1
|
Song M, Yang H, Liu R. Kaempferol modulates Wnt/ β-catenin pathway to alleviate preeclampsia- induced changes and protect renal and ovarian histomorphology. J Mol Histol 2024; 56:36. [PMID: 39644402 DOI: 10.1007/s10735-024-10321-2] [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: 10/14/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Preeclampsia (PE) is a form of hypertension that manifests in the later stages of pregnancy. Since Kaempferol (Ka) has remedial potential hence this research was conducted to examine its therapeutic effect on Preeclampsia rats by regulating Wingless-related integration site/β-catenin (Wnt/B-catenin) pathway. To achieve this, thirty-two SD female rats were randomly allocated into four groups: control, preeclampsia (PE, LPS, 1 mg/kg), preeclampsia with kaempferol (PE + Ka), and preeclampsia with Dickkopf - 1 (DKK-1) and kaempferol (PE + DKK-1 + Ka). Rats in the PE + Ka and PE + DKK-1 + Ka groups received intraperitoneal injections at 50 mg/kg/d of kaempferol, whereas the PE + DKK-1 + Ka group was administered with 60 µg/kg/d of recombinant rat DKK-1 protein, an inhibitor of the Wnt/β-catenin signaling pathway. Our findings revealed that systolic blood pressure (SBP) in the PE + Ka group was significantly reduced in comparison to PE group (P < 0.05). The urine albumin levels in the PE + Ka group decreased noticeably (P < 0.05), whereas serum concentrations of Tumor Necrosis Factor Alpha (TNF-α), Interleukin-1β (IL-1β), and Interleukin-6 (IL-6) in the PE + Ka group were reduced (P < 0.05) in comparison to PE group. Although PE + Ka group exhibited elevated levels of superoxide dismutases (SOD), glutathione (GSH), and catalase (CAT) in placental tissue relative to the PE group, whilst levels of malondialdehyde (MDA), alkaline phosphatase (ALP), serum glutamic-pyruvic transaminase (SGPT), and serum glutamic-oxaloacetic transaminase (SGOT) considerably decreased (P < 0.05). Comparatively mRNA levels of Wnt1 and β-catenin in the PE + Ka group were elevated, whereas mRNA level of DKK-1 was diminished (P < 0.05). Administration of DKK-1 counteracted kaempferol effects on these parameters in Preeclampsia rats (P < 0.05). Devastatingly, ovarian and kidney histomorphology in the PE group exhibited significant degenerative alterations, whereas kaempferol groups demonstrated normal histomorphology in comparison to the PE group. Conclusively, Kaempferol can significantly lower systolic blood pressure and urine albumin in PE female rats while mitigating excessive oxidative stress. The therapeutic efficacy of kaempferol on Preeclampsia may be mediatated via Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Meiyu Song
- Department of Obstetrics, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Haiyan Yang
- Department of Obstetrics, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Ronghui Liu
- Department of Obstetrics, Yantaishan Hospital, Yantai, Shandong, 264003, China.
| |
Collapse
|
2
|
Hetherington K, Thomas J, Nicholls SJ, Barsha G, Bubb KJ. Unique cardiometabolic factors in women that contribute to modified cardiovascular disease risk. Eur J Pharmacol 2024; 984:177031. [PMID: 39369878 DOI: 10.1016/j.ejphar.2024.177031] [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: 04/30/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Major risk factors of cardiovascular disease (CVD) include hypertension, obesity, diabetes mellitus and metabolic syndrome; all of which are considered inflammatory conditions. Women are disproportionately affected by inflammatory conditions, with sex differences emerging as early as adolescence. Hormonal fluctuations associated with reproductive events such as menarche, pregnancy and menopause, are hypothesized to promote a pro-inflammatory state in women. Moreover, women who have experienced inflammatory-type conditions such as polycystic ovarian syndrome (PCOS), gestational diabetes or pre-eclampsia, have a cardiometabolic phenotype that pre-disposes to increased risk of myocardial infarction, stroke and coronary heart disease. Women with no notable CVD risk factors are often relatively protected from CVD pre-menopause; but overtake men in risk of major cardiovascular events when the cardiovascular protective effects of oestrogen begin to wane. Sex differences and female-specific factors have long been considered challenging to study and this has led to an underrepresentation of females in clinical trials and lack of female-specific data from pre-clinical studies. However, there is now a clear prerogative to include females at all stages of research, despite inherent complexities and potential variability in data. This review explores recent advancements in our understanding of CVD in women. We summarise the underlying factors unique to women that can promote CVD risk factors, ultimately contributing to CVD burden and the emerging therapies aimed to combat this.
Collapse
Affiliation(s)
- Kara Hetherington
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3800, Australia; Victorian Heart Institute, Victorian Heart Hospital, Clayton, Victoria, 3168, Australia
| | - Jordyn Thomas
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3800, Australia; Victorian Heart Institute, Victorian Heart Hospital, Clayton, Victoria, 3168, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, Victorian Heart Hospital, Clayton, Victoria, 3168, Australia
| | - Giannie Barsha
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3800, Australia; Victorian Heart Institute, Victorian Heart Hospital, Clayton, Victoria, 3168, Australia
| | - Kristen J Bubb
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3800, Australia; Victorian Heart Institute, Victorian Heart Hospital, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
3
|
Kiolbasa M, Kotlarz A, Kaminiów K, Pastuszczak M. Elevated IL-10 serum levels are associated with slower serological response following syphilis treatment during pregnancy. Int J STD AIDS 2024:9564624241303816. [PMID: 39627163 DOI: 10.1177/09564624241303816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
BACKGROUND Approximately 50% of pregnant individuals treated for syphilis do not achieve a decline in non-treponemal titers by delivery. The serological response in pregnant persons is significantly slower compared to non-pregnant individuals, with unclear pathogenesis and clinical significance. This study aimed to determine the association between the host immune response and serological outcome in pregnant individuals with syphilis. METHODS Twenty-four females with early syphilis, including 14 pregnant individuals, were included. Pro-inflammatory and regulatory cytokines (IFN-γ, TNF-α, IL-4, IL-1β, IL-10, TGF-β) were measured before treatment and 6 months after penicillin injection. RESULTS The median time to serological cure was 5 months for pregnant individuals and 2 months for non-pregnant females. Pregnant individuals had significantly higher serum levels of IL-10 and TGF-β at baseline and at 6 months post-treatment compared to non-pregnant individuals (p < .05). CONCLUSIONS A robust regulatory immune response to syphilis may be associated with a slower serological response to therapy during pregnancy.
Collapse
Affiliation(s)
- Martyna Kiolbasa
- Clinical Department of Dermatology, Medical University of Silesia, Zabrze, Poland
| | - Agnieszka Kotlarz
- Department of Gynaecology and Obstetrics, Jagiellonian University Medical College, Krakow, Polnad
| | - Konrad Kaminiów
- Clinical Department of Dermatology, Medical University of Silesia, Zabrze, Poland
| | - Maciej Pastuszczak
- Clinical Department of Dermatology, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
4
|
Kindschuh WF, Austin GI, Meydan Y, Park H, Urban JA, Watters E, Pollak S, Saade GR, Chung J, Mercer BM, Grobman WA, Haas DM, Silver RM, Serrano M, Buck GA, McNeil R, Nandakumar R, Reddy U, Wapner RJ, Kav AB, Uhlemann AC, Korem T. Early prediction of preeclampsia using the first trimester vaginal microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626267. [PMID: 39677801 PMCID: PMC11642775 DOI: 10.1101/2024.12.01.626267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Preeclampsia is a severe obstetrical syndrome which contributes to 10-15% of all maternal deaths. Although the mechanisms underlying systemic damage in preeclampsia-such as impaired placentation, endothelial dysfunction, and immune dysregulation-are well studied, the initial triggers of the condition remain largely unknown. Furthermore, although the pathogenesis of preeclampsia begins early in pregnancy, there are no early diagnostics for this life-threatening syndrome, which is typically diagnosed much later, after systemic damage has already manifested. Here, we performed deep metagenomic sequencing and multiplex immunoassays of vaginal samples collected during the first trimester from 124 pregnant individuals, including 62 who developed preeclampsia with severe features. We identified multiple significant associations between vaginal immune factors, microbes, clinical factors, and the early pathogenesis of preeclampsia. These associations vary with BMI, and stratification revealed strong associations between preeclampsia and Bifidobacterium spp., Prevotella timonensis, and Sneathia vaginalis. Finally, we developed machine learning models that predict the development of preeclampsia using this first trimester data, collected ~5.7 months prior to clinical diagnosis, with an auROC of 0.78. We validated our models using data from an independent cohort (MOMS-PI), achieving an auROC of 0.80. Our findings highlight robust associations among the vaginal microbiome, local host immunity, and early pathogenic processes of preeclampsia, paving the way for early detection, prevention and intervention for this devastating condition.
Collapse
Affiliation(s)
- William F. Kindschuh
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - George I. Austin
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Yoli Meydan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia A. Urban
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily Watters
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Susan Pollak
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - George R. Saade
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Judith Chung
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of California Irvine, CA, USA
| | - Brian M. Mercer
- Departments of Obstetrics and Gynecology, Case Western Reserve University, Cleveland, OH, USA
| | | | - David M. Haas
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN, USA
| | - Robert M. Silver
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
| | - Myrna Serrano
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
| | - Gregory A. Buck
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
- Department of Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Renu Nandakumar
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Uma Reddy
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Aya Brown Kav
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
5
|
Palm K, Cluver C, Langenegger E, Tong S, Walker S, Imberg H, Hastie R, Bergman L. Circulating concentrations of pro-inflammatory cytokines in preeclampsia with varying disease severity. Pregnancy Hypertens 2024; 38:101168. [PMID: 39577064 DOI: 10.1016/j.preghy.2024.101168] [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: 06/20/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVES To assess whether plasma concentrations of the circulating inflammatory proteins Interleukin-6 (IL-6), Vascular Cell Adhesion Molecule-1 (VCAM-1) and C-Reactive Protein (CRP) are increased in women with preeclampsia with end-organ complications, compared with women with preeclampsia without end-organ complications. STUDY DESIGN We used samples from a large prospective biobank collection (Preeclampsia Obstetric Adverse Event biobank), and two large, randomized preeclampsia therapeutic treatment trials. All samples were collected in Cape Town, South Africa. The last plasma sample collected prior to birth was analyzed for IL-6, VCAM-1 and CRP concentrations. We categorized cases according to disease severity and compared circulating levels of these analytes. Covariate adjustment was performed. RESULTS 183 women were included. Compared with women without end-organ complications (n = 119), those with preeclampsia with two or more end-organ complications (n = 15) had a 4.9-fold (95 % CI, 1.81-13.09, p = 0.001) increase in IL-6 and a 1.7-fold (95 % CI, 1.11-2.72, p = 0.012) increase in VCAM-1 plasma concentrations. Comparing women with two or more end-organ complications to those with one end-organ complication (n = 49), plasma concentrations of IL-6 were 3.2-fold (95 % CI, 1.18-8.39, p = 0.018) increased, while there was no statistically significant difference for VCAM-1 (1.2-fold higher, 95 % CI, 0.79-1.91, p = 0.50). Plasma concentrations of CRP did not differ between the groups. CONCLUSIONS Plasma concentrations of IL-6 and VCAM-1, but not CRP, were increased among women with preeclampsia and end-organ complications, compared with women without end-organ complications. IL-6 and VCAM-1 could be drivers of disease in preeclampsia and potentially useful to identify women at high risk of severe disease.
Collapse
Affiliation(s)
- Klara Palm
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Catherine Cluver
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa; Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Eduard Langenegger
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Susan Walker
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Henrik Imberg
- Statistiska Konsultgruppen, Gothenburg, Sweden; Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | - Roxanne Hastie
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Lina Bergman
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa; Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Xu Y, Qin X, Zeng W, Wu F, Wei X, Li Q, Lin Y. DOCK1 deficiency drives placental trophoblast cell dysfunction by influencing inflammation and oxidative stress, hallmarks of preeclampsia. Hypertens Res 2024; 47:3434-3446. [PMID: 39379467 DOI: 10.1038/s41440-024-01920-3] [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: 04/15/2024] [Revised: 08/21/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024]
Abstract
Preeclampsia (PE) is a globally prevalent obstetric disorder, pathologically characterized by abnormal placental development. Dysfunctions of angiogenesis, vasculogenesis and spiral artery remodeling are demonstrated to be involved in PE pathogenesis; however, the underlying mechanisms remain largely unknown. Here, we investigated the role of the dedicator of cytokinesis 1 (DOCK1), crucial molecule in various cellular processes, in PE progression using HTR-8 cells derived from first-trimester placental extravillous trophoblasts. Our analysis revealed an aberrant DOCK1 expression in the placental villi of PE patients and its impact on essential cellular functions for vascular network formation. A deficiency of DOCK1 in HTR-8 cells impaired the vascular network formation, exacerbated the expression of anti-angiogenic factor ENG, and reduced VEGF levels. Moreover, DOCK1 knockout amplified apoptosis, as indicated by an altered BCL2: BAX ratio and enhanced levels of cleaved PARP. DOCK1 depletion also boosted NF-κB activation and pro-inflammatory cytokine production (IL-6 and TNF-α). Furthermore, the mice treated with DOCK1 inhibitor, TBOPP, exhibited PE-like symptoms. These findings highlight the multifaceted roles of DOCK1 in the pathophysiology of PE, demonstrating that its deficiency can lead to placental dysfunction by orchestrating inflammatory responses and oxidative stress. These insights emphasize the pathogenic role of DOCK1 in PE development and suggest potential treatment strategies that require further exploration. In the graphical abstract, a split image of placental villi contrasts the effects of normal and reduced DOCK1 expression on preeclampsia. The left side illustrates adequate DOCK1 levels supporting healthy trophoblast function and effective spiral artery remodeling. The right side highlights the consequences of DOCK1 deficiency, leading to trophoblast dysfunction and impaired spiral artery remodeling, accompanied by angiogenic imbalance, increased inflammation, oxidative stress, and apoptosis, contributing to placental dysfunction and the development of preeclampsia.
