1
|
Kulkarni V, Alexander M, Bhosale R, Jain D, Deshpande P, Gitlin ES, Vaidyanathan A, Chalem A, Naik S, Gupte N, Nawani N, Gupta A, Mathad J. Discordance of 3rd and 4th generation QuantiFERON-TB Gold assays by pregnancy stages in India. J Clin Tuberc Other Mycobact Dis 2025; 38:100504. [PMID: 39758562 PMCID: PMC11697401 DOI: 10.1016/j.jctube.2024.100504] [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] [Indexed: 01/07/2025] Open
Abstract
Background Pregnancy and HIV affect CD4+ T lymphocytes and impact performance of QuantiFERON-TB Gold (QFT). We compared the results of QFT with QuantiFERON-TB Gold Plus (QFT-Plus), which also measures CD8+ responses to TB antigens, during pregnancy and postpartum. Methods We screened 516 pregnant women for TB infection (TBI) with IGRA. From 165 IGRA + pregnant women, QFT vs QFT-Plus results were compared at delivery and postpartum. Longitudinal changes in QFT-Plus were assessed in 74 pregnant women who received QFT-Plus testing at pregnancy, delivery, and postpartum. Results Through cross-sectional analysis of the IGRA + cohort, QFT-Plus showed higher positivity than QFT (80 % vs 65 %, p = 0.04) at delivery but no difference postpartum. Among 35 women with HIV, QFT-Plus returned more positive results than QFT at delivery and postpartum (76 % vs 47 %, p = 0.08; 90 % vs 80 %, p = 0.54), though not statistically significant. Longitudinally, QFT-Plus positivity by TB1 or TB2 was highest antepartum vs. delivery and postpartum (74 % vs. 58 % vs. 62 %; p = 0.09) and performed better than TB1 alone (100 % vs 90 %, p = 0.04) in women without HIV but not in women with HIV. Conclusions Performance of QFT-Plus was consistent across pregnancy, including at delivery when QFT positivity is lower. QFT-Plus may enhance antenatal TBI detection among pregnant women.
Collapse
Affiliation(s)
- Vandana Kulkarni
- Byramjee-Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, 3rd Floor, Infosys Superspecialty Building, Byramjee Jeejeebhoy Government Medical College & Sassoon General Hospitals, Jai Prakash Narayan Road, Pune 411001, India
- Center for Infectious Diseases in India, Johns Hopkins India, G Block, C1 Shopping Arcade, Konark Estate, Cannaught Road, Pune 411001, India
| | - Mallika Alexander
- Byramjee-Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, 3rd Floor, Infosys Superspecialty Building, Byramjee Jeejeebhoy Government Medical College & Sassoon General Hospitals, Jai Prakash Narayan Road, Pune 411001, India
- Center for Infectious Diseases in India, Johns Hopkins India, G Block, C1 Shopping Arcade, Konark Estate, Cannaught Road, Pune 411001, India
| | - Ramesh Bhosale
- Byramjee Jeejeebhoy Government Medical College, 3rd Floor, Dept. of Gynaecology and Obstetrics, Jai Prakash Narayan Road, Pune 411001, Maharashtra, India
| | - Divyashri Jain
- Byramjee-Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, 3rd Floor, Infosys Superspecialty Building, Byramjee Jeejeebhoy Government Medical College & Sassoon General Hospitals, Jai Prakash Narayan Road, Pune 411001, India
| | - Prasad Deshpande
- Byramjee-Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, 3rd Floor, Infosys Superspecialty Building, Byramjee Jeejeebhoy Government Medical College & Sassoon General Hospitals, Jai Prakash Narayan Road, Pune 411001, India
- Center for Infectious Diseases in India, Johns Hopkins India, G Block, C1 Shopping Arcade, Konark Estate, Cannaught Road, Pune 411001, India
| | - Emily Shira Gitlin
- Weill Cornell Medicine, Center for Global Health, 402 East 67th Street, 2nd Floor, New York, NY 10065, USA
| | - Arthi Vaidyanathan
- Duke University School of Medicine, DUMC 2927,40 Duke Medicine Circle,124 Davison Building, Durham, NC 27710, USA
| | - Andrea Chalem
- University of North Carolina Gillings School of Global Public Health, 135 Dauer Dr, Chapel Hill, NC 27599, USA
| | - Shilpa Naik
- Byramjee Jeejeebhoy Government Medical College, 3rd Floor, Dept. of Gynaecology and Obstetrics, Jai Prakash Narayan Road, Pune 411001, Maharashtra, India
| | - Nikhil Gupte
- Byramjee-Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, 3rd Floor, Infosys Superspecialty Building, Byramjee Jeejeebhoy Government Medical College & Sassoon General Hospitals, Jai Prakash Narayan Road, Pune 411001, India
- Center for Infectious Diseases in India, Johns Hopkins India, G Block, C1 Shopping Arcade, Konark Estate, Cannaught Road, Pune 411001, India
| | - Neelu Nawani
- Microbial Diversity Research Centre, Dr D.Y. Patil Biotechnology and Bioinformatics Institute, Dr D. Y. Patil Vidyapeeth, Service Rd, Tathawade, Pimpri-Chinchwad, Pune 411033, India
| | - Amita Gupta
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, 1830 East Monument Street/4th Floor Baltimore, Maryland 21287-2100 USA
| | - Jyoti Mathad
- Weill Cornell Medicine, Center for Global Health, 402 East 67th Street, 2nd Floor, New York, NY 10065, USA
| |
Collapse
|
2
|
X M. A synthetic review: natural history of amniote reproductive modes in light of comparative evolutionary genomics. Biol Rev Camb Philos Soc 2025; 100:362-406. [PMID: 39300750 DOI: 10.1111/brv.13145] [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: 12/05/2022] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
