. [PMID: 33806342 PMCID: PMC8000934 DOI: 10.3390/nu13030834] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Elevated inflammation in pregnancy has been associated with multiple adverse pregnancy outcomes and potentially an increased susceptibility to future chronic disease. How maternal dietary patterns influence systemic inflammation during pregnancy requires further investigation. The purpose of this review was to comprehensively evaluate studies that assessed dietary patterns and inflammatory markers during pregnancy. This review was guided by the Preferred Reporting Items for Systematic Review and Meta-Analyses. Included studies were sourced from EMBASE, PubMed, Web of Science, and Scopus and evaluated using The Quality Assessment Tool for Quantitative Studies. Inclusion criteria consisted of human studies published in English between January 2007 and May 2020 that addressed associations between dietary patterns and inflammatory markers during pregnancy. Studies focused on a single nutrient, supplementation, or combined interventions were excluded. A total of 17 studies were included. Despite some inconsistent findings, maternal diets characterized by a higher intake of animal protein and cholesterol and/or a lower intake of fiber were shown to be associated with certain pro-inflammatory markers (C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor-α (TNF- α), IL-8, serum amyloid A (SAA), and glycoprotein acetylation (GlycA)). Future studies that explore a broader range of inflammatory markers in the pregnant population, reduce measurement errors, and ensure adequate statistical adjustment are warranted.
Collapse
|
35
|
Tuerxun D, Aierken R, Zhang YM, Huang Y, Sui S, Li XY, Abulikemu Z, Dilixiati N. Astragaloside IV alleviates lipopolysaccharide-induced preeclampsia-like phenotypes via suppressing the inflammatory responses. Kaohsiung J Med Sci 2021; 37:236-244. [PMID: 33089927 DOI: 10.1002/kjm2.12313] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/09/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Preeclampsia (PE) is a major cause of perinatal and maternal mortality and morbidity, which affects 2% to 8% of pregnancies in the world. The aberrant maternal inflammation and angiogenic imbalance have been demonstrated to contribute to the pathogenesis of PE. This research aimed to investigate the effect of Astragaloside IV (AsIV) in the treatment of PE and the underlying mechanisms. A rat PE-like model was established by tail vein injection of lipopolysaccharide (LPS) and different doses of AsIV (40 and 80 mg/kg) were treated at the same time. Systolic blood pressure, total urine protein and urine volume were observed. Serum and placenta inflammatory cytokines were measured by ELISA kit. The mRNA and protein expression of relative genes were analyzed by qRT-PCR and Western blotting. In PE-like rats, there were obvious increases in systolic blood pressure, total urine protein and urine volume, which were obviously alleviated by treatment with AsIV. Serum levels of interleukin (IL)-1β, tumor necrosis factor alpha (TNF-α), IL-6 and IL-18, as well as IL-4, IL-10, PIGF, VEGF and sFlt-1, were all reversed by treatment with AsIV. Meanwhile, AsIV treatment improved abnormal pregnancy outcomes, such as low litter size and low fetal weight. In addition, AsIV treatment downregulated the mRNA expression of inflammatory gene IL-1β and IL-6 in PE rats model, and AsIV treatment inhibited the activation of TLR-4, NF-κB, and sFlt-1 in the placenta of PE rats. Our findings indicated the first evidence that AsIV alleviated PE-like signs, and this improvement effect is possibly through inhibition of inflammation response via the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Dilihuma Tuerxun
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Remila Aierken
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Yan-Mei Zhang
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Ying Huang
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Shuang Sui
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Xiao-Ying Li
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Zulifeiya Abulikemu
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Nuerbiye Dilixiati
- Department of Obstestrics of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| |
Collapse
|
36
|
Spence T, Allsopp PJ, Yeates AJ, Mulhern MS, Strain JJ, McSorley EM. Maternal Serum Cytokine Concentrations in Healthy Pregnancy and Preeclampsia. J Pregnancy 2021; 2021:6649608. [PMID: 33680514 PMCID: PMC7925069 DOI: 10.1155/2021/6649608] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
The maternal immune response is essential for successful pregnancy, promoting immune tolerance to the fetus while maintaining innate and adaptive immunity. Uncontrolled, increased proinflammatory responses are a contributing factor to the pathogenesis of preeclampsia. The Th1/Th2 cytokine shift theory, characterised by bias production of Th2 anti-inflammatory cytokine midgestation, was frequently used to reflect the maternal immune response in pregnancy. This theory is simplistic as it is based on limited information and does not consider the role of other T cell subsets, Th17 and Tregs. A range of maternal peripheral cytokines have been measured in pregnancy cohorts, albeit the changes in individual cytokine concentrations across gestation is not well summarised. Using available data, this review was aimed at summarising changes in individual maternal serum cytokine concentrations throughout healthy pregnancy and evaluating their association with preeclampsia. We report that TNF-α increases as pregnancy progresses, IL-8 decreases in the second trimester, and IL-4 concentrations remain consistent throughout gestation. Lower second trimester IL-10 concentrations may be an early predictor for developing preeclampsia. Proinflammatory cytokines (TNF-α, IFN-γ, IL-2, IL-8, and IL-6) are significantly elevated in preeclampsia. More research is required to determine the usefulness of using cytokines, particularly IL-10, as early biomarkers of pregnancy health.