Collapse
Affiliation(s)
- Yichi Xu
- Reproductive Medicine Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoli Qin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weihong Zeng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowei Wei
- Reproductive Medicine Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- Reproductive Medicine Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Serrano-Lomelin J, Smith GN, Davidge ST, Riddell M, Chari R, Crawford S, Bakal JA, Ospina MB. Associations of Diabetes, Mental Health, and Asthma with Hypertensive Disorders of Pregnancy: A Population-based Case-Control Study in Alberta, Canada. Pregnancy Hypertens 2024; 38:101172. [PMID: 39581177 DOI: 10.1016/j.preghy.2024.101172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 06/25/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE To explore direct and indirect associations of diabetes, mental health, and asthma diagnosed before or during pregnancy with gestational hypertension (GH) or preeclampsia (PE). STUDY DESIGN This population-based case-control study conducted in Alberta, Canada, analyzed perinatal registry data from primiparous pregnant women aged 16 years and above, with no prior hypertension history, during the period 2010 to 2013. Cases of GH and PE were matched on gestational age with a random sample of controls at a 1:3 ratio. MAIN OUTCOME MEASURES We examined the presence of type 2 diabetes mellitus (T2DM) or gestational diabetes, depression, anxiety, and asthma diagnoses within five years before and during pregnancy. To estimate direct and indirect associations between these diagnoses and GH and PE, we used multivariable logistic and mediation models, adjusting for covariates. RESULTS The analysis included 18,381 women (3,443 GH cases, 1,152 PE cases, and 13,786 controls). We found a direct association between anxiety during pregnancy and GH (adjusted Odds Ratio [aOR] 2.18, 95 % confidence interval (CI) 1.43-3.31). Depression before pregnancy increased the odds of anxiety during pregnancy (aOR 4.78, 95 % CI 2.89-7.92) resulting in an indirect effect on GH (aOR 3.63, 95 % CI 1.67--7.87). For PE, we observed direct associations with pre-pregnancy T2DM (aOR 1.58, 95 % CI 1.12-2.24), gestational diabetes (aOR 1.28, 95 % CI 1.04-1.56), and asthma during pregnancy (aOR 2.23, 95 % CI 1.41-3.51). CONCLUSION These findings highlight the interplay of mental health factors in influencing GH and underscore the clinical importance of diabetes and asthma in the pathogenesis of PE.
Collapse
Affiliation(s)
- Jesus Serrano-Lomelin
- Department of Public Health Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Graeme N Smith
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sandra T Davidge
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada; Department of Obstetrics & Gynaecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Meghan Riddell
- Women and Children's Health Research Institute, Edmonton, Alberta, Canada; Department of Obstetrics & Gynaecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Radha Chari
- Department of Obstetrics & Gynaecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Crawford
- Maternal Newborn Child and Youth Strategic Clinical Network, Alberta Health Services, Calgary, Alberta, Canada
| | - Jeffrey A Bakal
- Provincial Research Data Services, Alberta Health Services, Edmonton, Alberta, Canada
| | - Maria B Ospina
- Department of Public Health Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada; Women and Children's Health Research Institute, Edmonton, Alberta, Canada; Department of Obstetrics & Gynaecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
8
|
Mack JA, Burkholder A, Akhtari FS, House JS, Sovio U, Smith GCS, Schmitt CP, Fargo DC, Hall JE, Motsinger-Reif AA. A multi-ancestry genome-wide association study identifies novel candidate loci in the RARB gene associated with hypertensive disorders of pregnancy. HGG ADVANCES 2024; 6:100385. [PMID: 39580622 DOI: 10.1016/j.xhgg.2024.100385] [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: 05/30/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Genetic factors related to pregnancy-related traits are understudied, especially in ancestrally diverse cohorts. To assess maternal contributions to hypertensive disorders of pregnancy (HDP), we performed a multi-ancestry genome-wide association study (GWAS) of HDP in data from the North Carolina-based Personalized Environment and Genes Study (PEGS) cohort with validation in the UK Biobank (UKBB). The GWAS revealed two maternal loci associated with HDP at the genome-wide significance level. The lead independent variants were rs114954125 on chromosome 2 (near LRP1B; odds ratio [OR] [95% confidence interval {CI}]): 2.96 [2.02-4.34]; p = 2.82 × 10-8) and rs61176331 on chromosome 3 (on RARB; OR (95% CI): 3.08 (2.12-4.48); p = 3.52 × 10-9). We validated the associations near RARB with a meta-analysis of PEGS and the UKBB. We also identified cis-expression quantitative trait loci in the candidate region associated with decreased RARB expression in macrophage cells exposed to Salmonella. Chromatin mapping in FUMA identified a significant interaction within chromosome 3's enhancer and open chromatin regions, with strong effects observed for RARB and H3P10 gene regulation in mesendoderm cells, mesenchymal stem cells, and trophoblast-like stem cells. We applied existing polygenic scores (PGS) for preeclampsia and gestational hypertension and found that the scores were significantly associated with HDP in PEGS. The findings demonstrate the power of multi-ancestry studies for genetic discovery and highlight the relationship between immune response, regulation, and HDP and the utility of PGS for risk prediction. (PEGS is registered at ClinicalTrials.gov: NCT00341237.).
Collapse
Affiliation(s)
- Jasmine A Mack
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Adam Burkholder
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Farida S Akhtari
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - John S House
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ulla Sovio
- University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Gordon C S Smith
- University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Charles P Schmitt
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David C Fargo
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janet E Hall
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
9
|
Cainelli E, Vedovelli L, Bisiacchi P. The mother-child interface: A neurobiological metamorphosis. Neuroscience 2024; 561:92-106. [PMID: 39427701 DOI: 10.1016/j.neuroscience.2024.10.027] [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: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
From the start of pregnancy, mother and child induce reciprocal neurobiological changes in the brain that will prove critical for neurodevelopment and survival of both. Molecular communication between mother and fetus is constantly active and persists even after the fetus starts to synthesize its hormones in late gestation. Intriguingly, some mother and fetus exchange cells remain in the other's brain and body with long-lasting effects and memories that do not follow the laws of classical genetics but involve complex epigenetic mechanisms. After childbirth, mother and child go through a transitional phase, a sort of limbo in which both will have a peculiar functioning profile, which is adaptive for contingencies but also renders them vulnerable. The interplay between these two "limbo" states allows for an easier transition to the subsequent phases of development. In this review, we will trace mother's and child's path from pregnancy to the months following birth and, in particular, unravel i) the key features of pregnancy and brain development and the reciprocal influences; ii) how a transitory pattern of functioning characterize mother and child, moving them toward more flexible and evolved forms; and iii) how mother and fetus act during childbirth to promote neuroprotection, pain reduction, and neurophysiological changes. Therefore, this review covers a wide range of topics, integrating neuroanatomical, neurological, biochemical, neurophysiological, and psychological studies in a meaningful way, trying to integrate them in a holistic view of the mother-child interface that is usually neglected.
Collapse
Affiliation(s)
- Elisa Cainelli
- Department of General Psychology, University of Padova, 35131 Padova, Italy.
| | - Luca Vedovelli
- Unit of Biostatistics, Epidemiology, and Public Health, Department of Cardiac, Thoracic, Vascular and Public Health Sciences, University of Padova, 35131 Padova, Italy.
| | - Patrizia Bisiacchi
- Department of General Psychology, University of Padova, 35131 Padova, Italy; Padova Neuroscience Center, PNC, 35131 Padova, Italy.
| |
Collapse
|
10
|
Garcia de Leon R, Hodges TE, Brown HK, Bodnar TS, Galea LAM. Inflammatory signalling during the perinatal period: Implications for short- and long-term disease risk. Psychoneuroendocrinology 2024; 172:107245. [PMID: 39561569 DOI: 10.1016/j.psyneuen.2024.107245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024]
Abstract
During pregnancy and the postpartum, there are dynamic fluctuations in steroid and peptide hormone levels as well as inflammatory signalling. These changes are required for a healthy pregnancy and can persist well beyond the postpartum. Many of the same hormone and inflammatory signalling changes observed during the perinatal period also play a role in symptoms related to autoimmune disorders, psychiatric disorders, and perhaps neurodegenerative disease later in life. In this review, we outline hormonal and immunological shifts linked to pregnancy and the postpartum and discuss the possible role of these shifts in increasing psychiatric, neurodegenerative disease risk and autoimmune symptoms during and following pregnancy. Furthermore, we discuss how key variables such as the number of births (parity) and sex of the fetus can influence inflammatory signalling, and possibly future disease risk, but are not often studied. We conclude by discussing the importance of studying female experiences such as pregnancy and parenting on physiology and disease.
Collapse
Affiliation(s)
- Romina Garcia de Leon
- Centre for Addiction and Mental Health, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada
| | | | | | | | - Liisa A M Galea
- Centre for Addiction and Mental Health, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Chen YH, Chang YC, Wu WJ, Chen M, Yen CC, Lan YW, Cheng HC, Chen CM. Kefir peptides mitigate L-NAME-induced preeclampsia in rats through modulating hypertension and endothelial dysfunction. Biomed Pharmacother 2024; 180:117592. [PMID: 39490048 DOI: 10.1016/j.biopha.2024.117592] [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: 08/06/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
AIM Preeclampsia is a complex and serious pregnancy disorder that leads to maternal and neonatal mortality worldwide. Kefir peptides (KPs), derived from various prebiotic fermentations in whole milk by kefir grains, were investigated for their potential therapeutic effects. In this study, we used the L-NAME in drinking water to induce a preeclampsia-like condition in spontaneous hypertension stroke-prone (SHRSP) pregnant rats. MAIN METHODS The rats were assigned to five groups: the normal group (WKY rats), the untreated group (SHRSP rat control pregnant), the L-NAME/Mock group (SHRSP rats fed with L-NAME water), the L-NAME/KPs-LD group (SHRSP rats fed with L-NAME water and low-dose KPs diets), and the L-NAME/KPs-HD group (SHRSP rats fed with L-NAME water and high-dose KPs diets) for a 20-day experiment. Chorioallantois membrane (CAM) assay was applied for ex vivo angiogenesis study of KPs treatment. KEY FINDINGS Data showed that rats in the L-NAME group developed severe hypertension, proteinuria, placental damage, and embryo resorption. Pre-administration of KPs significantly reduced hypertension, proteinuria, improved generalized endothelial dysfunction, and decreased levels of anti-HIF-1α, sFLT1, anti-TNF-α, and IL-6 in the placenta of SHRSP rats. In ex vivo CAM study, L-NAME administration in chicken embryos resulted in lower vessel density and hemorrhage; however, angiogenesis was observed after KPs-HD treatment. SIGNIFICANCE The results indicate that kefir peptides improve renal lesions, prevent renal parenchyma damage, and balance endothelial and angiogenic dysfunction in both maternal and fetal sites in L-NAME-induced SHRSP pregnant rats.
Collapse
Affiliation(s)
- Yu-Hsuan Chen
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan.
| | - Yo-Cheng Chang
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan.
| | - Wan-Ju Wu
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Min Chen
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan; Department of Genomic Medicine, Changhua Christian Hospital, Changhua 50046, Taiwan.
| | - Chih-Ching Yen
- Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung 404, Taiwan.
| | - Ying-Wei Lan
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Phoenix Children's Health Research Institute, Department of Child Health, University of Arizona College of Medicine, Phoenix 85004, USA.
| | - Hsu-Chen Cheng
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan.
| | - Chuan-Mu Chen
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Center for General Educational, National Quemoy University, Kinmen 892, Taiwan.
| |
Collapse
|
12
|
Bernier E, Couture C, Borchers A, Brien ME, Graham CH, Girard S. Circulating Immune Cells from Early- and Late-onset Pre-eclampsia Displays Distinct Profiles with Differential Impact on Endothelial Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1292-1304. [PMID: 39302114 PMCID: PMC11491498 DOI: 10.4049/jimmunol.2400196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
Pre-eclampsia (PE) affects 5-8% of pregnancies and has detrimental effects on maternal-fetal health. PE is characterized by de novo hypertension after 20 wk of gestation and end-organ damage. Systemic inflammatory imbalance has been associated with PE, but its contribution to the pathology is poorly understood. Our objective was to investigate maternal systemic immune changes in early-onset PE (EOPE) and late-onset PE (LOPE) versus uncomplicated pregnancies (control [CTRL]), and their contribution to endothelial activation, hallmark of hypertension. Blood samples were analyzed by flow cytometry, multiplex assay, intracellular cytokine staining, and single-cell RNA sequencing. We performed cocultures between circulating immune cells and HUVECs to assess endothelial activation. We found that EOPE had decreased regulatory T cells (4.64±0.33, p < 0.05) and monocytes (33.92±3.08, p < 0.01), whereas LOPE had decreased regulatory T cells (4.60±0.30, p < 0.05) and Th2 cells (7.50±0.62, p < 0.01) versus CTRL. Compared to CTRL, elevated cytokines/chemokines, and growth factors were observed in LOPE, whereas EOPE primarily showed decreased levels. Using intracellular cytokine staining, we observed more monocytes producing IL-12, TNF-α, and IL-1β (all p < 0.05) in LOPE versus CTRL. At the transcriptomic level, we found differentially expressed genes between EOPE and CTRL, predominantly related to upregulation of immune activation pathways. Lastly, EOPE PBMCs induced heightened endothelial activation in vitro observed by increased ICAM-1 and ET-1 (p < 0.05), whereas LOPE PBMCs required LPS stimulation. Although significant proteomic changes are observed in the LOPE group, the EOPE displayed changes mostly at the transcriptomic levels and could induce endothelial activation in vitro.
Collapse
Affiliation(s)
- Elsa Bernier
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Camille Couture
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Anna Borchers
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Immunology, Mayo Clinic, Rochester, MN
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Sylvie Girard
- Department of Immunology, Mayo Clinic, Rochester, MN
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN
| |
Collapse
|
13
|
Gharipour M, Craig JM, Stephenson G. Epigenetic programming of obesity in early life through modulation of the kynurenine pathway. Int J Obes (Lond) 2024:10.1038/s41366-024-01647-8. [PMID: 39424650 DOI: 10.1038/s41366-024-01647-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024]
Abstract
Childhood obesity is a global health concern that has its origins before birth. Although genetics plays a crucial role, increasing evidence suggests that epigenetic modifications during fetal life could also influence its incidence. In this model, during the fetal period, interactions between genetic makeup, intrauterine factors, and environmental conditions, increase the risk of childhood obesity. This is in accordance with the Developmental Origins of Health and Disease (DOHaD) hypothesis, in which specific intrauterine environments can have long-lasting effects on the immune system's essential functions during crucial stages of fetal growth, resulting in permanent changes to the immune function of the offspring. Consequently, dysfunction can consequently make the offspring more prone to inflammatory and immune-related disorders later in life. In this review, we examine how maternal inflammation could influence the risk of childhood obesity. We propose that during pregnancy, modification of the expression of critical genes in metabolic and signaling pathways, such as the kynurenine (Kyn) pathway, occurs due to increased levels of maternal inflammation. We also propose that such expression differences are mediated by epigenetic changes. Furthermore, we also hypothesize that the Kyn pathway produces metabolites that have immunoregulatory effects and may play a crucial role in regulating inflammation during pregnancy. As a result, interventions aimed at improving maternal inflammation may be able to help alleviate the risk of childhood obesity.