There is a current lack of consensus on whether the ancestral parity mode was oviparity (egg-laying) or viviparity (live-birth) in amniotes and particularly in squamates (snakes, lizards, and amphisbaenids). How transitions between parity modes occur at the genomic level has primary importance for how science conceptualises the origin of amniotes, and highly variable parity modes in Squamata. Synthesising literature from medicine, poultry science, reproductive biology, and evolutionary biology, I review the genomics and physiology of five broad processes (here termed the 'Main Five') expected to change during transitions between parity modes: eggshell formation, embryonic retention, placentation, calcium transport, and maternal-fetal immune dynamics. Throughout, I offer alternative perspectives and testable hypotheses regarding proximate causes of parity mode evolution in amniotes and squamates. If viviparity did evolve early in the history of lepidosaurs, I offer the nucleation site hypothesis as a proximate explanation. The framework of this hypothesis can be extended to amniotes to infer their ancestral state. I also provide a mechanism and hypothesis on how squamates may transition from viviparity to oviparity and make predictions about the directionality of transitions in three species. After considering evidence for differing perspectives on amniote origins, I offer a framework that unifies (i) the extended embryonic retention model and (ii) the traditional model which describes the amniote egg as an adaptation to the terrestrial environment. Additionally, this review contextualises the origin of amniotes and parity mode evolution within Medawar's paradigm. Medawar posited that pregnancy could be supported by immunosuppression, inertness, evasion, or immunological barriers. I demonstrate that this does not support gestation or gravidity across most amniotes but may be an adequate paradigm to explain how the first amniote tolerated internal fertilization and delayed egg deposition. In this context, the eggshell can be thought of as an immunological barrier. If serving as a barrier underpins the origin of the amniote eggshell, there should be evidence that oviparous gravidity can be met with a lack of immunological responses in utero. Rare examples of two species that differentially express very few genes during gravidity, suggestive of an absent immunological reaction to oviparous gravidity, are two skinks Lampropholis guichenoti and Lerista bougainvillii. These species may serve as good models for the original amniote egg. Overall, this review grounds itself in the historical literature while offering a modern perspective on the origin of amniotes. I encourage the scientific community to utilise this review as a resource in evolutionary and comparative genomics studies, embrace the complexity of the system, and thoughtfully consider the frameworks proposed.
Collapse
Affiliation(s)
- Maggs X
- Richard Gilder Graduate School at The American Museum of Natural History, 200 Central Park West, New York, NY, 10024, USA
- Christopher S. Bond Life Science Center at the University of Missouri, 1201 Rollins St, Columbia, MO, 65201, USA
- School of Life and Environmental Sciences at the University of Sydney, Heydon-Laurence Building A08, Sydney, NSW, 2006, Australia
| |
Collapse
|
3
|
Li K, Wang J, Liu X, Dang Y, Wang K, Li M, Zhang X, Liu Y. Identification of hub biomarkers and immune cell infiltrations participating in the pathogenesis of endometriosis. Sci Rep 2025; 15:2802. [PMID: 39843899 PMCID: PMC11754470 DOI: 10.1038/s41598-025-86164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
Endometriosis (EM) is a chronic disease that can cause pain and infertility in patients. As is well known, immune cell infiltrations (ICIs) play important roles in the pathogenesis of EM. However, the pathogenesis and biomarkers of EM that can be used in clinical practice and their relationship with ICIs still need to be elucidated. The gene expression datasets of EM and the healthy control were obtained from the Gene Expression Omnibus (GEO). To identify the central modules and explore the correlation between the gene network and EM, weighted gene co-expression network analysis (WGCNA) was executed. The hub genes were screened using machine learning. The qRT-PCR results showed that only CHMP4C and KAT2B differentially expressed in ectopic tissues compared to the normal. Subsequently, the samples were clustered based on the expression of CHMP4C and KAT2B. Depending on the differential expression genes of the two 2rG Clusters, the samples were divided into two gene Clusters. Significant differences in immune cell infiltrations were observed among the two 2rG Clusters and the two gene Clusters. Furthermore, varied immune checkpoint genes were shown to be correlated with EM. The qRT-PCR results showed that the two genes were significantly related to the ICI genes in EM. Hub genes CHMP4C and KAT2B are involved in the pathogenesis of EM by regulating ICI.