Collapse
Affiliation(s)
- Toni Spence
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Philip J. Allsopp
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Alison J. Yeates
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Maria S. Mulhern
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - J. J. Strain
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Emeir M. McSorley
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
37
|
Akbar MIA, Yosediputra A, Pratama RE, Fadhilah NL, Sulistyowati S, Amani FZ, Dachlan EG, Dikman Angsar M, Dekker GA. Pravastatin suppresses inflammatory cytokines and endothelial activation in patients at risk of developing preeclampsia: INOVASIA study. J Matern Fetal Neonatal Med 2021; 35:5375-5382. [PMID: 33522342 DOI: 10.1080/14767058.2021.1879785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The Indonesian INOVASIA study is an ongoing multicentre randomized, open controlled trial of pravastatin for the prevention of preeclampsia in patients deemed to be high risk. Here we evaluate the effects of pravastatin on circulating inflammatory and endothelial markers, i.e. Vascular Endothelial Growth Factor (VEGF), Interleukin-6 (IL-6), Endothelin-1 (ET-1), and Nitric Oxide (NO). METHODS Pregnant women deemed to be at a high risk of developing preeclampsia women were recruited based on the Fetal Medicine Foundation preeclampsia screening test or a history of preterm preeclampsia, or clinical risk factors in combination with an abnormal uterine artery Doppler flow pattern at 11-20 week's gestation. This is a nested cohort study within the larger trial (INOVASIA); 38 patients were consecutively recruited and assigned to the pravastatin group and the control group. Participants in the pravastatin group received pravastatin (2 × 20 mg p.o) in addition to a standard regimen of aspirin (80 mg p.o) and calcium (1 g p.o), from 14 to 20 weeks until delivery. Blood samples to measure the various biomarkers were obtained in consecutive patients before starting the research medication and just before delivery (pre and post-test examination). RESULT The number of samples on the 2 time points for the various biomarkers was: VEGF: 38, IL-6: 30, ET-1: 38, and NO: 35. IL-6 levels decreased significantly in the pravastatin group (mean ± SD): (191.87 ± 82.99 vs. 151.85 + 48.46, p = .013), while levels in the control group did not change significantly (median (interquartile range)) (144.17 (53.91) vs. 140.82 (16.18), p = .177). ET-1 levels decreased significantly in the pravastatin group (3.64 ± 0.85 vs. 3.01 ± 0.74, p = .006) while the control group had more or less stable levels (3.57 ± 1.12 vs. 3.78 ± 0.73 p = .594). NO was the only serum marker that showed significant changes in both groups. NO levels increased in pravastatin group (11.30 (17.43) vs. 41.90 (53.18), p = .044) and decreased in control group (38.70 (34.80) vs. 10.03 (26.96), p = .002). VEGF levels appeared to follow opposite trends in the 2 groups (NS) (Pravastatin: 3.22 (0.62) vs. 3.28 (0.75), p = .402. Control: 3.38 (0.83) vs. 3.06 (0.74), p = .287). CONCLUSION Administration of 40 mg pravastatin resulted in an improvement in NO levels, and a decrease in IL-6 and endothelin (ET-1) levels. The direction of the effect of pravastatin on these biomarkers appears to underpin the potential for a beneficial effect of pravastatin in the prevention of preeclampsia.