Collapse
Affiliation(s)
- Mojgan Gharipour
- School of Medicine, Faculty of Health at Deakin University, Melbourne, VIC, Australia.
| | - Jeffrey M Craig
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Murdoch Children's Research Institute, Department of Pediatrics, The University of Melbourne, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Garth Stephenson
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
14
|
Chen A, Tian M, Luo Z, Cao X, Gu Y. Analysis of the evolution of placental oxidative stress research from a bibliometric perspective. Front Pharmacol 2024; 15:1475244. [PMID: 39484166 PMCID: PMC11524950 DOI: 10.3389/fphar.2024.1475244] [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: 08/03/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Background Research on placental oxidative stress is pivotal for comprehending pregnancy-related physiological changes and disease mechanisms. Despite recent advancements, a comprehensive review of current status, hotspots, and trends remains challenging. This bibliometric study systematically analyzes the evolution of placental oxidative stress research, offering a reference for future studies. Objective To conduct a comprehensive bibliometric analysis of the literature on placental oxidative stress to identify research hotspots, trends, and key contributors, thereby providing guidance for future research. Methods Relevant data were retrieved from the Web of Science Core Collection database and analyzed using VOSviewer, CiteSpace, and the bibliometrix package. An in-depth analysis of 4,796 publications was conducted, focusing on publication year, country/region, institution, author, journal, references, and keywords. Data collection concluded on 29 April 2024. Results A total of 4,796 papers were retrieved from 1,173 journals, authored by 18,835 researchers from 4,257 institutions across 103 countries/regions. From 1991 to 2023, annual publications on placental oxidative stress increased from 7 to 359. The United States (1,222 publications, 64,158 citations), the University of Cambridge (125 publications, 13,562 citations), and Graham J. Burton (73 publications, 11,182 citations) were the most productive country, institution, and author, respectively. The journal Placenta had the highest number of publications (329) and citations (17,152), followed by the International Journal of Molecular Sciences (122 publications). The most frequent keywords were "oxidative stress," "expression," "pregnancy," "preeclampsia," and "lipid peroxidation." Emerging high-frequency keywords included "gestational diabetes mellitus," "health," "autophagy," "pathophysiology," "infection," "preterm birth," "stem cell," and "inflammation." Conclusion Over the past 3 decades, research has concentrated on oxidative stress processes, antioxidant mechanisms, pregnancy-related diseases, and gene expression regulation. Current research frontiers involve exploring pathophysiology and mechanisms, assessing emerging risk factors and environmental impacts, advancing cell biology and stem cell research, and understanding the complex interactions of inflammation and immune regulation. These studies elucidate the mechanisms of placental oxidative stress, offering essential scientific evidence for future intervention strategies, therapeutic approaches, and public health policies.
Collapse
Affiliation(s)
| | | | | | - Xiaohui Cao
- Department of Obstetrics and Gynecology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Yanfang Gu
- Department of Obstetrics and Gynecology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
15
|
Gatarayiha A, Ntaganira J, Brookes Z, Mutesa L, Gustafsson A, Rulisa S. Periodontitis and pre-eclampsia among pregnant women in Rwanda: A case-control study. PLoS One 2024; 19:e0312103. [PMID: 39401230 PMCID: PMC11472930 DOI: 10.1371/journal.pone.0312103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 10/01/2024] [Indexed: 10/17/2024] Open
Abstract
INTRODUCTION Several studies have indicated that the presence of periodontitis during pregnancy could increase the risk of developing pre-eclampsia, thereby negatively influencing pregnancy outcomes for both the mother and child. Notably, despite the high prevalence of both periodontitis and adverse pregnancy outcomes in Rwanda, there exists a crucial evidence gap concerning the precise relationship between periodontitis and pre-eclampsia. OBJECTIVES The aim of this study was to assess the association between periodontitis and pre-eclampsia amongst pregnant women in Rwanda. METHODS AND MATERIALS Employing an unmatched 1:2 case-control design, we studied 52 pre-eclamptic and 104 non-pre-eclamptic pregnant women aged ≥18 years at two referral hospitals in Rwanda. Pre-eclampsia was defined as a systolic blood pressure ≥ 140 and diastolic blood pressure ≥ 90 mm Hg, diagnosed after 20 weeks of gestation and proteinuria of ≥300mL in 24 hours of urine collection. Periodontitis was defined as the presence of two or more teeth with one or more sites with a pocket depth ≥ 4mm and clinical attachment loss >3 mm at the same site, assessed through clinical attachment loss measurement. Bivariate analysis and logistic regression were used to estimate Odds ratio (ORs) and 95% confidence interval. RESULTS The prevalence of periodontitis was significantly higher among women with pre-eclampsia, compared to pregnant women without pre-eclampsia, at 90.4% and 55.8%, respectively (p< 0.001). Pregnant Women with periodontitis were 3.85 times more likely to develop pre-eclampsia after controlling for relevant confounders (adjusted Odds Ratio [aOR] = 3.85, 95%CI = 1.14-12.97, p<0.05). CONCLUSION This study results indicates that periodontitis is significantly associated with pre-eclampsia among pregnant women in Rwanda. These findings suggest that future research should explore whether enhancing periodontal health during pregnancy could contribute to reducing pre-eclampsia in this specific population.
Collapse
Affiliation(s)
- Agnes Gatarayiha
- College of Medicine and Health Sciences, School of Dentistry, University of Rwanda, Kigali, Rwanda
| | - Joseph Ntaganira
- College of Medicine and Health Sciences, School of Public Health, University of Rwanda, Kigali, Rwanda
| | - Zoe Brookes
- Peninsula Dental School, University of Plymouth, Plymouth, United Kingdom
| | - Léon Mutesa
- Centre for Human Genetics, College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| | | | - Stephen Rulisa
- College of Medicine and Health Sciences, School of Medicine and Pharmacy, University of Rwanda, Kigali, Rwanda
| |
Collapse
|
16
|
Deer E, Herrock O, Simmons K, Campbell N, Amaral L, Zheng B, Morris R, Wallace K, Cleveland ELH, Belk S, Dodd C, LaMarca B. Hypertension and Cognitive Dysfunction in a Pregnant Rat Model of PE; a Role for CD4+ T Cells. Am J Reprod Immunol 2024; 92:e13935. [PMID: 39427297 DOI: 10.1111/aji.13935] [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: 05/10/2024] [Revised: 08/27/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE Preeclampsia (PE) is associated with hypertension (HTN) during pregnancy and activated CD4+ T cells, inflammatory cytokines, and autoantibodies to the angiotensin II type I receptor (AT1-AA). Having had COVID-19 (CV) during pregnancy is associated with an increased incidence of a PE-like phenotype. Both PE and CV have long-lasting neurological implications and studies show that nonpregnant COVID patients produce AT1-AA. We have shown that CD4+ T cells from PE women cause a PE phenotype in nude athymic rats. In this study, we sought to examine the role of CD4+ T cells from PE with a CV History (Hx) to contribute to a PE phenotype and to determine the importance of CD4+ T cells in cognitive dysfunction during pregnancy. METHODS At delivery, blood and placentas were collected, and one million placental CD4+ T cells from each PE and each normotensive patient, with (NT) or without (NP) a CV (Hx) during pregnancy, were isolated, purified, and injected i.p. into a gestational day (GD) 12 pregnant nude athymic rat (one patient/rat). At GD19, blood pressure (MAP) and circulating factors were assessed in recipient rats. Cognitive function and memory were assessed using Novel Object Recognition and Barnes Maze tests, respectively. Placental ACE-2 activity and AT1-AA were measured from COVID Hx patients. A one- or two-way ANOVA with Bonferroni's multiple comparisons test was used for statistical analysis. RESULTS Blood pressure was increased in patients with PE, with or without COVID, compared to NT patients. There were no significant changes in placental ACE activity in patients with COVID Hx with or without PE. AT1-AA was elevated in PE patients and in both PE and NT COVID Hx compared to control NP. In pregnant recipient rats, MAP increased in CV Hx PE (113 ± 2, n = 8) compared to CV Hx NT (101 ± 5, n = 6). PE and PE CV Hx CD4+ T Cell recipient rats exhibited impaired memory and cognitive dysfunction (p < 0.05), compared to control groups. Recipient rats of PE CV Hx CD4+ T cells had elevated AT1-AA compared to NT CV Hx recipients. Both COVID Hx groups and recipients of PE CD4+ T cells had elevated TNF alpha compared to NP. CONCLUSION Our findings indicate that pregnant patients with a Hx of COVID during pregnancy produce AT1-AA, with or without PE. Recipients of CD4+ T cells from PE with or without a CV Hx during pregnancy cause HTN and elevated AT1-AA. TNF-α is elevated in PE and in CV Hx NT and PE recipients. Interestingly, recipients of T cells from PE patients with or without a Hx of CV had worse cognitive function during pregnancy, compared to recipient rats of NP CD4+ T cells. These data demonstrate the importance of CD4+ T cells in HTN and impaired neurological function during PE in the presence or absence of a prior COVID-19 infection during pregnancy.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kimberly Simmons
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lorena Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Baoying Zheng
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Rachael Morris
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kedra Wallace
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Sheila Belk
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Cameronne Dodd
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
17
|
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
|
18
|
Kokori E, Aderinto N, Olatunji G, Komolafe R, Abraham IC, Babalola AE, Aboje JE, Ukoaka BM, Samuel O, Ayodeji A, Omoworare O, Olatunji D. Maternal and fetal neurocognitive outcomes in preeclampsia and eclampsia; a narrative review of current evidence. Eur J Med Res 2024; 29:470. [PMID: 39342384 PMCID: PMC11437679 DOI: 10.1186/s40001-024-02070-5] [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: 06/02/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP), such as preeclampsia and eclampsia, present significant risks to maternal and fetal health. While immediate complications are well-documented, emerging research highlights potential neurocognitive impacts on both mothers and their offspring. This narrative review synthesizes evidence on these neurocognitive outcomes associated with HDP, focusing on preeclampsia and eclampsia. A literature search was conducted for studies published from 2000 to February 2024. Maternal outcomes, including memory, executive function, and psychosocial well-being, were assessed across 11 studies, while fetal and neonatal neurocognitive outcomes were explored in five studies. Consistent findings indicate that preeclampsia and eclampsia are linked to impairments in maternal cognitive functions and psychosocial health. Offspring exposed to these conditions in utero also show cognitive deficits and alterations in brain connectivity. Contributing factors include placental dysfunction, altered angiokine levels, maternal stress, and socioeconomic variables. To mitigate these impacts, future research should focus on clarifying the underlying mechanisms and developing early interventions. This review emphasizes the necessity of multidisciplinary approaches to improve neurocognitive outcomes for both mothers and their children affected by preeclampsia and eclampsia.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine, Ladoke Akintola University of Technology, PMB 5000, Ogbomoso, Nigeria.
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Rosemary Komolafe
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | | | - John Ehi Aboje
- College of Health Sciences, Benue State University, Benue, Nigeria
| | | | | | - Akinmeji Ayodeji
- Department of Medicine and Surgery, Olabisi Onabanjo University, Ogun, Nigeria
| | | | - Doyin Olatunji
- Department of Health Sciences, Western Illinois University, Macomb, USA
| |
Collapse
|
19
|
Couture C, Caron M, St-Onge P, Brien ME, Sinnett D, Dal Soglio D, Girard S. Identification of divergent placental profiles in clinically distinct pregnancy complications revealed by the transcriptome. Placenta 2024; 154:184-192. [PMID: 39042974 DOI: 10.1016/j.placenta.2024.07.008] [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: 04/25/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Pregnancy complications, including preeclampsia (PE), preterm birth (PTB), and intra-uterine growth restriction (IUGR) have individually been associated with inflammation but the combined comparative analysis of their placental profiles at the transcriptomic and histological levels is lacking. METHODS Bulk RNA-sequencing of human placental biopsies from uncomplicated term pregnancies (CTL) and pregnancies complicated with early-onset (EO), and late-onset (LO) PE, as well as PTB and term IUGR were used to characterize individual molecular profiles. We also applied immune-cell-specific cellular deconvolution to address local immune cell compositions and analyzed placental lesions by histology to further characterize these complications. RESULTS Transcriptome analysis revealed that clinically distinct complications differentiated themselves in unique ways compared to CTLs. Only TMEM136 was commonly modulated. Compared to CTLs, we found that PTB and IUGR were the most distinct, with LOPE being the least distinct. PTB and IUGR revealed differently enhanced inflammatory pathways, where PTB had general inflammatory responses and IUGR had immune cell activation. This inflammation was reflected in the histological profile for PTB only, whereas structural lesions were elevated in all complications. Placental lesions additionally had corresponding enhancement in inflammatory and structural biological processes. We observed that having co-complications, particularly for PTB with or without IUGR, impacted placental transcriptomes. Lastly, cellular deconvolution uncovered shared immune features among the complications. DISCUSSION Overall, we provide evidence that these pregnancy complications are not only distinct in their clinical manifestations but also in their placental profiles, which could be leveraged to understand their underlying mechanisms and could offer therapeutic targets.
Collapse
Affiliation(s)
- Camille Couture
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada; Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Maxime Caron
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montreal, Quebec, Canada
| | - Pascal St-Onge
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Daniel Sinnett
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada; Department of Pediatrics, Université de Montreal, Montreal, Quebec, Canada
| | - Dorothée Dal Soglio
- Department of Pathology and Cellular Biology, Université de Montréal, Montreal, QC, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada; Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Elgazzaz M, Woodham PC, Maher J, Faulkner JL. Implications of pregnancy on cardiometabolic disease risk: preeclampsia and gestational diabetes. Am J Physiol Cell Physiol 2024; 327:C646-C660. [PMID: 39010840 PMCID: PMC11427017 DOI: 10.1152/ajpcell.00293.2024] [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: 05/02/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Cardiometabolic disorders, such as obesity, insulin resistance, and hypertension, prior to and within pregnancy are increasing in prevalence worldwide. Pregnancy-associated cardiometabolic disease poses a great risk to the short- and long-term well-being of the mother and offspring. Hypertensive pregnancy, notably preeclampsia, as well as gestational diabetes are the major diseases of pregnancy growing in prevalence as a result of growing cardiometabolic disease prevalence. The mechanisms whereby obesity, diabetes, and other comorbidities lead to preeclampsia and gestational diabetes are incompletely understood and continually evolving in the literature. In addition, novel therapeutic avenues are currently being explored in these patients to offset cardiometabolic-induced adverse pregnancy outcomes in preeclamptic and gestational diabetes pregnancies. In this review, we discuss the emerging pathophysiological mechanisms of preeclampsia and gestational diabetes in the context of cardiometabolic risk as well as the most recent preclinical and clinical updates in the pathogenesis and treatment of these conditions.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Padmashree C Woodham
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - James Maher
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
21
|
Liang R, Panelli DM, Stevenson DK, Rehkopf DH, Shaw GM, Sørensen HT, Pedersen L. Outcome of Pregnancy Oral Glucose Tolerance Test and Preterm Birth. Epidemiology 2024; 35:701-709. [PMID: 38771706 PMCID: PMC11305920 DOI: 10.1097/ede.0000000000001752] [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] [Indexed: 05/23/2024]
Abstract
BACKGROUND Gestational diabetes is associated with adverse outcomes such as preterm birth (<37 weeks). However, there is no international consensus on screening criteria or diagnostic levels for gestational diabetes, and it is unknown whether body mass index (BMI) or obesity modifies the relation between glucose level and preterm birth. METHODS We studied a pregnancy cohort restricted to two Danish regions from the linked Danish Medical Birth Register to study associations between glucose measurements from the 2-hour postload 75-g oral glucose tolerance test (one-step approach) and preterm birth from 2004 to 2018. In Denmark, gestational diabetes screening is a targeted strategy for mothers with identified risk factors. We used Poisson regression to estimate rate ratios (RR) of preterm birth with z-standardized glucose measurements. We assessed effect measure modification by stratifying analyses and testing for heterogeneity. RESULTS Among 11,337 pregnancies (6.2% delivered preterm), we observed an adjusted preterm birth RR of 1.2 (95% confidence interval [CI] = 1.1, 1.3) for a one-standard deviation glucose increase of 1.4 mmol/l from the mean of 6.7 mmol/l. There was evidence for effect measure modification by obesity, for example, adjusted RR for nonobese (BMI, <30): 1.2 (95% CI = 1.1, 1.3) versus obese (BMI, ≥30): 1.3 (95% CI = 1.2-1.5), P = 0.05 for heterogeneity. CONCLUSION Among mothers screened for gestational diabetes, increased glucose levels, even those below the diagnostic level for gestational diabetes in Denmark, were associated with increased preterm birth risk. Obesity (BMI, ≥30) may be an effect measure modifier, not just a confounder, of the relation between blood glucose and preterm birth risk.