Collapse
Affiliation(s)
- Kang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology & Embryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jiaxu Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology & Embryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xuyue Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology & Embryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yifei Dang
- Department of Gynecology & Obstetrics, Qilu Hospital of Shandong University, Jinan, China
| | - Kaiting Wang
- Department of Gynecology & Obstetrics, Qilu Hospital of Shandong University, Jinan, China
| | - Manyu Li
- Department of Gynecology & Obstetrics, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoli Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology & Embryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuan Liu
- Department of Gynecology & Obstetrics, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
4
|
Fraile-Martinez O, García-Montero C, Gomez-Lahoz AM, Sainz F, Bujan J, Barrena-Blázquez S, López-González L, Díaz-Pedrero R, Álvarez-Mon M, García-Honduvilla N, Saez MA, Monserrat J, Ortega MA. Evidence of Inflammatory Network Disruption in Chronic Venous Disease: An Analysis of Circulating Cytokines and Chemokines. Biomedicines 2025; 13:150. [PMID: 39857734 PMCID: PMC11763091 DOI: 10.3390/biomedicines13010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Chronic venous disease (CVD) comprises a set of vascular disorders that affect the venous system with important local and systemic repercussions. A growing body of evidence displays the relationship between suffering from CVD and a marked deregulation of the immune inflammatory system. In this sense, the previous literature has reported some significant changes in the level of various circulating inflammatory parameters in these patients. However, more research is required to detail and deepen this complex relationship. Methods: In this work, we studied, using a multiplex technique, the levels of circulating cytokines and chemokines detectable in the serum of 40 patients with CVD and compared it with 38 healthy controls (HCs). In parallel, we performed Spearman's correlation analysis to explore potential inflammatory networks in CVD. Results: In this study, we measured circulating cytokines and chemokines in CVD patients using a multiplex assay. Results showed increased levels of several pro-inflammatory mediators (IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-12, IL-17A, IL-23, TNF-α, IFN-γ, fractalkine, ITAC, and GM-CSF) and a decrease in IL-13, with no significant changes in IL-4, IL-10, IL-21, MIP-1α, MIP-1β, or MIP-3α. The Spearman correlation analysis revealed strong, positive correlations among several inflammatory mediators in HC, particularly between TNF-alpha, IL-1β, IL-17A, and IL-23, forming a highly interconnected cytokine network. In contrast, CVD patients showed fewer, weaker, and distinct correlations, with new associations such as IFN-γ with IL-1β and IL-23, suggesting a disrupted inflammatory profile. Conclusions: The distinct inflammatory profile in CVD patients, characterized by altered cytokine and chemokine levels and a less coordinated cytokine network, underscores the reconfiguration of inflammatory pathways in this condition. These findings highlight potential therapeutic targets aimed at restoring immune balance and mitigating chronic inflammation in CVD.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Ana María Gomez-Lahoz
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Felipe Sainz
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Angiology and Vascular Surgery Service, Central University Hospital of Defence—UAH, 28047 Madrid, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Silvestra Barrena-Blázquez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Laura López-González
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Raul Díaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcala de Henares, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| |
Collapse
|
5
|
Sun Z, Wu W, Xiang Z, Gao H, Ju W, Uhm C, Hagemann IS, Woodard PK, Zhong N, Cahill AG, Wang Q, Wang Y. Quantitative and longitudinal assessment of human placental inflammation using diffusion basis spectrum imaging. NPJ WOMEN'S HEALTH 2025; 3:1. [PMID: 39759173 PMCID: PMC11698687 DOI: 10.1038/s44294-024-00049-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
Besides exchanging nutrients, gases, and wastes, placenta is an intermediary between maternal and fetal immune systems. However, no method exists to safely image and monitor placental inflammation during pregnancy. We customized a Magnetic Resonance Imaging (MRI) method, diffusion basis spectrum imaging (DBSI), to measure immune cells in placenta. We validated placental DBSI in simulations and ex-vivo specimens, then applied it to 202 MRI scans from 82 patients whose placentas were classified as non-inflammation (n = 70) or inflammation (n = 12). Our method imaged the 3D distribution of immune cells, revealing significantly greater infiltration in the inflammation placentas from early (2.8% ± 0.7% vs. 4.8% ± 0.65%, p < 0.01) to late pregnancy (4.75% ± 0.9% vs. 7.25% ± 2.13%, p < 0.01). DBSI detects elevated immune cell infiltration associated with placental inflammation and enables non-invasive imaging of placental inflammation, offering early detection and monitoring throughout pregnancy, facilitating personalized care and potentially improving pregnancy outcomes without ionizing radiation.