Collapse
Affiliation(s)
- Muhammad Ilham Aldika Akbar
- Department Obstetrics & Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Universitas Airlangga Hospital, Surabaya, Indonesia
| | - Angelina Yosediputra
- Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Raditya Ery Pratama
- Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Nur Lailatul Fadhilah
- Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | | | - Fariska Zata Amani
- Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Erry Gumilar Dachlan
- Department Obstetrics & Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Universitas Airlangga Hospital, Surabaya, Indonesia
| | - Muhammad Dikman Angsar
- Department Obstetrics & Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Universitas Airlangga Hospital, Surabaya, Indonesia
| | - Gustaaf Albert Dekker
- Department Obstetrics & Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.,Department Obstetrics & Gynecology, Lyell McEwin Hospital, The University of Adelaide, Elizabeth Vale, Australia
| |
Collapse
|
38
|
Barron A, McCarthy CM, O'Keeffe GW. Preeclampsia and Neurodevelopmental Outcomes: Potential Pathogenic Roles for Inflammation and Oxidative Stress? Mol Neurobiol 2021; 58:2734-2756. [PMID: 33492643 DOI: 10.1007/s12035-021-02290-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
Preeclampsia (PE) is a common and serious hypertensive disorder of pregnancy that occurs in approximately 3-5% of first-time pregnancies and is a well-known leading cause of maternal and neonatal mortality and morbidity. In recent years, there has been accumulating evidence that in utero exposure to PE acts as an environmental risk factor for various neurodevelopmental disorders, particularly autism spectrum disorder and ADHD. At present, the mechanism(s) mediating this relationship are uncertain. In this review, we outline the most recent evidence implicating a causal role for PE exposure in the aetiology of various neurodevelopmental disorders and provide a novel interpretation of neuroanatomical alterations in PE-exposed offspring and how these relate to their sub-optimal neurodevelopmental trajectory. We then postulate that inflammation and oxidative stress, two prominent features of the pathophysiology of PE, are likely to play a major role in mediating this association. The increased inflammation in the maternal circulation, placenta and fetal circulation in PE expose the offspring to both prenatal maternal immune activation-a risk factor for neurodevelopmental disorders, which has been well-characterised in animal models-and directly higher concentrations of pro-inflammatory cytokines, which adversely affect neuronal development. Similarly, the exaggerated oxidative stress in the mother, placenta and foetus induces the placenta to secrete factors deleterious to neurons, and exposes the fetal brain to directly elevated oxidative stress and thus adversely affects neurodevelopmental processes. Finally, we describe the interplay between inflammation and oxidative stress in PE, and how both systems interact to potentially alter neurodevelopmental trajectory in exposed offspring.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland.,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Cathal M McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland. .,Cork Neuroscience Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
39
|
Mohseni Z, Derksen E, Oben J, Al-Nasiry S, Spaanderman MEA, Ghossein-Doha C. Cardiac dysfunction after preeclampsia; an overview of pro- and anti-fibrotic circulating effector molecules. Pregnancy Hypertens 2020; 23:140-154. [PMID: 33388730 DOI: 10.1016/j.preghy.2020.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/29/2020] [Accepted: 12/08/2020] [Indexed: 01/09/2023]
Abstract
Preeclampsia (PE) is strongly associated with heart failure (HF) later in life. The aberrant cardiac remodelling is likely initiated or amplified during preeclamptic pregnancy. Aberrant remodelling often persists after delivery and is known to relate strongly to cardiac fibrosis. This review provides an overview of pro- and anti- fibrotic circulating effector molecules that are involved in cardiac fibrosis and their association with PE. Women with PE complicated pregnancies show increased ANG-II sensitivity and elevated levels of the pro-fibrotic factors IL-6, TNF-α, TGs and FFAs compared to uncomplicated pregnancies. In the postpartum period, PE pregnancies compared to uncomplicated pregnancies have increased ANG-II sensitivity, elevated levels of the pro-fibrotic factors IL-6, TNF-α, LDL cholesterol and leptin, as well as decreased levels of the anti-fibrotic factor adiponectin. The review revealed several profibrotic molecules that associate to cardiac fibrosis during and after PE. The role that these fibrotic factors have on the heart during and after PE may improve the understanding of the link between PE and HF. Furthermore they may provide insight into the pathways in which the relation between both diseases can be understood as potential mechanisms which interfere in the process of cardiovascular disease (CVD). Unravelling the molecular mechanism and pathways involved might bring the diagnostic and therapeutic abilities of those factors a step closer.
Collapse
Affiliation(s)
- Zenab Mohseni
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), The Netherlands.
| | - Elianne Derksen
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), The Netherlands
| | - Jolien Oben
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), The Netherlands
| | - Salwan Al-Nasiry
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), The Netherlands
| | - Marc E A Spaanderman
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), The Netherlands; Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Center, The Netherlands
| | - Chahinda Ghossein-Doha
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre (MUMC+), The Netherlands; Department of Cardiology, Maastricht University Medical Centre (MUMC+), The Netherlands
| |
Collapse
|
40
|
Preeclampsia Among African American Pregnant Women: An Update on Prevalence, Complications, Etiology, and Biomarkers. Obstet Gynecol Surv 2020; 75:111-120. [PMID: 32105335 DOI: 10.1097/ogx.0000000000000747] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Importance Preeclampsia is a devastating disease of pregnancy associated with increased risk of fetal and maternal complications. African American pregnant women have a high prevalence of preeclampsia, but there is a need of systemic analyses of this high-risk group regarding complications, etiology, and biomarkers. Objective The aim of this study was to provide a synopsis of current research of preeclampsia specifically related to African American women. Evidence Acquisition A comprehensive search was performed in the bibliographic database PubMed with keywords "preeclampsia" and "African American." Results African American women with preeclampsia were at an increased risk of preterm birth, which resulted in low-birth-weight infants. Intrauterine fetal death among African American preeclamptic patients occurs at twice the rate as in other races. On the maternal side, African American mothers with preeclampsia have more severe hypertension, antepartum hemorrhage, and increased mortality. Those who survive preeclampsia have a high risk of postpartum cardiometabolic disease. Preexisting conditions (eg, systemic lupus erythematosus) and genetic mutations (eg, sickle cell disease in the mother, FVL or APOL1 mutations in the fetus) may contribute to the higher prevalence and worse outcomes in African American women. Many blood factors, for example, the ratio of proteins sFlt/PlGF, hormones, and inflammatory factors, have been studied as potential biomarkers for preeclampsia, but their specificity needs further investigation. Conclusions Further studies of preeclampsia among African American women addressing underlying risk factors and etiologies, coupled with identification of preeclampsia-specific biomarkers allowing early detection and intervention, will significantly improve the clinical management of this devastating disease.