Collapse
Affiliation(s)
- Richard Liang
- Stanford University School of Medicine, Department of Epidemiology and Population Health
| | - Danielle M. Panelli
- Stanford University School of Medicine, Division of Maternal-Fetal Medicine and Obstetrics, Department of Obstetrics and Gynecology
| | - David K. Stevenson
- Stanford University School of Medicine, Department of Pediatrics, Division of Neonatal and Developmental Medicine, March of Dimes Prematurity Research Center at Stanford University School of Medicine
| | - David H. Rehkopf
- Stanford University School of Medicine, Department of Epidemiology and Population Health
- Stanford University School of Medicine, Division of Primary Care and Population Health
- Stanford University, Department of Sociology
- Stanford University, Center for Population Health Sciences
| | - Gary M. Shaw
- Stanford University School of Medicine, Department of Pediatrics, Division of Neonatal and Developmental Medicine, March of Dimes Prematurity Research Center at Stanford University School of Medicine
| | - Henrik Toft Sørensen
- Aarhus University and Aarhus University Hospital, Department of Clinical Epidemiology
- Stanford University, Clinical Excellence Research Center
| | - Lars Pedersen
- Aarhus University and Aarhus University Hospital, Department of Clinical Epidemiology
| |
Collapse
|
22
|
Manoharan MM, Montes GC, Acquarone M, Swan KF, Pridjian GC, Nogueira Alencar AK, Bayer CL. Metabolic theory of preeclampsia: implications for maternal cardiovascular health. Am J Physiol Heart Circ Physiol 2024; 327:H582-H597. [PMID: 38968164 PMCID: PMC11442029 DOI: 10.1152/ajpheart.00170.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Preeclampsia (PE) is a multisystemic disorder of pregnancy that not only causes perinatal mortality and morbidity but also has a long-term toll on the maternal and fetal cardiovascular system. Women diagnosed with PE are at greater risk for the subsequent development of hypertension, ischemic heart disease, cardiomyopathy, cerebral edema, seizures, and end-stage renal disease. Although PE is considered heterogeneous, inefficient extravillous trophoblast (EVT) migration leading to deficient spiral artery remodeling and increased uteroplacental vascular resistance is the likely initiation of the disease. The principal pathophysiology is placental hypoxia, causing subsequent oxidative stress, leading to mitochondrial dysfunction, mitophagy, and immunological imbalance. The damage imposed on the placenta in turn results in the "stress response" categorized by the dysfunctional release of vasoactive components including oxidative stressors, proinflammatory factors, and cytokines into the maternal circulation. These bioactive factors have deleterious effects on systemic endothelial cells and coagulation leading to generalized vascular dysfunction and hypercoagulability. A better understanding of these metabolic factors may lead to novel therapeutic approaches to prevent and treat this multisystemic disorder. In this review, we connect the hypoxic-oxidative stress and inflammation involved in the pathophysiology of PE to the resulting persistent cardiovascular complications in patients with preeclampsia.
Collapse
Affiliation(s)
- Mistina M Manoharan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
| | - Guilherme C Montes
- Department of Pharmacology and Psychobiology, Roberto Alcântara Gomes Institute Biology (IBRAG), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Mariana Acquarone
- Department of Neurology, Tulane University, New Orleans, Louisiana, United States
| | - Kenneth F Swan
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | - Gabriella C Pridjian
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | | | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
23
|
Che M, Xu Y, Zang Y, Zhang R, Hu J, Liu S, Zhang J. Association of NLRP3 and IL-4 VNTR polymorphisms and genetic susceptibility to preeclampsia: A case-control study. Pregnancy Hypertens 2024; 37:101142. [PMID: 38959653 DOI: 10.1016/j.preghy.2024.101142] [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: 12/29/2023] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Abnormalities in the maternal immune system and insufficient gestational immune tolerance may significantly contribute to the development of preeclampsia (PE). The NLR family pyrin domain containing 3 (NLRP3) functions as a pattern recognition receptor that identifies pathogen-associated molecular patterns. Interleukin-4 (IL-4) is a potent anti-inflammatory cytokine that modulates the immune response. Therefore, this study aims to elucidate the impact of NLRP3 and IL-4 variable number of tandem repeats (VNTR) polymorphisms on susceptibility to PE. MATERIALS AND METHODS A total of 1,018 patients with PE and 1,007 normal pregnant women were recruited as the case group and the control group, respectively. Peripheral blood DNA was extracted, and NLRP3 and IL-4 VNTR polymorphisms were genotyped using polymerase chain reaction and gel electrophoresis. Genotypes and allele frequencies of pregnant women were assessed in both cohorts. RESULTS The NLRP3 VNTR 9-7 genotype in the PE group was significantly lower than that in the control group, but 9 and 14 allele frequencies were significantly higher in patients with PE. Individuals with IL-4 VNTR genotypes 1-2 had a lower risk of PE than controls, and the IL-4 VNTR 2 allele frequency was significantly lower in patients with PE. CONCLUSIONS This study, the first of its kind in the literature, evaluates the impact of NLRP3 VNTR and IL-4 VNTR polymorphisms on PE, revealing a significant correlation with PE susceptibility. This investigation contributes to understanding the pathogenesis of PE and provides a reference point for developing strategies to prevent and treat the disease in the future.
Collapse
Affiliation(s)
- Mingxuan Che
- Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yinglei Xu
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yucui Zang
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru Zhang
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jian Hu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jidong Zhang
- Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
24
|
Youssef KK, Nieto MA. Epithelial-mesenchymal transition in tissue repair and degeneration. Nat Rev Mol Cell Biol 2024; 25:720-739. [PMID: 38684869 DOI: 10.1038/s41580-024-00733-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Epithelial-mesenchymal transitions (EMTs) are the epitome of cell plasticity in embryonic development and cancer; during EMT, epithelial cells undergo dramatic phenotypic changes and become able to migrate to form different tissues or give rise to metastases, respectively. The importance of EMTs in other contexts, such as tissue repair and fibrosis in the adult, has become increasingly recognized and studied. In this Review, we discuss the function of EMT in the adult after tissue damage and compare features of embryonic and adult EMT. Whereas sustained EMT leads to adult tissue degeneration, fibrosis and organ failure, its transient activation, which confers phenotypic and functional plasticity on somatic cells, promotes tissue repair after damage. Understanding the mechanisms and temporal regulation of different EMTs provides insight into how some tissues heal and has the potential to open new therapeutic avenues to promote repair or regeneration of tissue damage that is currently irreversible. We also discuss therapeutic strategies that modulate EMT that hold clinical promise in ameliorating fibrosis, and how precise EMT activation could be harnessed to enhance tissue repair.
Collapse
Affiliation(s)
| | - M Angela Nieto
- Instituto de Neurociencias (CSIC-UMH), Sant Joan d'Alacant, Spain.
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.
| |
Collapse
|
25
|
Tomruk C, Şirin Tomruk C, Denizlioğlu B, Olukman M, Ercan G, Duman S, Köse T, Çetin Uyanıkgil EÖ, Uyanıkgil Y, Uysal A. Effects of apelin on neonatal brain neurogenesis in L-NAME-induced maternal preeclampsia. Sci Rep 2024; 14:19347. [PMID: 39164321 PMCID: PMC11335761 DOI: 10.1038/s41598-024-69326-2] [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: 02/18/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
The aim of this study was to investigate the possible protective effects of apelin, which is known to have antioxidant and anti-inflammatory effects, on changes in neurogenesis in newborns of pregnant rats with L-NAME-induced preeclampsia. Wistar albino female rats were divided into four experimental groups: Control, Apelin, Preeclampsia and Preeclampsia + Apelin. Blood pressure was measured on the 5th, 11th and 17th days of gestation, urine protein was analyzed from urine samples collected for 24 h on the 6th, 12th and 18th days and serum creatinine was analyzed from serum samples. Maternal kidney and placenta tissues were obtained to establish the preeclampsia model, and neonatal brain tissues including the cortex, hippocampus and cerebellum regions were obtained to investigate neurogenesis and examined by histological and immunohistochemical methods. The number of newborns, body weight and brain weight of the newborns were measured. eNOS, IL-10, nNOS and NO levels in the brain analyzed via ELISA. Mean arterial pressure, urine protein and serum creatinine increased in the preeclampsia. Newborn weight decreased in the Preeclampsia group, the values in the Preeclampsia + Apelin group were closer to the Control and Apelin groups. In the Preeclampsia group, edema and dilatation in the proximal and distal tubules of kidneys, perivillous fibrin deposition and increase in syncytial nodules of placenta were observed. VEGF immunoreactivity decreased and iNOS immunoreactivity increased in both kidney and placenta. In neonatal brain tissue examinations, cytotoxic edema accompanied by thinning of cortex, delayed migration and lower cell counts in the hippocampus, and increase in intercellular spaces and EGL thickening in the cerebellum were observed in the preeclampsia. Expression of NeuN, GFAP, MBP, IL-10, eNOS, nNOS and NO levels decreased, whereas expression of Iba-1 increased in the preeclampsia. In the Preeclampsia + Apelin group, these findings were similar to the Control and Apelin groups. Apelin administration was found to be beneficial for preventing the adverse consequences of preeclampsia, but further experimental and clinical studies are needed to better understand these effects.
Collapse
Affiliation(s)
- Canberk Tomruk
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
- Histology and Embryology, Samsun Training and Research Hospital, İlkadim, Samsun, Türkiye
| | - Cansın Şirin Tomruk
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Burcu Denizlioğlu
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
- Emergency Medicine, Aydın State Hospital, Efeler, Aydın, Türkiye
| | - Murat Olukman
- Department of Medical Pharmacology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Gülinnaz Ercan
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Soner Duman
- Department of Internal Medicine, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Timur Köse
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Emel Öykü Çetin Uyanıkgil
- Department of Pharmaceutical Technology, Department of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, Ege University, Bornova, İzmir, Türkiye
| | - Yiğit Uyanıkgil
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye.
| | - Ayşegül Uysal
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| |
Collapse
|
26
|
Najeeb MN, Munir U, Hamza MA, Mehmood S, Qureshi JA, Maqbool T. Interleukin-6 (-174G/C), Interleukin-1β (-511 C/T), and Apolipoprotein B-100 (2488 C/T) Gene Polymorphism in Pre-Eclampsia. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1307. [PMID: 39202588 PMCID: PMC11356735 DOI: 10.3390/medicina60081307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024]
Abstract
Background and objectives: Pre-eclampsia (PE) is a pregnancy-specific condition characterized by significant health risks for pregnant women worldwide due to its status as a multi-organ disorder. High blood pressure (hypertension) with or without proteinuria is usually considered an initial clinical sign of PE. The pathogenesis of pre-eclampsia is highly complex and likely involves multiple factors, including poorly developed uterine spiral arterioles, immunological issues, placental ischemia or infarction, and genetic abnormalities. Inflammatory cytokine production, regulated by cytokine gene polymorphisms, is one of the factors likely contributing to the development of PE. The present study aimed to assess IL-6, IL-1β, and Apo B-100 gene polymorphism and to evaluate the association of these polymorphisms with PE. Materials and Methods: This cross-sectional observational study involved 99 participants aged 16 to 45 years from Bahawal Victoria Hospital Bahawalpur, Punjab, Pakistan. The participants were divided into three groups: Group 1 (PE with severe hypertension), Group 2 (PE with hypertension), and Group 3 (control), each comprising 33 individuals. Maternal blood samples were collected, DNA was extracted, and molecular genetic analysis of the IL-6, IL-1β, and Apo B-100 genes was performed using the PCR-RFLP method. Allelic frequencies were compared, and statistical analysis was conducted using SPSS 25, applying the Hardy-Weinberg equation and chi-square test to evaluate the results. Results: There are differences in the distribution of allelic frequencies for IL-6 -174G/C (CC, GC, GG), IL-1β-511C/T (CC, CT, TT), and Apo B-100 2488 C/T (CC, CT, TT) between pre-eclamptic patients and the control group. The analysis using the Hardy-Weinberg equilibrium and chi-square test showed an association between the IL-6-174 G/C polymorphism and the severity of pre-eclampsia. Conclusions: The polymorphisms of the IL-6, IL-1β, and Apo B-100 genes revealed different alleles. The IL-6 gene alone was found to be in disequilibrium according to the Hardy-Weinberg equation, indicating a potential link to the severity of pre-eclampsia in the population studied.