Collapse
Affiliation(s)
- Zhexian Sun
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
| | - Wenjie Wu
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
| | - Zezhen Xiang
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
- Department of Electrical & Systems Engineering, Washington University in St Louis, St Louis, MO USA
| | - Hansong Gao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
- Department of Electrical & Systems Engineering, Washington University in St Louis, St Louis, MO USA
| | - Weina Ju
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY USA
| | - Cherilyn Uhm
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY USA
| | - Ian S. Hagemann
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
- Department of Pathology & Immunology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
| | - Pamela K. Woodard
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
| | - Nanbert Zhong
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY USA
| | - Alison G. Cahill
- Department of Women’s Health, Dell Medical School, University of Texas, Austin, TX USA
| | - Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
| | - Yong Wang
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
- Department of Electrical & Systems Engineering, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO USA
| |
Collapse
|
6
|
Pandit A, Shah SM, Shah RA, Qureshi S, Sethi RS, Bhat F, Malik A, Parray O, Yaqoob H, Saleem M. Regulatory T cells in bovine fertility: Current understanding and future prospects. Anim Reprod Sci 2025; 272:107655. [PMID: 39616725 DOI: 10.1016/j.anireprosci.2024.107655] [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: 05/26/2024] [Revised: 09/17/2024] [Accepted: 11/24/2024] [Indexed: 12/20/2024]
Abstract
Regulatory T cells (Tregs) have emerged as crucial players in maintaining maternal-fetal tolerance and promoting successful pregnancy outcomes. This review examines the importance of these cells in pregnancy, drawing on human and animal-based studies, with a focus on their role in bovine fertility. Tregs employ various mechanisms to mediate maternal-fetal tolerance, including regulation of effector T-cell responses, interactions with innate immune cells in the uterine microenvironment, and modulation of trophoblast function. In humans, Treg dynamics during normal pregnancy and alterations in pregnancy complications provide compelling evidence for their involvement in maintaining fetal-maternal harmony. Animal models, particularly mouse studies, have further elucidated the importance of Tregs in preventing fetal rejection and promoting successful pregnancy outcomes. The review also explores the characterization of bovine Tregs, highlighting their similarities and unique features compared to human and rodent counterparts. Recent studies have indicated the presence and potential significance of Tregs in the bovine uterine environment during early pregnancy. Translational applications of Treg research in livestock fertility are discussed, with a focus on immunomodulatory strategies for enhancing Treg function, such as antigen-specific tolerance induction, pharmacological targeting of Treg pathways, and cell-based therapies using autologous or allogeneic Tregs. The review concludes by emphasizing the potential impact of Treg-based strategies on the livestock industry and the broader implications for human reproductive health. Future research directions are outlined, underscoring the need for further investigations into the role of Tregs in bovine reproductive tissues and their relationship with fertility outcomes.
Collapse
Affiliation(s)
- Arif Pandit
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India.
| | - Syed M Shah
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Riaz A Shah
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Sabia Qureshi
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Sciences, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - R S Sethi
- College of Dairy Sciences, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - Faheem Bhat
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Abrar Malik
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Oveas Parray
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Hilal Yaqoob
- Center of Excellence in Animal Reproductive Biotechnology, Mountain Livestock Research Institute, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Masood Saleem
- Directorate of Research, Sher E Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, Jammu and Kashmir, India.
| |
Collapse
|
7
|
Panneerselvam D, Murugesan A, Raveendran SK, Kumar JS, Venkataraman P. Examining the hidden dangers: Understanding how microplastics affect pregnancy. Eur J Obstet Gynecol Reprod Biol 2025; 304:53-62. [PMID: 39580908 DOI: 10.1016/j.ejogrb.2024.11.024] [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: 07/22/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Microplastics, a fast-growing environmental concern, play a crucial role in developing the major pollution crisis that affects nearly the entire surface of the planet. Microplastics are tiny particles, measuring less than 5 mm which are ubiquitous, in occurrence, and found in a wide array of products including plastic packaging, synthetic textiles, seafood, fruits, vegetables, salt, sugar, bottled water, and even personal care products. The presence of microplastics in our environment and the potential adverse health effects they may cause have made them a significant perturbation in recent years. Pregnancy is a potentially life-changing experience that entails several apprehensions and new responsibilities for women. For expectant mothers, it is imperative to be aware of the implications of microplastics during pregnancy. One threatened concern is the potential transfer of microplastics across the placenta, which could expose the developing fetus to these particles. Although research on the impact of microplastics on pregnancy is still in its early stages, preliminary findings indicate potential risks that expectant mothers should be aware of. The timing of exposure during pregnancy may play a significant role in the potential risks associated with these tiny particles. In this review, we will delve into the topic, exploring how microplastics enter the body and the potential mechanism by which they pose risks to pregnancy outcomes.