Collapse
|
41
|
Kajimoto E, Endo M, Fujimoto M, Matsuzaki S, Fujii M, Yagi K, Kakigano A, Mimura K, Tomimatsu T, Serada S, Takeuchi M, Yoshino K, Ueda Y, Kimura T, Naka T. Evaluation of leucine-rich alpha-2 glycoprotein as a biomarker of fetal infection. PLoS One 2020; 15:e0242076. [PMID: 33211747 PMCID: PMC7676652 DOI: 10.1371/journal.pone.0242076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/27/2020] [Indexed: 11/21/2022] Open
Abstract
This study aimed to determine the association between umbilical cord leucine-rich alpha-2 glycoprotein (LRG) and fetal infection and investigate the underlying mechanism of LRG elevation in fetuses. We retrospectively reviewed the medical records of patients who delivered at Osaka University Hospital between 2012 and 2017 and selected those with histologically confirmed chorioamnionitis (CAM), which is a common pregnancy complication that may cause neonatal infection. The participants were divided into two groups: CAM with fetal infection (CAM-f[+] group, n = 14) and CAM without fetal infection (CAM-f[−] group, n = 31). Fetal infection was defined by the histological evidence of funisitis. We also selected 50 cases without clinical signs of CAM to serve as the control. LRG concentrations in sera obtained from the umbilical cord were unaffected by gestational age at delivery, neonatal birth weight, nor the presence of noninfectious obstetric complications (all, p > 0.05). Meanwhile, the LRG levels (median, Interquartile range [IQR]) were significantly higher in the CAM-f(+) group (10.37 [5.21–13.7] μg/ml) than in the CAM-f(−) (3.61 [2.71–4.65] μg/ml) or control group (3.39 [2.81–3.93] μg/ml; p < 0.01). The area under the receiver operating characteristic (ROC) curve of LRG for recognizing fetal infection was 0.92 (optimal cutoff, 5.08 μg/ml; sensitivity, 86%; specificity, 88%). In a mouse CAM model established by lipopolysaccharide administration, the fetal LRG protein in sera and LRG mRNA in the liver were significantly higher than those in phosphate-buffered saline (PBS)-administered control mice (p < 0.01). In vitro experiments using a fetal liver-derived cell line (WRL68) showed that the expression of LRG mRNA was significantly increased after interleukin (IL)-6, IL-1β, and tumor necrosis factor- alpha (TNF-α) stimulation (p < 0.01); the induction was considerably stronger following IL-6 and TNF-α stimulation (p < 0.01). In conclusion, LRG is an effective biomarker of fetal infection, and fetal hepatocytes stimulated with inflammatory cytokines may be the primary source of LRG production in utero.
Collapse
Affiliation(s)
- Etsuko Kajimoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Obstetrics and Gynecology, Japan Community Health Care Organization Osaka Hospital, Osaka, Japan
| | - Masayuki Endo
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Children and Women’s Health, Osaka University Graduate School of Medicine, Osaka, Japan
- Division of Health Science, Graduate School of medicine, StemRIM Institute of Regeneration-Inducting Medicine, Osaka University, Osaka, Japan
| | - Minoru Fujimoto
- Center for Intractable Immune Disease, Kochi Medical School, Kochi University, Kochi, Japan
- * E-mail:
| | - Shinya Matsuzaki
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Fujii
- Division of Health Science, Graduate School of medicine, StemRIM Institute of Regeneration-Inducting Medicine, Osaka University, Osaka, Japan
| | - Kazunobu Yagi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Aiko Kakigano
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Mimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takuji Tomimatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Serada
- Center for Intractable Immune Disease, Kochi Medical School, Kochi University, Kochi, Japan
| | - Makoto Takeuchi
- Department of Pathology, Osaka Women’s and Children’s Hospital, Osaka, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuji Naka
- Center for Intractable Immune Disease, Kochi Medical School, Kochi University, Kochi, Japan
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
42
|
Ghaneifar Z, Yousefi Z, Tajik F, Nikfar B, Ghalibafan F, Abdollahi E, Momtazi-Borojeni AA. The potential therapeutic effects of curcumin on pregnancy complications: Novel insights into reproductive medicine. IUBMB Life 2020; 72:2572-2583. [PMID: 33107698 DOI: 10.1002/iub.2399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 10/11/2020] [Indexed: 01/13/2023]
Abstract
Pregnancy complications including preeclampsia, preterm birth, intrauterine growth restriction, and gestational diabetes are the main adverse reproductive outcomes. Excessive inflammation and oxidative stress play crucial roles in the pathogenesis of pregnancy disorders. Curcumin, the main polyphenolic compound derived from Curcuma longa, is mainly known by its anti-inflammatory and antioxidant properties. There are in vitro and in vivo reports revealing the preventive and ameliorating effects of curcumin against pregnancy complications. Here, we aimed to seek mechanisms underlying the modulatory effects of curcumin on dysregulated inflammatory and oxidative responses in various pregnancy complications.