Collapse
Affiliation(s)
- Muhammad Naveed Najeeb
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 40100, Punjab, Pakistan;
- Quaid-e-Azam Medical College, Bahawalpur 63100, Punjab, Pakistan;
| | - Umaira Munir
- Quaid-e-Azam Medical College, Bahawalpur 63100, Punjab, Pakistan;
- Shahida Islam Medical College, Lodhran 59320, Punjab, Pakistan;
| | | | - Sadia Mehmood
- Bakhtawar Ameen Medical College, Multan 60800, Punjab, Pakistan;
| | - Javed Anver Qureshi
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 40100, Punjab, Pakistan;
| | - Tahir Maqbool
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 40100, Punjab, Pakistan;
| |
Collapse
|
27
|
Varshavsky JR, Meeker JD, Zimmerman E, Woodbury ML, Aung MT, Rosario-Pabon ZY, Cathey AL, Vélez-Vega CM, Cordero J, Alshawabkeh A, Eick SM. Association of Phenols, Parabens, and Their Mixture with Maternal Blood Pressure Measurements in the PROTECT Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:87004. [PMID: 39140735 PMCID: PMC11323763 DOI: 10.1289/ehp14008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Phenols and parabens are two classes of high production volume chemicals that are used widely in consumer and personal care products and have been associated with reproductive harm and pregnancy complications, such as preeclampsia and gestational diabetes. However, studies examining their influence on maternal blood pressure and gestational hypertension are limited. OBJECTIVES We investigated associations between individual phenols, parabens, and their mixture on maternal blood pressure measurements, including systolic and diastolic blood pressure (SBP and DBP) and hypertension during pregnancy (defined as stage 1 or 2 hypertension), among N = 1,433 Puerto Rico PROTECT study participants. METHODS We examined these relationships cross-sectionally at two time points during pregnancy (16-20 and 24-28 wks gestation) and longitudinally using linear mixed models (LMMs). Finally, we used quantile g-computation to examine the mixture effect on continuous (SBP, DBP) and binary (hypertension during pregnancy) blood pressure outcomes. RESULTS We observed a trend of higher odds of hypertension during pregnancy with exposure to multiple analytes and the overall mixture [including bisphenol A (BPA), bisphenol S (BPS), triclocarbon (TCC), triclosan (TCS), benzophenone-3 (BP-3), 2,4-dichlorophenol (2,4-DCP), 2,5-dichlorophenol (2,5-DCP), methyl paraben (M-PB), propyl paraben (P-PB), butyl paraben (B-PB), and ethyl paraben (E-PB)], especially at 24-28 wk gestation, with an adjusted mixture odds ratio ( OR ) = 1.57 (95% CI: 1.03, 2.38). Lower SBP and higher DBP were also associated with individual analytes, with results from LMMs most consistent for methyl paraben (M-PB) or propyl paraben (P-PB) and increased DBP across pregnancy [adjusted M-PB β = 0.78 (95% CI: 0.17, 1.38) and adjusted P-PB β = 0.85 (95% CI: 0.19, 1.51)] and for BPA, which was associated with decreased SBP (adjusted β = - 0.57 ; 95% CI: - 1.09 , - 0.05 ). Consistent with other literature, we also found evidence of effect modification by fetal sex, with a strong inverse association observed between the overall exposure mixture and SBP at visit 1 among participants carrying female fetuses only. CONCLUSIONS Our findings indicate that phenol and paraben exposure may collectively increase the risk of stage 1 or 2 hypertension during pregnancy, which has important implications for fetal and maternal health. https://doi.org/10.1289/EHP14008.
Collapse
Affiliation(s)
- Julia R. Varshavsky
- Department of Public Health and Health Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
- Department of Civil and Environmental Engineering, College of Engineering, Northeastern University, Boston, Massachusetts, USA
| | - John D. Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Emily Zimmerman
- Department of Communication Sciences and Disorders, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Megan L. Woodbury
- Department of Civil and Environmental Engineering, College of Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Max T. Aung
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zaira Y. Rosario-Pabon
- Department of Social Sciences, Graduate School of Public Health, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico, USA
| | - Amber L. Cathey
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Carmen M. Vélez-Vega
- Department of Social Sciences, Graduate School of Public Health, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico, USA
| | - José Cordero
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, Georgia, USA
| | - Akram Alshawabkeh
- Department of Civil and Environmental Engineering, College of Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Stephanie M. Eick
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Mittelberger J, Seefried M, Löb S, Kuhn C, Franitza M, Garrido F, Ditsch N, Jeschke U, Dannecker C. The expression of TIM-3 and Gal-9 on macrophages and Hofbauer cells in the placenta of preeclampsia patients. J Reprod Immunol 2024; 164:104296. [PMID: 38972269 DOI: 10.1016/j.jri.2024.104296] [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: 11/09/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Preeclampsia is a disorder of pregnancy characterized by endothelial dysfunction, abnormal placentation, systemic inflammation, and altered immune reaction. The aim of this study was to investigate the immune checkpoint molecules TIM-3 and Gal-9 on macrophages and Hofbauer cells (HBC) in the placenta of preeclampsia patients. Immunohistochemistry and Immunofluorescence was used to characterize the expression of the macrophage markers CD68 and CD163, CK7 and the proteins TIM-3 and Gal-9 in the placentas of preeclampsia patients comparing it to the placentas of healthy pregnancies. Double immunofluorescence staining (TIM-3 with CD3/CD19/CD56) was used to analyze the TIM-3 expression on other immune cells (T cells, B cells, NK cells) within the chorionic villi. The expression of TIM-3 on decidual macrophages did not significantly differ between the preeclamptic and the control group (p = 0.487). When looking at the different offspring we saw an upregulation of TIM-3 expression on decidual macrophages in preeclamptic placentas with female offspring (p = 0.049). On Hofbauer cells within the chorionic villi, the TIM-3 expression was significantly downregulated in preeclamptic cases without a sex-specific difference (p < 0.001). Looking at the protein Gal-9 the expression was proven to be downregulated both, on decidual macrophages (p = 0.003) and on Hofbauer cells (p = 0.002) within preeclamptic placentas compared to healthy controls. This was only significant in male offspring (p < 0.001 and p = 0.013) but not in female offspring (p = 0.360 and p = 0.068). While TIM-3 expression within the extravillious trophoblast and the syncytiotrophoblast was significantly downregulated (p < 0.001 and p = 0.012) in preeclampsia, the expression of Gal-9 was upregulated in (p < 0.001 and p < 0.001) compared to healthy controls. The local variations of the immune checkpoint molecules TIM-3 and Gal-9 in the placenta may contribute to the inflammation observed in preeclamptic patients. It could therefore contribute to the pathogenesis and be an important target in the treatment of preeclampsia.
Collapse
Affiliation(s)
- Johanna Mittelberger
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| | - Marina Seefried
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| | - Sanja Löb
- Department of Obstetrics and Gynecology, University Hospital, University of Wuerzburg, Josef-Schneider-Str. 4, Würzburg 97080, Germany
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| | - Manuela Franitza
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| | - Fabian Garrido
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| | - Nina Ditsch
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany.
| | - Christian Dannecker
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstraße 2, Augsburg 86156, Germany
| |
Collapse
|
29
|
Drago G, Aloi N, Ruggieri S, Longo A, Contrino ML, Contarino FM, Cibella F, Colombo P, Longo V. Guardians under Siege: Exploring Pollution's Effects on Human Immunity. Int J Mol Sci 2024; 25:7788. [PMID: 39063030 PMCID: PMC11277414 DOI: 10.3390/ijms25147788] [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: 06/20/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chemical pollution poses a significant threat to human health, with detrimental effects on various physiological systems, including the respiratory, cardiovascular, mental, and perinatal domains. While the impact of pollution on these systems has been extensively studied, the intricate relationship between chemical pollution and immunity remains a critical area of investigation. The focus of this study is to elucidate the relationship between chemical pollution and human immunity. To accomplish this task, this study presents a comprehensive review that encompasses in vitro, ex vivo, and in vivo studies, shedding light on the ways in which chemical pollution can modulate human immunity. Our aim is to unveil the complex mechanisms by which environmental contaminants compromise the delicate balance of the body's defense systems going beyond the well-established associations with defense systems and delving into the less-explored link between chemical exposure and various immune disorders, adding urgency to our understanding of the underlying mechanisms and their implications for public health.
Collapse
Affiliation(s)
- Gaspare Drago
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| | - Noemi Aloi
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| | - Silvia Ruggieri
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| | - Alessandra Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| | - Maria Lia Contrino
- Azienda Sanitaria Provinciale di Siracusa, Corso Gelone 17, 96100 Siracusa, Italy; (M.L.C.); (F.M.C.)
| | - Fabio Massimo Contarino
- Azienda Sanitaria Provinciale di Siracusa, Corso Gelone 17, 96100 Siracusa, Italy; (M.L.C.); (F.M.C.)
| | - Fabio Cibella
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| | - Valeria Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.D.); (N.A.); (S.R.); (A.L.); (F.C.); (V.L.)
| |
Collapse
|
30
|
Xing WY, Sun JN, Liu FH, Shan LS, Yin JL, Li YZ, Xu HL, Wei YF, Liu JX, Zheng WR, Zhang YY, Song XJ, Liu KX, Liu JC, Wang JY, Jia MQ, Chen X, Li XY, Liu C, Gong TT, Wu QJ. Per- and polyfluoroalkyl substances and human health outcomes: An umbrella review of systematic reviews with meta-analyses of observational studies. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134556. [PMID: 38735187 DOI: 10.1016/j.jhazmat.2024.134556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/27/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Although evidence on the association between per- and polyfluoroalkyl substances (PFASs) and human health outcomes has grown exponentially, specific health outcomes and their potential associations with PFASs have not been conclusively evaluated. METHODS We conducted a comprehensive search through the databases of PubMed, Embase, and Web of Science from inception to February 29, 2024, to identify systematic reviews with meta-analyses of observational studies examining the associations between the PFASs and multiple health outcomes. The quality of included studies was evaluated using the A Measurement Tool to Assess Systematic Reviews (AMSTAR) tool, and credibility of evidence was assessed using the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) criteria. The protocol of this umbrella review (UR) had been registered in PROSPERO (CRD 42023480817). RESULTS The UR identified 157 meta-analyses from 29 articles. Using the AMSTAR measurement tool, all articles were categorized as of moderate-to-high quality. Based on the GRADE assessment, significant associations between specific types of PFASs and low birth weight, tetanus vaccine response, and triglyceride levels showed high certainty of evidence. Moreover, moderate certainty of evidence with statistical significance was observed between PFASs and health outcomes including lower BMI z-score in infancy, poor sperm progressive motility, and decreased risk of preterm birth as well as preeclampsia. Fifty-two (33%) associations (e.g., PFASs and gestational hypertension, cardiovascular disease, etc) presented low certainty evidence. Additionally, eighty-five (55%) associations (e.g., PFASs with infertility, lipid metabolism, etc) presented very low certainty evidence. CONCLUSION High certainty of evidence supported that certain PFASs were associated with the incidence of low birth weight, low efficiency of the tetanus vaccine, and low triglyceride levels.
Collapse
Affiliation(s)
- Wei-Yi Xing
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Nan Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Department of Pediatric, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Li Yin
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Zi Li
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Li Xu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Fan Wei
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Xin Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wen-Rui Zheng
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying-Ying Zhang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin-Jian Song
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ke-Xin Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Cheng Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Yi Wang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming-Qian Jia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xing Chen
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Ying Li
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Chuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| |
Collapse
|
31
|
Puttaiah A, Kirthan JPA, Sadanandan DM, Somannavar MS. Inflammatory markers and their association with preeclampsia among pregnant women: A systematic review and meta-analysis. Clin Biochem 2024; 129:110778. [PMID: 38876455 DOI: 10.1016/j.clinbiochem.2024.110778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
The goal of this review was to investigate the levels of pro-inflammatory markers such as Tumour necrosis factor-α (TNF-α) Interlukin-6 (IL-6), C-reactive protein (CRP), Transforming growth factor-β1 (TGF-β1) and ferritin in pre-eclamptic and normotensive pregnant women. Using PubMed, ProQuest and Google Scholar databases, a literature search was carried out and case-control studies showing associations between inflammatory markers and preeclampsia in pregnancy published between 2010 and 2023 were included. The risk of bias was assessed by using the Newcastle Ottawa quality assessment scale. A random effect meta-analysis was performed and pooled difference in means with 95 % CI were reported. All statistical analyses were performed using R software. Out of 660 articles, 25 articles were included in the systematic review. The differences in means for TGF-β1, CRP, ferritin and TNF-α levels between the preeclamptic women and normotensive women were 2.37 pg/mL [95 % CI: -1.66,6.39], 5.62 mg/L [95 % CI: -4.11,15.36], 32.93 ng/mL [95 % CI: -7.66,58.19] and 13.67 pg/mL [95 % CI: 4.20,23.14] respectively which showed moderate increase. The pooled differences in means for hs-CRP and IL-6 levels between the preeclamptic and normotensive women were 3.20 mg/L [95 % CI: 0.27,6.12] and 17.64 pg/mL [95 % CI: -8.36,43.64] respectively which showed significant increase. Sub-group analysis showed significant differences for CRP, ferritin and TNF-α levels across ethnicities. Meta-analysis demonstrates an increase in the maternal circulating levels of inflammatory markers such as hs-CRP, IL-6 and showed moderate increase in TGF-β1, CRP, ferritin, TNF-α markers among women affected by preeclampsia compared to those with normotensive pregnancies.
Collapse
Affiliation(s)
- Ananda Puttaiah
- Department of Biochemistry, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belagavi, Karnataka 590010, India.
| | - J P Akshay Kirthan
- Department of Biochemistry, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belagavi, Karnataka 590010, India
| | - Deepthy Melepurakkal Sadanandan
- Women's & Children's Health Research Unit, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belagavi, Karnataka 590010, India
| | - Manjunath S Somannavar
- Department of Biochemistry, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belagavi, Karnataka 590010, India
| |
Collapse
|
32
|
Kim S, Shim S, Kwon J, Ryoo S, Byeon J, Hong J, Lee JH, Kwon YG, Kim JY, Kim YM. Alleviation of preeclampsia-like symptoms through PlGF and eNOS regulation by hypoxia- and NF-κB-responsive miR-214-3p deletion. Exp Mol Med 2024; 56:1388-1400. [PMID: 38825645 PMCID: PMC11263402 DOI: 10.1038/s12276-024-01237-8] [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: 10/07/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 06/04/2024] Open
Abstract
Preeclampsia is caused by placental hypoxia and systemic inflammation and is associated with reduced placental growth factor (PlGF) and endothelial nitric oxide synthase (eNOS) levels. The molecular signaling axes involved in this process may play a role in the pathogenesis of preeclampsia. Here, we found that hypoxic exposure increased hypoxia-inducible factor-1α (HIF-1α)/Twist1-mediated miR-214-3p biogenesis in trophoblasts, suppressing PlGF production and trophoblast invasion. TNF-α stimulation increased NF-κB-dependent miR-214-3p expression in endothelial cells, impairing eNOS expression and causing endothelial dysfunction. Synthetic miR-214-3p administration to pregnant mice decreased PlGF and eNOS expression, resulting in preeclampsia-like symptoms, including hypertension, proteinuria, and fetal growth restriction. Conversely, miR-214-3p deletion maintained the PlGF and eNOS levels in hypoxic pregnant mice, alleviating preeclampsia-like symptoms and signs. These findings provide new insights into the role of HIF-1/Twist1- and NF-κB-responsive miR-214-3p-dependent PlGF and eNOS downregulation in the pathogenesis of preeclampsia and establish miR-214-3p as a therapeutic or preventive target for preeclampsia and its complications.