Collapse
Affiliation(s)
- Deboral Panneerselvam
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anuradha Murugesan
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| | - Sajeetha Kumari Raveendran
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Janardanan Subramonia Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - P Venkataraman
- Department of Medical Research, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
8
|
Christian LM, Brown RL, Carroll JE, Thayer JF, Lewis TT, Gillespie SL, Fagundes CP. Pathways to maternal health inequities: Structural racism, sleep, and physiological stress. Brain Behav Immun 2025; 123:502-509. [PMID: 39362504 PMCID: PMC11624070 DOI: 10.1016/j.bbi.2024.09.037] [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: 12/20/2023] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024] Open
Abstract
Racial inequities in health are vast and well-documented, particularly regarding maternal and infant health. Sleep health, including but not limited to duration and quality, is central to overall health and well-being. However, research has not adequately addressed how racism embedded in structures and systems, in addition to individual experiences, may affect maternal health by impacting sleep. In this critical review, we aim to 1) synthesize findings, emphasizing collaborative studies within our group, 2) highlight gaps in knowledge, and 3) propose a theoretical framework and methodological approach for moving the field forward. Specifically, we focus on findings and future directions linking perinatal sleep, cardiovascular and immune function, and racial disparities in maternal health. Because too few studies look beyond individual-level determinants of sleep deficiencies among Black Americans, we assert a critical need for research that bridges multiple levels of analysis (e.g., individual, community, society) and provides recommendations for specific health parameters that researchers in this area can target. Although the need to understand and address perinatal health disparities is clear, the goal of identifying multilevel mechanisms underlying how racism in one's environment and daily life may interact to affect health extends far beyond pregnancy research.
Collapse
Affiliation(s)
- Lisa M Christian
- Department of Psychiatry & Behavioral Health and the Institute for Behavioral Medicine Research and The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Ryan L Brown
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Judith E Carroll
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles, USA
| | - Julian F Thayer
- Department of Psychological Science, University of California at Irvine, Irvine, CA, USA
| | - Tené T Lewis
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Shannon L Gillespie
- Martha S. Pitzer Center for Women, Children and Youth, College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Christopher P Fagundes
- Department of Psychological Sciences, Rice University, Houston, TX, USA; Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
9
|
D'Ippolito S, Gavi F, Granieri C, De Waure C, Giuliano S, Cosentino F, Tersigni C, Scambia G, Di Simone N. Efficacy of Corticosteroids in Patients With Recurrent Pregnancy Loss: A Systematic Review and Meta-Analysis. Am J Reprod Immunol 2025; 93:e70037. [PMID: 39777851 DOI: 10.1111/aji.70037] [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: 10/22/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Recurrent pregnancy loss (RPL) represents a complication of pregnancy occurring in 1%-3% of all couples trying to conceive. About 50%-60% of RPL cases remain idiopathic, therefore therapeutic strategies seem empirical and based on unproven evidence. We investigated the efficacy of corticosteroids in women with RPL. We conducted a systematic review and meta-analysis, up to August 2024, in the PubMed, Scopus, and Web of Science databases, including studies on idiopathic RPL women and comparing corticosteroids versus control treatment. Primary outcome was the ongoing pregnancy rate beyond 12 weeks of gestation; secondary outcomes were live birth rate (LBR), stillbirth, birth weight, incidence of preeclampsia and/or gestational diabetes, gestational age at delivery, and fetal abnormalities. Four studies comprising 417 RPL women randomly assigned to steroid or control treatment were included. We found that oral corticosteroids significantly increase the ongoing pregnancy rate beyond 12 weeks of gestation compared to the control group (log OR [odds ratio] = 1.49 [0.32, 2.67], p = 0.01), with high heterogeneity (I2 = 75%), and improve LBR (log OR = 0.9 [0.11, 1.69], p = 0.03), with low heterogeneity (I2 = 0.05%). However, the limited number of studies significantly limits the strength of the findings. Also, the benefit/risk assessment of the use of corticosteroids in early pregnancy for RPL is still unclear.
Collapse
Affiliation(s)
- Silvia D'Ippolito
- Dipartimento di Medicina e Scienze della Salute "Vincenzo Tiberio,", University of Molise UNIMOL, Campobasso, Italy
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Filippo Gavi
- Department of Urology, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Chiara Granieri
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Chiara De Waure
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sara Giuliano
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesco Cosentino
- Dipartimento di Medicina e Scienze della Salute "Vincenzo Tiberio,", University of Molise UNIMOL, Campobasso, Italy
| | - Chiara Tersigni
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giovanni Scambia
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
10
|
Gharipour M, Craig JM, Stephenson G. Epigenetic programming of obesity in early life through modulation of the kynurenine pathway. Int J Obes (Lond) 2025; 49:49-53. [PMID: 39424650 DOI: 10.1038/s41366-024-01647-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
11
|
Zhang Q, Yang D, Han X, Ren Y, Fan Y, Zhang C, Sun L, Ye T, Wang Q, Ban Y, Cao Y, Zou H, Zhang Z. Alarmins and their pivotal role in the pathogenesis of spontaneous abortion: insights for therapeutic intervention. Eur J Med Res 2024; 29:640. [PMID: 39741354 DOI: 10.1186/s40001-024-02236-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
Alarmins are a class of molecules released when affected cells damaged or undergo apoptosis. They contain various chemotactic and immunomodulatory proteins or peptides. These molecules regulate the immune response by interacting with pattern recognition receptors (PRRs) and play important roles in inflammatory response, tissue repair, infection defense, and cancer treatment. Spontaneous abortion (SA) is a common pregnancy-related disease, and its pathogenesis has been puzzling clinicians, so it needs to be further studied. In this paper, we first reviewed the research status of various alarmins and SA, focusing on the role of high mobility box 1 (HMGB1), interleukin33 (IL-33), interleukin1β (IL-1β) and S-100 protein (S100 protein) in immune response, inflammation, embryonic development and abortion. Subsequently, this paper summarized the effect of alarmins on pregnancy outcome by influencing angiogenesis-related factors. Finally, from the perspective of aseptic inflammation, the pro-inflammatory signaling pathways involved in various alarmins and their targeted drugs were reviewed. By focusing on specific molecules in alarmins and their receptors and signaling pathways, we can more accurately conduct drug research and development. The purpose of this review is to explore the role of alarmins in SA, and provide important references for early detection of abortion risk, revealing the disease mechanism, developing new therapies and improving the prognosis of patients.