Collapse
Affiliation(s)
- Zahra Ghaneifar
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fatemeh Tajik
- Faculty of medicine, Azad University of Tehran, Tehran, Iran
| | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghalibafan
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Abdollahi
- Department of Medical Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
43
|
Ritter A, Roth S, Kreis NN, Friemel A, Hoock SC, Steglich Souto A, Eichbaum C, Neuhoff A, Chen Q, Solbach C, Louwen F, Yuan J. Primary Cilia in Trophoblastic Cells: Potential Involvement in Preeclampsia. Hypertension 2020; 76:1491-1505. [PMID: 33026915 DOI: 10.1161/hypertensionaha.120.15433] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathogenesis of preeclampsia, a pregnancy-related disease, is not completely understood. The primary cilium transduces a diverse array of signaling pathways important for vital cellular activities. Primary cilia were reported to facilitate trophoblastic cell invasion. We hypothesized their further functions in trophoblasts and were interested in related molecular mechanisms. We systematically examined the presence, length and percentage of the primary cilium, its mediated signal transduction, and its connection to trophoblast function. Various cellular and molecular methods were used including immunofluorescence staining, spheroid formation, gene analysis, invasion and tube formation assays with trophoblastic cell lines, primary trophoblasts, and placental tissues. We show that primary cilia are present in various trophoblastic cell lines derived from first trimester placentas. Cilia are also observable in primary trophoblasts, though in a small quantity. Importantly, primary cilia are shortened in trophoblastic cells derived from preeclamptic placentas. Mechanistically, interleukin-6, tumor necrosis factor-α or sera from patients with preeclampsia are able to reduce the length of primary cilia and impair the important sonic hedgehog signaling pathway. Functionally, trophoblastic cells with defective cilia display severe failures in their key functions, like migration, invasion and tube formation, also observed in trophoblastic cells depleted of the intraflagellar transport protein 88. This is accompanied by reduced gene expression of matrix metallopeptidases, vascular endothelial growth factor, and placental growth factor. This work highlights the significance of primary cilia in the functions of trophoblastic cells. Dysfunctional cilia may lead to compromised migration, invasion, and endothelial remodeling of trophoblastic cells, contributing to the development of preeclampsia.
Collapse
Affiliation(s)
- Andreas Ritter
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Susanne Roth
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Nina-Naomi Kreis
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Alexandra Friemel
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Samira Catharina Hoock
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Alice Steglich Souto
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Christine Eichbaum
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Annemarie Neuhoff
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Qi Chen
- Department of Obstetrics and Gynaecology, University of Auckland, New Zealand (Q.C.).,Hospital of Obstetrics and Gynaecology, Fudan University, Shanghai, China (Q.C.)
| | - Christine Solbach
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Frank Louwen
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| | - Juping Yuan
- From the Division of Obstetrics and Prenatal Medicine, Department of Gynaecology and Obstetrics, University Hospital, Goethe University, Germany (A.R., S.R., N.-N.K., A.F., S.C.H., A.S.S., C.E., A.N., C.S., F.L., J.Y.)
| |
Collapse
|
44
|
Mauro AK, Berdahl DM, Khurshid N, Clemente L, Ampey AC, Shah DM, Bird IM, Boeldt DS. Conjugated linoleic acid improves endothelial Ca2+ signaling by blocking growth factor and cytokine-mediated Cx43 phosphorylation. Mol Cell Endocrinol 2020; 510:110814. [PMID: 32259635 PMCID: PMC7253345 DOI: 10.1016/j.mce.2020.110814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
Sustained Ca2+ burst signaling is crucial for endothelial vasodilator production and is disrupted by growth factors and cytokines. Conjugated linoleic acid (CLA), a Src inhibitor in certain preparations, is generally regarded as safe during pregnancy by the FDA. Multiple CLA preparations; t10, c12 or c9, t11 CLA, or a 1:1 mixture of the two were administered before growth factor or cytokine treatment. Growth factors and cytokines caused a significant decrease in Ca2+ burst numbers in response to ATP stimulation. Both t10, c12 CLA and the 1:1 mixture rescued VEGF165 or TNFα inhibited Ca2+ bursts and correlated with Src-specific phosphorylation of connexin 43. VEGF165, TNFα, and IL-6 in combination at physiologic concentrations revealed IL-6 amplified the inhibitory effects of lower dose of VEGF165 and TNFα. Again, the 1:1 CLA mixture was most effective at rescue of function. Therefore, CLA formulations may be a promising treatment for endothelial dysfunction in diseases such as preeclampsia.