Collapse
Affiliation(s)
- Suji Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jisoo Kwon
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea
| | - Sungwoo Ryoo
- Department of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Junyoung Byeon
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jungwoo Hong
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Young-Guen Kwon
- Advanced Institute of Technology, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Ji-Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea.
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
33
|
Rao A, Subedi R, Kundu I, Idicula-Thomas S, Shinde U, Bansal V, Balsarkar G, Mayadeo N, Das DK, Balasinor N, Madan T. Differential proteomics of circulating extracellular vesicles of placental origin isolated from women with early-onset preeclampsia reveal aberrant innate immune and hemostasis processes. Am J Reprod Immunol 2024; 91:e13860. [PMID: 38804582 DOI: 10.1111/aji.13860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
PROBLEM Early-onset preeclampsia (EOPE) is a severe gestational hypertensive disorder with significant feto-maternal morbidity and mortality due to uteroplacental insufficiency. Circulating extracellular vesicles of placental origin (EV-P) are known to be involved in the pathophysiology of EOPE and might serve as an ideal reservoir for its specific biomarkers. Therefore, we aimed to characterize and perform comparative proteomics of circulating EV-P from healthy pregnant and EOPE women before delivery. METHOD OF STUDY The EV-P from both groups were isolated using immunoaffinity and were characterized using transmission electron microscopy, dynamic light scattering, nanoparticle tracking analysis, and immunoblotting. Following IgG albumin depletion, the pooled proteins that were isolated from EV-P of both groups were subjected to quantitative TMT proteomics. RESULTS Circulating term EV-P isolated from both groups revealed ∼150 nm spherical vesicles containing CD9 and CD63 along with placental PLAP and HLA-G proteins. Additionally, the concentration of EOPE-derived EV-P was significantly increased. A total of 208 proteins were identified, with 26 among them being differentially abundant in EV-P of EOPE women. This study linked the pathophysiology of EOPE to 19 known and seven novel proteins associated with innate immune responses such as complement and TLR signaling along with hemostasis and oxygen homeostasis. CONCLUSION The theory suggesting circulating EVs of placental origin could mimic molecular information from the parent organ-"the placenta"-is strengthened by this study. The findings pave the way for possible discovery of novel prognostic and predictive biomarkers as well as provide insight into the mechanisms driving the pathogenesis of EOPE.
Collapse
Affiliation(s)
- Aishwarya Rao
- Innate Immunity Department, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Rambhadur Subedi
- Innate Immunity Department, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Indra Kundu
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Uma Shinde
- Neuroendocrinology Department, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Vandana Bansal
- Nowrosjee Wadia Maternity Hospital (NWMH), Mumbai, India
| | | | - Niranjan Mayadeo
- King Edward Memorial Hospital and Seth Gordhandas Sunderdas Medical College, Mumbai, India
| | - Dhanjit Kumar Das
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Nafisa Balasinor
- Neuroendocrinology Department, ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Taruna Madan
- Development Research, Indian Council of Medical Research, V. Ramalingaswami Bhawan, New Delhi, India
| |
Collapse
|
34
|
Wang Y, Bao J, Hua S, Yin L. Association between rs1799724 of TNF- α gene and early onset preeclampsia in Chinese: A pilot study. Eur J Obstet Gynecol Reprod Biol X 2024; 22:100303. [PMID: 39011057 PMCID: PMC11247142 DOI: 10.1016/j.eurox.2024.100303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/10/2024] [Accepted: 03/20/2024] [Indexed: 07/17/2024] Open
Abstract
Objective To investigate the association between polymorphisms of TNF- α (rs1799724, rs1800629), VEGF (rs3025039) and VEGFR1 (rs 722503) and early onset preeclampsia (EOPE) in Chinese. Methods A total of 132 EOPE patients from January 2016 to December 2018 at the Second Hospital of Tianjin Medical University were selected as the EOPE group, and 156 normal pregnant patients as the Control group. In both groups, 5 ml of peripheral venous blood was obtained after admission. The characteristics of genotype and allele distribution at the four SNPs in the study subjects were examined by matrix-assisted laser desorption ionization time-of-flight mass spectrometric genotyping. Results The genotype frequency distribution and allele frequency distribution of rs1799724 were significantly different between the EOPE group and the Control group (P = 0.002,P = 0.003). The T allele was statistically associated with the development of EOPE under a dominant genetic model (P = 0.001). The genotype and allele frequency distributions of rs1800629, rs3025039, and rs 722503 did not differ significantly between the EOPE group and the Control group (P > 0.05). There was no linkage disequilibrium among rs1799724, rs1800629 and rs3025039 loci, the corresponding haploid cannot be formed. Conclusions The rs1799724 of TNF- α gene is a genetic susceptibility locus for EOPE and may be a potential predictors of preeclampsia.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jianheng Bao
- Second Department of Hepatopancreatobiliary Surgery, Tianjin Nankai Hospital, Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin, China
| | - Shaofang Hua
- Department of Obstetrics and Gynecology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Lirong Yin
- Department of Obstetrics and Gynecology, the Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
35
|
Hu H, Ma J, Peng Y, Feng R, Luo C, Zhang M, Tao Z, Chen L, Zhang T, Chen W, Yin Q, Zhai J, Chen J, Yin A, Wang CC, Zhong M. Thrombospondin-1 Regulates Trophoblast Necroptosis via NEDD4-Mediated Ubiquitination of TAK1 in Preeclampsia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309002. [PMID: 38569496 PMCID: PMC11151050 DOI: 10.1002/advs.202309002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/05/2024] [Indexed: 04/05/2024]
Abstract
Preeclampsia (PE) is considered as a disease of placental origin. However, the specific mechanism of placental abnormalities remains elusive. This study identified thrombospondin-1 (THBS1) is downregulated in preeclamptic placentae and negatively correlated with blood pressure. Functional studies show that THBS1 knockdown inhibits proliferation, migration, and invasion and increases the cycle arrest and apoptosis rate of HTR8/SVneo cells. Importantly, THBS1 silencing induces necroptosis in HTR8/SVneo cells, accompanied by the release of damage-associated molecular patterns (DAMPs). Necroptosis inhibitors necrostatin-1 and GSK'872 restore the trophoblast survival while pan-caspase inhibitor Z-VAD-FMK has no effect. Mechanistically, the results show that THBS1 interacts with transforming growth factor B-activated kinase 1 (TAK1), which is a central modulator of necroptosis quiescence and affects its stability. Moreover, THBS1 silencing up-regulates the expression of neuronal precursor cell-expressed developmentally down-regulated 4 (NEDD4), which acts as an E3 ligase of TAK1 and catalyzes K48-linked ubiquitination of TAK1 in HTR8/SVneo cells. Besides, THBS1 attenuates PE phenotypes and improves the placental necroptosis in vivo. Taken together, the down-regulation of THBS1 destabilizes TAK1 by activating NEDD4-mediated, K48-linked TAK1 ubiquitination and promotes necroptosis and DAMPs release in trophoblast cells, thus participating in the pathogenesis of PE.
Collapse
Affiliation(s)
- Haoyue Hu
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangzhou Key Laboratory of Forensic Multi‐Omics for Precision IdentificationSchool of Forensic MedicineSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jing Ma
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangzhou Key Laboratory of Forensic Multi‐Omics for Precision IdentificationSchool of Forensic MedicineSouthern Medical UniversityGuangzhouGuangdong510515China
| | - You Peng
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangzhou Key Laboratory of Forensic Multi‐Omics for Precision IdentificationSchool of Forensic MedicineSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Rixuan Feng
- School of NursingSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chenling Luo
- School of NursingSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Minyi Zhang
- Department of EpidemiologySchool of Public HealthSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zixin Tao
- Department of Obstetrics and GynecologyGuangzhou First People's HospitalSchool of MedicineSouth China University of TechnologyGuangzhouGuangdong510180China
| | - Lu Chen
- Department of Obstetrics and Gynaecology;Li Ka Shing Institute of Health Sciences;School of Biomedical Sciences;Chinese University of Hong Kong‐Sichuan University Joint Laboratory in Reproductive Medicine; The Chinese University of Hong KongHong Kong SARNTChina
| | - Tao Zhang
- Department of Obstetrics and Gynaecology;Li Ka Shing Institute of Health Sciences;School of Biomedical Sciences;Chinese University of Hong Kong‐Sichuan University Joint Laboratory in Reproductive Medicine; The Chinese University of Hong KongHong Kong SARNTChina
| | - Wenqian Chen
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangzhou Key Laboratory of Forensic Multi‐Omics for Precision IdentificationSchool of Forensic MedicineSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Qian Yin
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jinguo Zhai
- School of NursingSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jun Chen
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Ailan Yin
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology;Li Ka Shing Institute of Health Sciences;School of Biomedical Sciences;Chinese University of Hong Kong‐Sichuan University Joint Laboratory in Reproductive Medicine; The Chinese University of Hong KongHong Kong SARNTChina
| | - Mei Zhong
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
36
|
Shaw LJ, Patel K, Lala-Trindade A, Feltovich H, Vieira L, Kontorovich A, Ananth C, Taqueti VR, Mitrani L, Stern T, DeBolt C, Kase N, Smith RT, Narula J, Mehran R, Bianco A, Bhatt DL, Stone JL. Pathophysiology of Preeclampsia-Induced Vascular Dysfunction and Implications for Subclinical Myocardial Damage and Heart Failure. JACC. ADVANCES 2024; 3:100980. [PMID: 38938863 PMCID: PMC11198310 DOI: 10.1016/j.jacadv.2024.100980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/04/2024] [Accepted: 02/28/2024] [Indexed: 06/29/2024]
Abstract
Tragically, preeclampsia is a leading cause of pregnancy-related complications and is linked to a heightened risk for morbid and fatal cardiovascular disease (CVD) outcomes. Although the mechanism connecting preeclampsia to CVD risk has yet to be fully elucidated, evidence suggests distinct pathways of early and late preeclampsia with shared CV risk factors but with profound differences in perinatal and postpartum risk to the mother and infant. In early preeclampsia, <34 weeks of gestation, systemic vascular dysfunction contributes to near-term subclinical myocardial damage. Hypertrophy and diastolic abnormalities persist postpartum and contribute to early onset heart failure (HF). This HF risk remains elevated decades later and contributes to premature death. Black women are at the highest risk of preeclampsia and HF. These findings support closer monitoring of women postpartum, especially for those with early and severe preeclampsia to control chronic hypertension and reduce the potentially preventable sequelae of heightened CVD and HF risk.
Collapse
Affiliation(s)
- Leslee J. Shaw
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Women’s Heart and Vascular Center at Mount Sinai Heart, New York, New York, USA
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
- Department of Population Health Science and Policy, New York, New York, USA
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Krishna Patel
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Women’s Heart and Vascular Center at Mount Sinai Heart, New York, New York, USA
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
- Department of Population Health Science and Policy, New York, New York, USA
| | - Anuradha Lala-Trindade
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Helen Feltovich
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Luciana Vieira
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Amy Kontorovich
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Cande Ananth
- Division of Epidemiology and Biostatistics, Department of Obstetrics, Gynecology, and Reproductive Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Viviany R. Taqueti
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lindsey Mitrani
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Toni Stern
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Chelsea DeBolt
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Nathan Kase
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - R. Theodore Smith
- Department of Opthamology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jagat Narula
- UT Health Houston, McGovern Medical School, Houston, Texas, USA
| | - Roxana Mehran
- Women’s Heart and Vascular Center at Mount Sinai Heart, New York, New York, USA
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Angela Bianco
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Deepak L. Bhatt
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Joanne L. Stone
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| |
Collapse
|
37
|
Liu Y, Zhang Y, Du L, Chen D. The genetic relationships between immune cell traits, circulating inflammatory proteins and preeclampsia/eclampsia. Front Immunol 2024; 15:1389843. [PMID: 38873604 PMCID: PMC11170637 DOI: 10.3389/fimmu.2024.1389843] [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: 02/26/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Objectives Preeclampsia/eclampsia (PE), a critical complication during pregnancy, has been suggested to correlate with immune cell phenotypes and levels of circulating inflammatory proteins. Our study aimed to employ a two-sample mendelian randomization (MR) analysis to assess the potential causal effects of immune cell phenotypes and circulating inflammatory proteins on the onset of PE. Methods We utilized summary-level data from genome-wide association studies (GWAS). This included statistics for 371 immune cell phenotypes from 3,757 individuals in the Sardinian founder population, and data on 91 circulating inflammatory proteins from 14,824 European ancestry participants. Additionally, genetic associations related to PE were extracted from the FinnGen consortium, involving 1,413 cases and 287,137 controls. We applied inverse variance weighting (IVW) and supplementary methods like MR-Egger, weighted median, and weighted mode to comprehensively assess potential causal links. Results Our analysis revealed significant causal associations of several immune cells type and inflammatory proteins with PE. Out of the immune cell phenotypes analyzed, six immune phenotypes emerged as significant risk factors (p <0.01), mainly include CD4 on activated and secreting CD4 regulatory T cells, CD28 on CD39+ CD4+ T cells, CD127- CD8+ T cell absolute cell (AC) counts, HLA DR on HLA DR+ CD8+ T cell, CD66b on CD66b++ myeloid cells, and HLA DR on dendritic cells. And ten were identified as protective factors (p <0.01). Such as CD45 on CD33br HLA DR+ CD14-, CD33+ HLA DR+ AC, CD33+ HLA DR+ CD14- AC, CD33+ HLA DR+ CD14dim AC, CD27 on CD24+ CD27+ B cell, CD20- CD38- %B cell, IgD- CD24- %B cell CD80 on plasmacytoid DC, CD25 on CD4+ T cell, and CD25 on activated & secreting CD4 regulatory T cell. Furthermore, among the inflammatory proteins studied, five showed a significant association with PE, with three offering protective effects mainly include that C-X-C motif chemokine 1, tumor necrosis factor ligand superfamily member 14, and C-C motif chemokine 19 and two exacerbating PE risk such as STAM-binding domain and Interleukin-6 (p <0.05). Conclusions Our study highlights the pivotal roles played by diverse immune cell phenotypes and circulating inflammatory proteins in the pathophysiology of PE. These findings illuminate the underlying genetic mechanisms, emphasizing the criticality of immune regulation during pregnancy. Such insights could pave the way for novel intervention strategies in managing PE, potentially enhancing maternal and neonatal health outcomes.