Collapse
Affiliation(s)
- Qiqi Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China
| | - Dandan Yang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China
| | - Xingxing Han
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Yu Ren
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, Anhui, China
| | - Yongqi Fan
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Chao Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, China
| | - Lei Sun
- Department of Clinical Medical, The First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Tingting Ye
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Qiushuang Wang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Youhao Ban
- Hefei Anhua Trauma Rehabilitation Hospital, Hefei, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Huijuan Zou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China.
| | - Zhiguo Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
12
|
Yongkai W, Shuhui Z, Li M. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the immune system maternal-fetal interface during palatal development. J Mol Histol 2024; 56:60. [PMID: 39730832 DOI: 10.1007/s10735-024-10331-0] [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: 10/02/2024] [Accepted: 12/05/2024] [Indexed: 12/29/2024]
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is an important environmental pollutant that disturbs the immune balance of the maternal-fetal interface (MFI) and is also a common environmental factor for the formation of cleft palate (CP). Therefore, the purpose is to investigate whether TCDD can cause CP by disrupting the immune balance of the maternal-fetal interface. Fifteen C57BL/6J mice were randomly assigned to three groups: control group, TCDD group, and TCDD plus Freund's complete adjuvant (FCA) (TCDD + FCA) group. Peripheral blood, placentas, and palatal tissues were collected for H&E, flow cytometry, and ELISA. In the TCDD group, the placental diameter, the number of placental labyrinth vessels, and the area of sponge layer cells were all significantly reduced. At embryonic day (E) 17.0, there was a significant decrease in T-helper 1 (Th1) and Th2 cells in the peripheral blood of pregnant mice. Additionally, the levels of interferon-γ (IFN-γ) and interleukin-4 (IL-4), particularly IL-4, were significantly decreased. However, after treatment with FCA, the distance between the palatal shelves was reduced, and the placental weight, the number of labyrinth vessels, and the area of the cavernous cells in the placenta also increased. The number of Th1 and Th2 cells significantly increased, returning to the levels observed in the control group, with a more pronounced increase in the number of Th2 cells. In conclusion, TCDD may induce CP by disrupting the homeostasis of the MFI. The precise mechanisms by which TCDD impacts the immune system at the MFI require further investigation.2,3,7,8-- (TCDD) , (MFI) , (CP) 。, TCDD CP。 15 C57BL/6J 3 :、TCDD TCDD (FCA) (TCDD + FCA) 。、 H&E、 ELISA。TCDD 、。 17.0 (E) , T 1 (Th1) Th2 。,-γ (IFN-γ) -4 (IL-4), IL-4 。, FCA ,,、。Th1 Th2 ,,Th2 。,TCDD MFI CP。TCDD MFI 。.
Collapse
Affiliation(s)
- Wang Yongkai
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.51, Weiliu Road, Jinan, Shandong Province, 250021, China
- Stomatological College, Shandong First Medical University, Jinan, Shandong Province, China
| | - Zhang Shuhui
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.51, Weiliu Road, Jinan, Shandong Province, 250021, China
- Stomatological College, Shandong First Medical University, Jinan, Shandong Province, China
| | - Ma Li
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.51, Weiliu Road, Jinan, Shandong Province, 250021, China.