Collapse
Affiliation(s)
- Amanda K Mauro
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Danielle M Berdahl
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA; Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Nauman Khurshid
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA; Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Luca Clemente
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Amanda C Ampey
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Dinesh M Shah
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Ian M Bird
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA; Department of Pediatrics, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Derek S Boeldt
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA.
| |
Collapse
|
45
|
Li L, Chen W, Ma L, Liu ZB, Lu X, Gao XX, Liu Y, Wang H, Zhao M, Li XL, Cong L, Xu DX, Chen YH. Continuous association of total bile acid levels with the risk of small for gestational age infants. Sci Rep 2020; 10:9257. [PMID: 32518361 PMCID: PMC7283485 DOI: 10.1038/s41598-020-66138-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 05/11/2020] [Indexed: 12/27/2022] Open
Abstract
The association between maternal serum total bile acid (TBA) levels and small-for-gestational-age (SGA) infants is unclear. We investigated the association between various degrees of serum TBA levels and the risk of SGA infants in a Chinese population. The current study performed a cohort study among 11811 mothers with singleton pregnancy. Subjects were divided into seven categories according to maternal serum TBA levels. Interestingly, birth sizes were reduced, whereas the rate of SGA infants was increased across increasing categories of serum TBA. Compared to category 1, adjusted ORs (95%CI) for SGA infants were 0.99 (0.82-1.21) in category 2, 1.22 (0.97-1.53) in category 3, 1.99 (1.53-2.58) in category 4, 2.91 (2.16-3.93) in category 5, 4.29 (3.33-5.54) in category 6, and 9.01 (5.99-13.53) in category 7, respectively. Furthermore, adjusted ORs (95%CI) for SGA infants for each 1-SD increase in serum TBA levels were 1.36 (1.29-1.43) among all subjects, 2.40 (1.82-3.45) among subjects without cholestasis, and 1.13 (1.06-1.22) among subjects with cholestasis, respectively. These results suggest that gestational cholestasis increases the risk of SGA infants. Additionally, our results indicate strong, continuous associations of serum TBA levels below those diagnostic of cholestasis with a decreased birth sizes and an increased risk of SGA infants.
Collapse
Affiliation(s)
- Li Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, HeFei, 230022, China
| | - Wei Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Li Ma
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Zhi Bing Liu
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Xue Lu
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Xing Xing Gao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yan Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Mei Zhao
- School of Nursing, Anhui Medical University, Hefei, 230032, China
| | - Xiao Lan Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, HeFei, 230022, China
| | - Lin Cong
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, HeFei, 230022, China
| | - De Xiang Xu
- School of Public Health, Anhui Medical University, Hefei, 230032, China.
| | - Yuan Hua Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.
| |
Collapse
|
46
|
Comparative Analysis of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells between Preeclampsia and Normal Pregnant Women. Stem Cells Int 2020; 2020:8403192. [PMID: 32587622 PMCID: PMC7298345 DOI: 10.1155/2020/8403192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 12/26/2022] Open
Abstract
Preeclampsia is a syndrome characterized by deterioration of either the maternal condition or the fetal condition. The adverse intrauterine environment made by preeclampsia results into intrauterine growth restriction and increased risk of a variety of diseases in future life. Given the adverse environment of fetal circulation made in the preeclamptic condition, and the role of mesenchymal stem cell (MSC) as a multipotent progenitor cell, we hypothesized that MSCs derived from human umbilical cord blood (hUCB-MSCs) obtained from preeclampsia are adversely altered or affected compared with normal pregnancy. The aim of this study was to analyze the biological characteristics and compare the functional abilities and gene expression patterns of hUCB-MSCs originating from pregnant women with and without severe preeclampsia. hUCB-MSCs were isolated and cultured from 28 pregnant women with severe preeclampsia and 30 normal pregnant women. hUCB-MSCs obtained from women with preeclampsia were less proliferative and more senescent and had lower telomerase activity and higher ROS activity than cells from women with normal pregnancy. In addition, many senescence-related differentially expressed genes (DEGs) were identified by analysis of microarray gene expression profiles and significantly associated with the Gene Ontology term cell aging. In conclusion, hUCB-MSCs obtained from women with preeclampsia showed the poorly proliferative, more senescent, and decreased telomerase activity, and these characters may be related with functional impairment of MSC from preeclampsia compared with cells from normal pregnancy.