Collapse
Affiliation(s)
- Yu Liu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
- Guangdong-Hong Kong Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
| | - Yuliang Zhang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
- Guangdong-Hong Kong Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
| | - Lili Du
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
- Guangdong-Hong Kong Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
- Guangdong-Hong Kong Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Talebi S, Mehrabani S, Ghoreishy SM, Wong A, Moghaddam A, Feyli PR, Amirian P, Zarpoosh M, Kermani MAH, Moradi S. The association between ultra-processed food and common pregnancy adverse outcomes: a dose-response systematic review and meta-analysis. BMC Pregnancy Childbirth 2024; 24:369. [PMID: 38750456 PMCID: PMC11097443 DOI: 10.1186/s12884-024-06489-w] [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: 01/18/2024] [Accepted: 04/07/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVES Given the increasing incidence of negative outcomes during pregnancy, our research team conducted a dose-response systematic review and meta-analysis to investigate the relationship between ultra-processed foods (UPFs) consumption and common adverse pregnancy outcomes including gestational diabetes mellitus (GDM), preeclampsia (PE), preterm birth (PTB), low birth weight (LBW), and small for gestational age (SGA) infants. UPFs are described as formulations of food substances often modified by chemical processes and then assembled into ready-to-consume hyper-palatable food and drink products using flavors, colors, emulsifiers, and other cosmetic additives. Examples include savory snacks, reconstituted meat products, frozen meals that have already been made, and soft drinks. METHODS A comprehensive search was performed using the Scopus, PubMed, and Web of Science databases up to December 2023. We pooled relative risk (RR) and 95% confidence intervals (CI) using a random-effects model. RESULTS Our analysis (encompassing 54 studies with 552,686 individuals) revealed a significant association between UPFs intake and increased risks of GDM (RR = 1.19; 95% CI: 1.10, 1.27; I2 = 77.5%; p < 0.001; studies = 44; number of participants = 180,824), PE (RR = 1.28; 95% CI: 1.03, 1.59; I2 = 80.0%; p = 0.025; studies = 12; number of participants = 54,955), while no significant relationships were found for PTB, LBW and SGA infants. Importantly, a 100 g increment in UPFs intake was related to a 27% increase in GDM risk (RR = 1.27; 95% CI: 1.07, 1.51; I2 = 81.0%; p = 0.007; studies = 9; number of participants = 39,812). The non-linear dose-response analysis further indicated a positive, non-linear relationship between UPFs intake and GDM risk Pnonlinearity = 0.034, Pdose-response = 0.034), although no such relationship was observed for PE (Pnonlinearity = 0.696, Pdose-response = 0.812). CONCLUSION In summary, both prior to and during pregnancy, chronic and excessive intake of UPFs is associated with an increased risk of GDM and PE. However, further observational studies, particularly among diverse ethnic groups with precise UPFs consumption measurement tools, are imperative for a more comprehensive understanding.
Collapse
Affiliation(s)
- Sepide Talebi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanaz Mehrabani
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Mojtaba Ghoreishy
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
- Student research committee, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Alexei Wong
- Department of Health and Human Performance, Marymount University, Arlington, VA, USA
| | - Aliasghar Moghaddam
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Peyman Rahimi Feyli
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Parsa Amirian
- General Practitioner, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Mahsa Zarpoosh
- General Practitioner, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Mohammad Ali Hojjati Kermani
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Moradi
- Department of Nutrition and Food Sciences, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
39
|
Alemayehu E, Mohammed O, Belete MA, Mulatie Z, Debash H, Gedefie A, Weldehanna DG, Eshetu B, Shibabaw A, Tekele SG, Tilahun M, Ebrahim H. Association of prothrombin time, thrombin time and activated partial thromboplastin time levels with preeclampsia: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2024; 24:354. [PMID: 38741046 DOI: 10.1186/s12884-024-06543-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Preeclampsia (PE), an obstetric disorder, remains one of the leading causes of maternal and infant mortality worldwide. In individuals with PE, the coagulation-fibrinolytic system is believed to be among the most significantly impacted systems due to maternal inflammatory responses and immune dysfunction. Therefore, this systematic review and meta-analysis aimed to assess the association of prothrombin time (PT), thrombin time (TT) and activated partial thromboplastin time (APTT) levels with preeclampsia. METHODS This systematic review and meta-analysis was conducted in accordance with the PRISMA guidelines. Articles relevant to the study, published from July 26, 2013, to July 26, 2023, were systematically searched across various databases including PubMed, Scopus, Embase, and Hinari. The methodological quality of the articles was evaluated using the Joanna Briggs Institute critical appraisal checklist. Utilizing Stata version 14.0, a random-effects model was employed to estimate the pooled standardized mean difference (SMD) along with the respective 95% CIs. The I2 statistics and Cochrane Q test were utilized to assess heterogeneity, while subgroup analyses were performed to explore its sources. Furthermore, Egger's regression test and funnel plot were employed to assess publication bias among the included studies. RESULTS A total of 30 articles, involving 5,964 individuals (2,883 with PE and 3,081 as normotensive pregnant mothers), were included in this study. The overall pooled SMD for PT, APTT, and TT between PE and normotensive pregnant mothers were 0.97 (95% CI: 0.65-1.29, p < 0.001), 1.05 (95% CI: 0.74-1.36, p < 0.001), and 0.30 (95% CI: -0.08-0.69, p = 0.11), respectively. The pooled SMD indicates a significant increase in PT and APTT levels among PE patients compared to normotensive pregnant mothers, while the increase in TT levels among PE patients was not statistically significant. CONCLUSIONS The meta-analysis underscores the association between PE and prolonged PT and APTT. This suggests that evaluating coagulation parameters like PT, APTT, and TT in pregnant women could offer easily accessible and cost-effective clinical indicators for assessing PE. However, multicenter longitudinal studies are needed to evaluate their effectiveness across various gestational weeks of pregnancy.
Collapse
Affiliation(s)
- Ermiyas Alemayehu
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia.
| | - Ousman Mohammed
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Melaku Ashagrie Belete
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Zewudu Mulatie
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Habtu Debash
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Alemu Gedefie
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Daniel Gebretsadik Weldehanna
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Bruktawit Eshetu
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Agumas Shibabaw
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Saba Gebremichael Tekele
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Mihret Tilahun
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Hussen Ebrahim
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| |
Collapse
|
40
|
Mao Y, Li X, Ren R, Yuan Y, Wang L, Zhang X. Identification of hub glutamine metabolism-associated genes and immune characteristics in pre-eclampsia. PLoS One 2024; 19:e0303471. [PMID: 38718074 PMCID: PMC11078374 DOI: 10.1371/journal.pone.0303471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVE Preeclampsia (PE) is a severe complication of unclear pathogenesis associated with pregnancy. This research aimed to elucidate the properties of immune cell infiltration and potential biomarkers of PE based on bioinformatics analysis. MATERIALS AND METHODS Two PE datasets were imported from the Gene ExpressioOmnibus (GEO) and screened to identify differentially expressed genes (DEGs). Significant module genes were identified by weighted gene co-expression network analysis (WGCNA). DEGs that interacted with key module genes (GLu-DEGs) were analyzed further by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. The diagnostic value of the genes was assessed using receiver operating characteristic (ROC) curves and protein-protein interaction (PPI) networks were constructed using GeneMANIA, and GSVA analysis was performed using the MSigDB database. Immune cell infiltration was analyzed using the TISIDB database, and StarBase and Cytoscape were used to construct an RBP-mRNA network. The identified hub genes were validated in two independent datasets. For further confirmation, placental tissue from healthy pregnant women and women with PE were collected and analyzed using both RT-qPCR and immunohistochemistry. RESULTS A total of seven GLu-DEGs were obtained and were found to be involved in pathways associated with the transport of sulfur compounds, PPAR signaling, and energy metabolism, shown by GO and KEGG analyses. GSVA indicated significant increases in adipocytokine signaling. Furthermore, single-sample Gene Set Enrichment Analysis (ssGSEA) indicated that the levels of activated B cells and T follicular helper cells were significantly increased in the PE group and were negatively correlated with GLu-DEGs, suggesting their potential importance. CONCLUSION In summary, the results showed a correlation between glutamine metabolism and immune cells, providing new insights into the understandingPE pathogenesis and furnishing evidence for future advances in the treatment of this disease.
Collapse
Affiliation(s)
- Yan Mao
- First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of Gynecology and Obstetrics, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xinye Li
- Department of Gynecology and Obstetrics, General Hospital of Lanzhou Petrochemical Corporation, Lanzhou, Gansu, China
| | - Rui Ren
- First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of Gynecology and Obstetrics, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yue Yuan
- First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Li Wang
- Department of Gynecology and Obstetrics, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xuehong Zhang
- Gansu Key Laboratory for Reproductive Medicine and Embryology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
41
|
Zahid S, Mohamed MS, Rajendran A, Minhas AS, Khan MZ, Nazir NT, Ocon AJ, Weber BN, Isiadinso I, Michos ED. Rheumatoid arthritis and cardiovascular complications during delivery: a United States inpatient analysis. Eur Heart J 2024; 45:1524-1536. [PMID: 38427130 DOI: 10.1093/eurheartj/ehae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND AND AIMS Persons with rheumatoid arthritis (RA) have an increased risk of obstetric-associated complications, as well as long-term cardiovascular (CV) risk. Hence, the aim was to evaluate the association of RA with acute CV complications during delivery admissions. METHODS Data from the National Inpatient Sample (2004-2019) were queried utilizing ICD-9 or ICD-10 codes to identify delivery hospitalizations and a diagnosis of RA. RESULTS A total of 12 789 722 delivery hospitalizations were identified, of which 0.1% were among persons with RA (n = 11 979). Individuals with RA, vs. those without, were older (median 31 vs. 28 years, P < .01) and had a higher prevalence of chronic hypertension, chronic diabetes, gestational diabetes mellitus, obesity, and dyslipidaemia (P < .01). After adjustment for age, race/ethnicity, comorbidities, insurance, and income, RA remained an independent risk factor for peripartum CV complications including preeclampsia [adjusted odds ratio (aOR) 1.37 (95% confidence interval 1.27-1.47)], peripartum cardiomyopathy [aOR 2.10 (1.11-3.99)], and arrhythmias [aOR 2.00 (1.68-2.38)] compared with no RA. Likewise, the risk of acute kidney injury and venous thromboembolism was higher with RA. An overall increasing trend of obesity, gestational diabetes mellitus, and acute CV complications was also observed among individuals with RA from 2004-2019. For resource utilization, length of stay and cost of hospitalization were higher for deliveries among persons with RA. CONCLUSIONS Pregnant persons with RA had higher risk of preeclampsia, peripartum cardiomyopathy, arrhythmias, acute kidney injury, and venous thromboembolism during delivery hospitalizations. Furthermore, cardiometabolic risk factors among pregnant individuals with RA rose over this 15-year period.
Collapse
Affiliation(s)
- Salman Zahid
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Mohamed S Mohamed
- Sands-Constellation Heart Institute, Rochester General Hospital, Rochester, NY, USA
| | - Aardra Rajendran
- Division of Cardiology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Anum S Minhas
- Division of Cardiology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Muhammad Zia Khan
- Heart and Vascular Institute, West Virginia University, Morgantown, WV, USA
| | - Noreen T Nazir
- Division of Cardiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Anthony J Ocon
- Division of Allergy, Immunology & Rheumatology, Rochester Regional Health, Rochester, NY, USA
| | - Brittany N Weber
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ijeoma Isiadinso
- Division of Cardiology, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| |
Collapse
|
42
|
Gumusoglu SB, Kiel MD, Gugel A, Schickling BM, Weaver KR, Lauffer MC, Sullivan HR, Coulter KJ, Blaine BM, Kamal M, Zhang Y, Devor EJ, Santillan DA, Gantz SC, Santillan MK. Anti-angiogenic mechanisms and serotonergic dysfunction in the Rgs2 knockout model for the study of psycho-obstetric risk. Neuropsychopharmacology 2024; 49:864-875. [PMID: 37848733 PMCID: PMC10948883 DOI: 10.1038/s41386-023-01749-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023]
Abstract
Psychiatric and obstetric diseases are growing threats to public health and share high rates of co-morbidity. G protein-coupled receptor signaling (e.g., vasopressin, serotonin) may be a convergent psycho-obstetric risk mechanism. Regulator of G Protein Signaling 2 (RGS2) mutations increase risk for both the gestational disease preeclampsia and for depression. We previously found preeclampsia-like, anti-angiogenic obstetric phenotypes with reduced placental Rgs2 expression in mice. Here, we extend this to test whether conserved cerebrovascular and serotonergic mechanisms are also associated with risk for neurobiological phenotypes in the Rgs2 KO mouse. Rgs2 KO exhibited anxiety-, depression-, and hedonic-like behaviors. Cortical vascular density and vessel length decreased in Rgs2 KO; cortical and white matter thickness and cell densities were unchanged. In Rgs2 KO, serotonergic gene expression was sex-specifically changed (e.g., cortical Htr2a, Maoa increased in females but all serotonin targets unchanged or decreased in males); redox-related expression increased in paraventricular nucleus and aorta; and angiogenic gene expression was changed in male but not female cortex. Whole-cell recordings from dorsal raphe serotonin neurons revealed altered 5-HT1A receptor-dependent inhibitory postsynaptic currents (5-HT1A-IPSCs) in female but not male KO neurons. Additionally, serotonin transporter blockade by the SSRI sertraline increased the amplitude and time-to-peak of 5-HT1A-IPSCs in KO neurons to a greater extent than in WT neurons in females only. These results demonstrate behavioral, cerebrovascular, and sertraline hypersensitivity phenotypes in Rgs2 KOs, some of which are sex-specific. Disruptions may be driven by vascular and cell stress mechanisms linking the shared pathogenesis of psychiatric and obstetric disease to reveal future targets.
Collapse
Affiliation(s)
- Serena B Gumusoglu
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
| | - Michaela D Kiel
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Aleigha Gugel
- Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Brandon M Schickling
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Kaylee R Weaver
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Marisol C Lauffer
- Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
- Neural Circuits and Behavior Core, Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
| | - Hannah R Sullivan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Kaylie J Coulter
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Brianna M Blaine
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Mushroor Kamal
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Yuping Zhang
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Stephanie C Gantz
- Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Mark K Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, USA.
| |
Collapse
|
43
|
Tan L, Kluivers AC, Cruz-López EO, Broekhuizen M, Chen Z, Neuman RI, Schoenmakers S, Ruijgrok L, van de Velde D, de Winter BC, van den Bogaerdt AJ, Lu X, Danser AJ, Verdonk K. Statins Prevent the Deleterious Consequences of Placental Chemerin Upregulation in Preeclampsia. Hypertension 2024; 81:861-875. [PMID: 38361240 PMCID: PMC10956680 DOI: 10.1161/hypertensionaha.123.22457] [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: 11/24/2023] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Chemerin, an inflammatory adipokine, is upregulated in preeclampsia, and its placental overexpression results in preeclampsia-like symptoms in mice. Statins may lower chemerin. METHODS Chemerin was determined in a prospective cohort study in women suspected of preeclampsia and evaluated as a predictor versus the sFlt-1 (soluble fms-like tyrosine kinase-1)/PlGF (placental growth factor) ratio. Chemerin release was studied in perfused placentas and placental explants with or without the statins pravastatin and fluvastatin. We also addressed statin placental passage and the effects of chemerin in chorionic plate arteries. RESULTS Serum chemerin was elevated in women with preeclampsia, and its addition to a predictive model yielded significant effects on top of the sFlt-1/PlGF ratio to predict preeclampsia and its fetal complications. Perfused placentas and explants of preeclamptic women released more chemerin and sFlt-1 and less PlGF than those of healthy pregnant women. Statins reversed this. Both statins entered the fetal compartment, and the fetal/maternal concentration ratio of pravastatin was twice that of fluvastatin. Chemerin constricted plate arteries, and this was blocked by a chemerin receptor antagonist and pravastatin. Chemerin did not potentiate endothelin-1 in chorionic plate arteries. In explants, statins upregulated low-density lipoprotein receptor expression, which relies on the same transcription factor as chemerin, and NO release. CONCLUSIONS Chemerin is a biomarker for preeclampsia, and statins both prevent its placental upregulation and effects, in an NO and low-density lipoprotein receptor-dependent manner. Combined with their capacity to improve the sFlt-1/PlGF ratio, this offers an attractive mechanism by which statins may prevent or treat preeclampsia.