- Stomatological College, Shandong First Medical University, Jinan, Shandong Province, China.
| |
Collapse
|
13
|
Wang J, Li C, Li W, Tao Y, Li Y. Epicardial adipose tissue thickness associated with preeclampsia and birth weight in early pregnancy. Hypertens Pregnancy 2024; 43:2390531. [PMID: 39129211 DOI: 10.1080/10641955.2024.2390531] [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: 03/29/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Preeclampsia (PE) increases the risk of many adverse maternal and fetal outcomes. This study was to investigate the correlation between epicardial adipose tissue (EAT) thickness and PE and birth weight. METHODS This was a single-center retrospective study, 221 patients with PE were selected, and 81 women without hypertension and proteinuria were selected as a comparison. Echocardiogram was performed in their first prenatal examinations at 11-13 gestational weeks, and the thickness of EAT was measured. At the subsequent follow-up, the birth weight was recorded. RESULTS EAT thickness was significantly elevated (6.60 ± 1.34 vs. 5.71 ± 1.79 mm, p < 0.001) in severe PE compared to mild PE. In the multivariate analysis, EAT thickness (OR 5.671, 95% CI, 1.991-16.150, p = 0.001), and C reactive protein (OR 4.097, 95% CI, 2.323-7.224, p < 0.001) were found as significant independent predictors of severe PE after adjusting for other risk factors. Linear regression analysis showed that hs-CRP, EAT thickness, and severe PE significantly negatively affected birth weight. CONCLUSION EAT thickness can be used to identify pregnant women with severe PE risks and low birth weight. It is an independent risk factor for severe PE but is not a valuable sign of mild PE.
Collapse
Affiliation(s)
- Jing Wang
- Department of obstetrics and gynecology, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Chunfeng Li
- Department of obstetrics and gynecology, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Wen Li
- Department of Cardiology, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Yexiao Tao
- Department of Cardiology, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Yong Li
- Department of Cardiology, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Siwakoti RC, Harris SM, Ferguson KK, Hao W, Cantonwine DE, Mukherjee B, McElrath TF, Meeker JD. Prenatal exposure to per- and polyfluoroalkyl substances (PFAS) and their influence on inflammatory biomarkers in pregnancy: Findings from the LIFECODES cohort. ENVIRONMENT INTERNATIONAL 2024; 194:109145. [PMID: 39550829 DOI: 10.1016/j.envint.2024.109145] [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: 09/04/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are fluorinated chemicals linked to adverse pregnancy and birth outcomes. However, the underlying mechanisms, specifically their effects on maternal inflammatory processes, are not well characterized. OBJECTIVE We examined associations between prenatal PFAS exposure and repeated measures of inflammatory biomarkers, including C-reactive protein (CRP) and four cytokines [Interleukin-10 (IL-10), IL-1β, IL-6, and tumor necrosis factor-α (TNF-α)]. METHODS We analyzed data from 469 pregnant women in a nested case-control study of preterm birth at Brigham and Women's Hospital in Boston, Massachusetts (2006-2008). We measured nine PFAS in early pregnancy plasma samples (median gestation: 10 weeks), with inflammatory biomarkers measured at median gestations of 10, 18, 26, and 35 weeks. We used linear mixed models for repeated measures and multivariable regression for visit-specific analysis to examine associations between each PFAS and inflammation biomarker, adjusting for maternal demographics, pre-pregnancy BMI, and parity. We examined the effects of PFAS mixture using sum of all PFAS (∑PFAS) and quantile-based g-computation approaches. RESULTS We observed consistent inverse associations between most PFAS and cytokines, specifically IL-10, IL-6, and TNF-α, in both single pollutant and mixture analyses. For example, an interquartile range increase in perfluorooctanesulfonic acid was associated with -10.87 (95% CI: -19.75, -0.99), -13.91 (95% CI: -24.11, -2.34), and -8.63 (95% CI: -14.51, -2.35) percent change in IL-10, IL-6, and TNF-α levels, respectively. Fetal sex, maternal race, and visit-specific analyses showed associations between most PFAS and cytokines were generally stronger in mid-pregnancy and among women who delivered males or identified as African American. CONCLUSIONS The observed suppression of both regulatory (IL-10) and pro-inflammatory (TNF-α) cytokines suggests that PFAS may alter maternal inflammatory processes or immune functions during pregnancy. Further research is needed to understand the effects of both legacy and newer PFAS on inflammatory pathways and their broader clinical implications.
Collapse
Affiliation(s)
- Ram C Siwakoti
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sean M Harris
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Kelly K Ferguson
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Wei Hao
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - David E Cantonwine
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Thomas F McElrath
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John D Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Song L, Yang C, Ji G, Hu R. The role and potential treatment of macrophages in patients with infertility and endometriosis. J Reprod Immunol 2024; 166:104384. [PMID: 39442472 DOI: 10.1016/j.jri.2024.104384] [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: 05/18/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
Endometriosis is characterized as a macrophage-related ailment due to its strong link with immune dysfunction. Understanding the status of macrophage polarization in the context of endometriosis-related infertility is crucial for advancing diagnostic and therapeutic strategies. Our comprehensive review delves into the foundational understanding of macrophages and their profound influence on both endometriosis and infertility. Additionally, we illuminate the complex role of macrophages in infertility and endometriosis specifically. Finally, we focused on four critical dimensions: follicular fluid, the intraperitoneal environment, endometrial receptivity, and strategies for managing endometriosis. It is clear that throughout the progression of endometriosis, the diverse polarization states of macrophages play a pivotal role in the internal reproductive environment of infertile individuals grappling with this condition. Modulating macrophage polarization in the reproductive environment of endometriosis patients could address infertility challenges more effectively, offering a promising pathway for treating infertility associated with endometriosis.