Collapse
|
47
|
Lumbreras-Marquez MI, Lumbreras-Marquez J, Barraza-Salas M, Castillo-Reyther RA, De la Maza-Labastida S, Hernandez-Rayon YI, Farber MK, Vazquez-Alaniz F. Maternal and umbilical cord procalcitonin, high-sensitivity C-reactive protein, and interleukin-6 levels in preeclamptic and normotensive patients: A cross-sectional study. Pregnancy Hypertens 2020; 21:218-223. [PMID: 32683282 DOI: 10.1016/j.preghy.2020.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/07/2020] [Accepted: 04/25/2020] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To report maternal and umbilical vein levels of procalcitonin (PCT) in patients with preeclampsia (PE) compared to controls. As secondary aims, we measured high-sensitivity C-reactive protein (hs-CRP), and interleukin-6 (IL-6). Moreover, correlation analyses were performed between the inflammatory biomarkers and mean arterial pressure (MAP). STUDY DESIGN This was a single center, cross-sectional study. MAIN OUTCOME MEASURES After Institutional Review Board approval and written informed consent, patients with or without PE were enrolled. PCT, hs-CRP, and IL-6 levels were compared between groups using multiple linear regression models. We calculated the adjusted ratios of geometric means (aRGM) for the comparison of patients with and without PE. Correlation analysis between the inflammatory biomarkers and MAP was performed using Spearman's method. RESULTS A total of 156 participants were enrolled, yielding 156 venous blood samples and umbilical venous blood samples. Seventy-six patients were in the PE group, and 80 patients were in the control group. Maternal plasma and serum concentrations of PCT (aRGM 3.35 (95% confidence interval [CI]: 2.26, 4.95; p < 0.001)), hs-CRP (aRGM 1.85 (95% CI: 1.30, 2.63; p = 0.003)), and IL-6 (aRGM 1.49 (95% CI: 1.08, 2.04; p = 0.045)) were higher in the PE group. In umbilical venous samples, the concentrations of PCT (aRGM 2.54 (95% CI: 1.46, 4.44; p = 0.003)) and hs-CRP (aRGM 1.45 (95% CI: 1.13, 1.87; p = 0.012)) in the PE group were higher than the controls. No difference in umbilical venous IL-6 concentrations were detected between PE vs. control groups (aRGM 1.46; 95% CI: 1.07, 1.98; p = 0.051). There was positive correlation for both PCT and hs-CRP with MAP in maternal and umbilical venous samples. However, there was no correlation between IL and 6 and MAP in maternal or umbilical venous samples. CONCLUSIONS PCT levels were elevated in maternal and umbilical venous samples of patients with PE, and correlated with disease severity.
Collapse
Affiliation(s)
- Mario I Lumbreras-Marquez
- Department of Obstetrics and Gynecology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Jesus Lumbreras-Marquez
- Department of Obstetrics and Gynecology, Hospital Central Dr. Ignacio Morones Prieto, San Luis Potosi, Mexico
| | - Marcelo Barraza-Salas
- Molecular Genetic Laboratory, Facultad de Ciencias Químicas, Universidad Juarez del Estado de Durango, Mexico
| | - Roberto A Castillo-Reyther
- Department of Obstetrics and Gynecology, Hospital Central Dr. Ignacio Morones Prieto, San Luis Potosi, Mexico
| | | | | | - Michaela K Farber
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Fernando Vazquez-Alaniz
- Molecular Genetic Laboratory, Facultad de Ciencias Químicas, Universidad Juarez del Estado de Durango, Mexico; Clinical Investigation Unit, Hospital General 450, Servicios de Salud de Durango, Mexico.
| |
Collapse
|
48
|
Dela Justina V, Priviero F, Dos Passos RR, Webb RC, Lima VV, Giachini FR. O-GlcNAc impairs endothelial function in uterine arteries from virgin but not pregnant rats: The role of GSK3β. Eur J Pharmacol 2020; 880:173133. [PMID: 32343970 DOI: 10.1016/j.ejphar.2020.173133] [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: 03/13/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023]
Abstract
Increased O-Linked β-N-acetylglucosamine (O-GlcNAc) is observed in several pathologies, and unbalanced O-GlcNAcylation levels favor endothelial dysfunction. Whether augmented O-GlcNAc impacts the uterine artery (UA) function and how it affects the UA during pregnancy remains to be elucidated. We hypothesized that glucosamine treatment increases O-GlcNAc, leading to uterine artery dysfunction and this effect is prevented by pregnancy. Pregnant (P) and non-pregnant (NP) Wistar rats were treated with glucosamine (300 mg/kg; i.p.) for 21 days. Concentration response-curves (CRC) to acetylcholine (in the presence or absence of L-NAME) and sodium nitroprusside were performed in UAs. In NP rats, glucosamine treatment increased O-GlcNAc expression in UAs accompanied by decreased endothelium-dependent relaxation, which was abolished by L-NAME. Endothelial nitric oxide synthase (eNOS) activity and total Akt expression were decreased by glucosamine-treatment in NP rats. Further, NP rats treated with glucosamine displayed increased glycogen synthase kinase 3 beta (GSK3β) activation and O-GlcNAc-transferase (OGT) expression in the UA. P rats treated with glucosamine displayed decreased O-GlcNAc in UAs and it was accompanied by improved relaxation to acetylcholine, whereas eNOS and GSK3β activity and total Akt and OGT expression were unchanged. Sodium nitroprusside-induced relaxation was not changed in all groups, indicating that glucosamine treatment led to endothelial dysfunction in NP rats. The underlying mechanism is, at least in part, dependent on Akt/GSK3β/OGT modulation. We speculate that during pregnancy, hormonal alterations play a protective role in preventing O-GlcNAcylation-induced endothelial dysfunction in the UAs.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | | | - Rinaldo Rodrigues Dos Passos
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil; Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, MT, Brazil
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Victor Vitorino Lima
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, MT, Brazil
| | - Fernanda R Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil; Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, MT, Brazil.