Collapse
Affiliation(s)
- Lunbo Tan
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, China (L.T., X.L.)
| | - Ans C.M. Kluivers
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Department of Obstetrics and Gynecology (A.C.M.K., R.I.N., S.S.), Erasmus MC, Rotterdam, the Netherlands
| | - Edwyn O. Cruz-López
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
| | - Michelle Broekhuizen
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care (M.B.), Erasmus MC, Rotterdam, the Netherlands
| | - Zhongli Chen
- Department of Internal Medicine, Academic Center for Thyroid Diseases (Z.C.), Erasmus MC, Rotterdam, the Netherlands
| | - Rugina I. Neuman
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Department of Obstetrics and Gynecology (A.C.M.K., R.I.N., S.S.), Erasmus MC, Rotterdam, the Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and Gynecology (A.C.M.K., R.I.N., S.S.), Erasmus MC, Rotterdam, the Netherlands
| | - Liesbeth Ruijgrok
- Department of Hospital Pharmacy (L.R., D.v.d.V., B.C.M.d.W.), Erasmus MC, Rotterdam, the Netherlands
| | - Daan van de Velde
- Department of Hospital Pharmacy (L.R., D.v.d.V., B.C.M.d.W.), Erasmus MC, Rotterdam, the Netherlands
| | - Brenda C.M. de Winter
- Department of Hospital Pharmacy (L.R., D.v.d.V., B.C.M.d.W.), Erasmus MC, Rotterdam, the Netherlands
| | - Antoon J. van den Bogaerdt
- Heart Valve Department, Euro Tissue Bank-Bio Implant Services LIFE (ETB-BISLIFE), Beverwijk, the Netherlands (A.J.v.d.B.)
| | - Xifeng Lu
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, China (L.T., X.L.)
| | - A.H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
| | - Koen Verdonk
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
44
|
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
|
45
|
Lin Z, Wu S, Jiang Y, Chen Z, Huang X, Wen Z, Yuan Y. Unraveling the molecular mechanisms driving enhanced invasion capability of extravillous trophoblast cells: a comprehensive review. J Assist Reprod Genet 2024; 41:591-608. [PMID: 38315418 PMCID: PMC10957806 DOI: 10.1007/s10815-024-03036-6] [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: 11/28/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Precise extravillous trophoblast (EVT) invasion is crucial for successful placentation and pregnancy. This review focuses on elucidating the mechanisms that promote heightened EVT invasion. We comprehensively summarize the pivotal roles of hormones, angiogenesis, hypoxia, stress, the extracellular matrix microenvironment, epithelial-to-mesenchymal transition (EMT), immunity, inflammation, programmed cell death, epigenetic modifications, and microbiota in facilitating EVT invasion. The molecular mechanisms underlying enhanced EVT invasion may provide valuable insights into potential pathogenic mechanisms associated with diseases characterized by excessive invasion, such as the placenta accreta spectrum (PAS), thereby offering novel perspectives for managing pregnancy complications related to deficient EVT invasion.
Collapse
Affiliation(s)
- Zihan Lin
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Shuang Wu
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Yinghui Jiang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Ziqi Chen
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Xiaoye Huang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Zhuofeng Wen
- The Sixth Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Yi Yuan
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
46
|
Song R, Baker TL, Watters JJ, Kumar S. Obstructive Sleep Apnea-Associated Intermittent Hypoxia-Induced Immune Responses in Males, Pregnancies, and Offspring. Int J Mol Sci 2024; 25:1852. [PMID: 38339130 PMCID: PMC10856042 DOI: 10.3390/ijms25031852] [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: 12/27/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Obstructive sleep apnea (OSA), a respiratory sleep disorder associated with cardiovascular diseases, is more prevalent in men. However, OSA occurrence in pregnant women rises to a level comparable to men during late gestation, creating persistent effects on both maternal and offspring health. The exact mechanisms behind OSA-induced cardiovascular diseases remain unclear, but inflammation and oxidative stress play a key role. Animal models using intermittent hypoxia (IH), a hallmark of OSA, reveal several pro-inflammatory signaling pathways at play in males, such as TLR4/MyD88/NF-κB/MAPK, miRNA/NLRP3, and COX signaling, along with shifts in immune cell populations and function. Limited evidence suggests similarities in pregnancies and offspring. In addition, suppressing these inflammatory molecules ameliorates IH-induced inflammation and tissue injury, providing new potential targets to treat OSA-associated cardiovascular diseases. This review will focus on the inflammatory mechanisms linking IH to cardiovascular dysfunction in males, pregnancies, and their offspring. The goal is to inspire further investigations into the understudied populations of pregnant females and their offspring, which ultimately uncover underlying mechanisms and therapeutic interventions for OSA-associated diseases.
Collapse
Affiliation(s)
- Ruolin Song
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Tracy L. Baker
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Jyoti J. Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
47
|
Valiati N, Puel EM, Stefani CM, Lataro RM. Does probiotic ingestion reduce the risk of preeclampsia? A systematic review. Appl Physiol Nutr Metab 2024; 49:135-147. [PMID: 37844331 DOI: 10.1139/apnm-2023-0089] [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] [Indexed: 10/18/2023]
Abstract
We aimed to systematically review the literature on the effects of probiotic consumption on the risk of preeclampsia (PE) development. Eight databases, clinical trial registries, and grey literature were searched until February 2022. Studies were included if they (1) were randomized clinical trials (RCTs), (2) included pregnant women aged ≥ 18 years old, (3) used probiotics products, and (4) were written in the Latin alphabet. A random-effects meta-analysis was performed using the risk ratio as the effect measure with 95% confidence intervals (CI) for PE. The search strategy identified 359 records, from which six RCTs were included. The six RCTs evaluated pregnant women with comorbidities and enrolled 593 women that received probiotics and 625 receiving placebo. None of the included RCTs analyzed healthy women. Probiotics increased by 12% the PE risk (RR 1.12, 95% CI, CI = 0.83-1.53, p = 0.46, χ2 = 3.31, df = 5 (p = 0.65), I2 = 0%). The certainty of the evidence, evaluated through the Grading of Recommendations Assessment, Development and Evaluation approach, was rated as very low. In conclusion, probiotics supplementation may slightly increase PE rates in pregnant women with comorbidities. The risk may be higher in obese women and for periods of ingestion longer than eight weeks. However, the evidence certainty is very low. PROSPERO registration No.CRD42021278611.
Collapse
Affiliation(s)
- Nayara Valiati
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Esthela M Puel
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cristine M Stefani
- Department of DentistryFaculty of Health Sciences, University of Brasília, Brasília, Brazil
| | - Renata M Lataro
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
48
|
Zhou W, Li X, Li X, Liu Y, Song W, Yang Q. The role of circular RNA in preeclampsia: From pathophysiological mechanism to clinical application. Life Sci 2024; 338:122407. [PMID: 38184270 DOI: 10.1016/j.lfs.2023.122407] [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: 11/05/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
Preeclampsia (PE) is a common pregnancy-induced hypertension disorder that poses a significant threat to the health of pregnant women and fetuses, and has become a leading cause of maternal, fetal, and neonatal mortality. Currently, the therapy strategy for PE is mainly prevention management and symptomatic treatment, and only delivery can completely terminate PE. Therefore, a deeper understanding of the pathogenesis of PE is needed to make treatment and prevention more effective and targeted. With the deepening of molecular etiology research, circular RNAs (circRNAs) have been found to be widely involved in various processes of PE pathogenesis. As a kind of RNA with a special "head to tail" loop structure, the characteristics of circRNAs enable them to play diverse roles in the pathophysiology of PE, and can also serve as ideal biomarkers for early prediction and monitoring progression of PE. In this review, we summarized the latest research on PE-related circRNAs, trying to elucidate the unique or shared roles of circRNAs in various pathophysiological mechanisms of PE, aiming to provide a whole picture of current research on PE-related circRNAs, and extend a new perspective for the precise screening and targeted therapy of PE.
Collapse
Affiliation(s)
- Wenjing Zhou
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China; Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiuying Li
- Medical Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| | - Xin Li
- Medical College, Jilin Engineering Vocational College, Siping, Jilin, China.
| | - Yaojia Liu
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China.
| | - Wenling Song
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Qiwei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
49
|
Rowe CW, Rosee P, Sathiakumar A, Ramesh S, Qiao V, Huynh J, Dennien G, Weaver N, Wynne K. Factors associated with maternal hyperglycaemia and neonatal hypoglycaemia after antenatal betamethasone administration in women with diabetes in pregnancy. Diabet Med 2024; 41:e15262. [PMID: 38017692 DOI: 10.1111/dme.15262] [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: 07/26/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
AIMS Bespoke glycaemic control strategies following antenatal corticosteroids for women with diabetes in pregnancy (DIP) may mitigate hyperglycaemia. This study aims to identify predictive factors for the glycaemic response to betamethasone in a large cohort of women with DIP. METHODS Evaluation of a prospective cohort study of 347 consecutive DIP pregnancies receiving two doses of 11.4 mg betamethasone 24 h apart between 2017 and 2021 and treated with the Pregnancy-IVI intravenous insulin protocol. Regression modelling identified factors associated with maternal glycaemic time-in-range (TIR) and maternal insulin requirements following betamethasone. Factors associated with neonatal hypoglycaemia (glucose <2.6 mmol/L) in infants born within 48 h of betamethasone administration (n = 144) were investigated. RESULTS The mean maternal age was 31.9 ± 5.8 years, with gestational age at betamethasone of 33.5 ± 3.4 weeks. Gestational diabetes was present in 81% (12% type 1; 7% type 2). Pre-admission subcutaneous insulin was prescribed for 63%. On-infusion maternal glucose TIR (4.0-7.8 mmol/L) was 83% [IQR 77%-90%] and mean on-IVI glucose was 6.6 ± 0.5 mmol/L. Maternal hypoglycaemia (<3.8 mmol/L) was uncommon (0.47 h/100 on-IVI woman hours). Maternal glucose TIR was negatively associated with indicators of insulin resistance (type 2 diabetes, polycystic ovary syndrome), late-pregnancy complications (pre-eclampsia, chorioamnionitis) and the 1-h OGTT result. Intravenous insulin requirements were associated with type of diabetes, pre-eclampsia and intrauterine infection, the 1-h OGTT result and the timing of betamethasone administration. Neonatal hypoglycaemia was associated with pre-existing diabetes but not with measures of glycaemic control. CONCLUSION An intravenous infusion protocol effectively controls maternal glucose after betamethasone. A risk-factor-based approach may allow individualisation of therapy.
Collapse
Affiliation(s)
- Christopher W Rowe
- Department of Endocrinology and Diabetes, John Hunter Hospital, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Patrick Rosee
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Angeline Sathiakumar
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Soundarya Ramesh
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Vivian Qiao
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Jason Huynh
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Georgia Dennien
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Natasha Weaver
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Katie Wynne
- Department of Endocrinology and Diabetes, John Hunter Hospital, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
50
|
Cui Y, Wu S, Liu K, Zhao H, Ma B, Gong L, Zhou Q, Li X. Extra villous trophoblast-derived PDL1 can ameliorate macrophage inflammation and promote immune adaptation associated with preeclampsia. J Reprod Immunol 2024; 161:104186. [PMID: 38134680 DOI: 10.1016/j.jri.2023.104186] [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: 08/31/2023] [Revised: 10/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Severe preeclampsia (sPE) is a systemic syndrome that may originate from chronic inflammation. Maintaining maternal-fetal hemostasis by the co-inhibitory molecule programmed death ligand 1 (PDL1) can be favorable for ameliorating inflammation from immune cells. Apart from programmed death 1 (PD1) expression, decidual macrophages (dMs) produce inflammatory cytokines, in response to cells which express PDL1. However, strong evidence is lacking regarding whether the PDL1/PD1 interaction between trophoblasts and decidual macrophages affects inflammation during sPE development. METHODS To determine whether the trophoblast-macrophage crosstalk via the PDL1/PD1 axis modulates the inflammatory response in sPE-like conditions, at first, maternal-fetal tissues from sPE and normal patients were collected, and the PDL1/PD1 distribution was analyzed by Western blot, immunohistochemistry/ immunofluorescence and flow cytometry. Next, a coculture system was established and flow cytometry was used to identify how PDL1 was involved in macrophage-related inflammation under hypoxic stress. Transcriptional analysis was performed to clarify the inflammation-associated pathway induced by the PDL1/PD1 interaction. Finally, the Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME) mouse model was used to examine the effect of PDL1 on macrophage-related inflammation by measuring PE-like symptoms. RESULTS In maternal-fetal tissue from sPE patients, placental extravillous trophoblasts (EVTs) and dMs had a surprisingly increase of PDL1 and PD1 expression, respectively, accompanied by a higher percentage of CD68 +CD86 + dMs. In vitro experiments showed that trophoblast-derived PDL1 under hypoxia interacted with PD1 on CD14 +CD80 +macrophages, leading to suppression of inflammation through the TNFα-p38/NFκB pathway. Accordingly, the PE-like mouse model showed a reversal of PE-like symptoms and a reduced F4/80 + CD86 + macrophage percentage in the uterus in response to recombinant PDL1 protein administration, indicating the protective effect of PDL1. DISCUSSION Our results initially explained an immunological adaptation of trophoblasts under placental hypoxia, although this protection was insufficient. Our findings suggest the possible capacity of modulating PDL1 expression as a potential therapeutic strategy to target the inflammatory response in sPE.
Collapse
Affiliation(s)
- Yutong Cui
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Suwen Wu
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Ketong Liu
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Huanqiang Zhao
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Bo Ma
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Lili Gong
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Qiongjie Zhou
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.
| | - Xiaotian Li
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China; Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|