| |
Collapse
|
49
|
HSPA1L rs1061581 polymorphism is associated with the risk of preeclampsia in Han Chinese women. Biosci Rep 2020; 40:222071. [PMID: 32039449 PMCID: PMC7048671 DOI: 10.1042/bsr20194307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 11/17/2022] Open
Abstract
Preeclampsia (PE) is an excessive systemic inflammation response with dysfunction of endothelial. As a stress protein, heat shock protein 70 (HSP70) plays a pivotal role in protecting cells against apoptosis, oxidative damage and genetic damage. In humans, three genes encode members of the HSP70 class: HSPA1A, HSPA1B and HSPA1L. Our study was to investigate the association between genetic variations of HSPA1L and the susceptibility for PE in Chinese Han population. The polymorphisms of rs2227956, rs1043618 and rs1061581 in HSPA1L were genotyped by TaqMan allelic discrimination real time polymerase chain reaction (PCR) in 929 PE patients and 1024 healthy pregnant women. Statistic difference of the genotypic and allelic frequencies were found in HSPA1L rs1061581 between PE patients and controls (χ2 = 29.863, P < 0.001 by genotype; χ2 = 27.298, P < 0.001, OR = 1.874, 95%CI 1.476-2.379 by allele) and HSPA1L rs1061581 A alleles occurred more frequently in PE patients compared with healthy controls (PE vs. controls 10.28% vs. 5.76%). Furthermore, we divided the PE cases into early-onset/late-onset PE and mild/severe PE subgroups and found statistical differences in genotypic and allelic frequencies of the HSPA1L rs1061581 between early-onset PE, late-onset PE, mild PE, severe PE and controls, respectively. Moreover, HSPA1L rs1061581 A alleles were more frequent in early-onset PE, late-onset PE, mild PE and severe PE than controls respectively. Therefore, we concluded that HSPA1L rs1061581 polymorphism is associated with the risk of PE in Han Chinese women and A alleles may play a role in the susceptibility for PE.
Collapse
|
50
|
Cakir SC, Dorum BA, Koksal N, Ozkan H. The effects of maternal preeclampsia on inflammatory cytokines and clinical outcomes in premature infants. Pak J Med Sci 2020; 36:26-31. [PMID: 32063926 PMCID: PMC6994880 DOI: 10.12669/pjms.36.2.1316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective: To investigate the effects of maternal preeclampsia on inflammatory cytokines and neonatal outcomes in premature infants. Methods: The study included preterm infants born at gestational age ≤32 weeks in a tertiary university hospital between January 2016 and January 2017. The study group consisted of infants born from mothers with preeclampsia (Group-1), and the control group consisted of infants born from normotensive mothers (Group-2). Demographic characteristics and clinical outcomes of the infants were recorded. IL-6, IL-8, IL-10, and TNF-α cytokine levels were measured from umbilical cord blood samples. Results: A total of 108 infants were included in the study, of which 34 were in the Group-1 and 74 in the Group-2. Gestational ages (29 vs 30 weeks) of the infants in both groups were similar. There was no significant difference between the cytokine levels of infants with and without preeclampsia. The rate of small for gestational age, retinopathy of prematurity, intraventricular hemorrhage, necrotizing enterocolitis, neutropenia, and thrombocytopenia were significantly higher at the infants with preeclampsia. Conclusion: Maternal preeclampsia leads to an increase at the neonatal morbidities in premature infants without causing a significant alteration at the cytokine levels in cord blood.
Collapse
Affiliation(s)
- Salih Cagri Cakir
- Salih Cagri Cakir, Medical Doctor, Department of Pediatrics, Division of Neonatology, Uludag University Medical Faculty, Gorukle, 16059, Nilufer-Bursa-Turkey
| | - Bayram Ali Dorum
- Bayram Ali Dorum, Department of Pediatrics, Division of Neonatology, Uludag University Medical Faculty, Gorukle, 16059, Nilufer-Bursa-Turkey
| | - Nilgun Koksal
- Prof. Nilgun Koksal, Medical Doctor, Department of Pediatrics, Division of Neonatology, Uludag University Medical Faculty, Gorukle, 16059, Nilufer-Bursa-Turkey
| | - Hilal Ozkan
- Hilal Ozkan, Associate Professor, Department of Pediatrics, Division of Neonatology, Uludag University Medical Faculty, Gorukle, 16059, Nilufer-Bursa-Turkey
| |
Collapse
|