Collapse
Affiliation(s)
- Linlin Song
- Department of Gynecology, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Caihong Yang
- Department of Gynecology, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Guiyi Ji
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Rong Hu
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
16
|
Kyvelidou C, Haselrieder S, von Gierke M, Gostner JM, Biasio W, Wirleitner B, Heufler C, Toth B, Hofer-Tollinger S. Dendritic cells under the control of the preimplantation embryo secretome: an in vitro study. Reprod Biol Endocrinol 2024; 22:150. [PMID: 39578791 PMCID: PMC11585248 DOI: 10.1186/s12958-024-01319-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
OBJECTIVE To study the crosstalk between maternal immune cells and the developing embryo by investigating the immunogenic properties of human blastocyst spent media (SM) on dendritic cells. METHODS In this prospective multicenter experimental study, human preimplantation embryo spent media were collected after blastocyst formation, grouped based on successful or unsuccessful implantation, and analyzed by protein array or used to stimulate monocyte derived dendritic cells (moDC). The immunomodulatory properties of SM on moDC were investigated by analyzing changes in phenotype, cytokine secretion, indoleamine 2,3-dioxygenase (IDO) activity, and ability to activate T cells. RESULTS A plethora of cytokines and growth factors secreted from preimplantation embryos was detected. Exposure to embryo SM altered the phenotype of moDC in a manner dependent on the implantation outcome. Specifically, SM from non-implanted embryos increased the expression of co-stimulatory molecules and activation markers on moDC. Furthermore, SM treated dendritic cells secreted low levels of cytokines and growth factors and were able to stimulate naïve T cells. Activation of IDO was decreased in moDC after stimulation with SM. CONCLUSIONS Our findings show that human preimplantation embryos secrete an abundance of molecules with the ability to significantly affect and even regulate immune cells in their environment.
Collapse
Affiliation(s)
- Christiana Kyvelidou
- Department of Gynaecological Endocrinology and Reproductive Medicine, Medical University of Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria
| | - Sofia Haselrieder
- Department of Gynaecological Endocrinology and Reproductive Medicine, Medical University of Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria
| | - Maria von Gierke
- Department of Gynaecological Endocrinology and Reproductive Medicine, Medical University of Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria
| | - Johanna M Gostner
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Biasio
- Department of Gynaecological Endocrinology and Reproductive Medicine, Medical University of Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria
| | | | - Christine Heufler
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Bettina Toth
- Department of Gynaecological Endocrinology and Reproductive Medicine, Medical University of Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria
| | - Susanne Hofer-Tollinger
- Department of Gynaecological Endocrinology and Reproductive Medicine, Medical University of Innsbruck, Anichstraße 35, Innsbruck, 6020, Austria.
| |
Collapse
|
17
|
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
|
18
|
Mor G, Singh A, Yang J, Adzibolosu N, Cai S, Kauf E, Yang L, Li Q, Li H, Werner A, Parthasarathy S, Ding J, Fortier J, Rodriguez-Garcia M, Diao LH. Decoding Functional and Developmental Trajectories of Tissue-Resident Uterine Dendritic Cells Through Integrative Omics. RESEARCH SQUARE 2024:rs.3.rs-5424920. [PMID: 39606471 PMCID: PMC11601813 DOI: 10.21203/rs.3.rs-5424920/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Uterine dendritic cells (uDCs) are critical for endometrial function, yet their origin, molecular characteristics, and specific roles during the pre- and post-implantation periods in the human endometrium remain largely unknown. The complexity of the endometrial environment makes defining the contributions of uDCs subtypes challenging. We hypothesize that distinct uDC subsets carry out specialized functions, and that resident progenitor DCs generate these subtypes. Employing single-cell RNA sequencing on uterine tissues collected across different menstrual phases and during early pregnancy, we identify several uDCs subtypes, including resident progenitor DCs. CITE-seq was performed on endometrial single-cell suspensions to link surface protein expression with key genes identified by the RNAseq analysis. Our analysis revealed the developmental trajectory of the uDCs along with the distinct functional roles of each uDC subtype, including immune regulation, antigen presentation, and creating a conducive environment for embryo implantation. This study provides a comprehensive characterization of uDCs, serving as a foundational reference for future studies for better understanding female reproductive disorders such as infertility and pregnancy complications.
Collapse
Affiliation(s)
| | | | - Jing Yang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | | | - Songchen Cai
- Shenzhen Zhongshan Obstetrics & Gynecology Hospital
| | | | | | - Qiyuan Li
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Hanjie Li
- Shenzhen Institutes of Advanced Technology
| | | | | | | | | | | | | |
Collapse
|
19
|
Rezaei M, Moghoofei M. The role of viral infection in implantation failure: direct and indirect effects. Reprod Biol Endocrinol 2024; 22:142. [PMID: 39529140 PMCID: PMC11552308 DOI: 10.1186/s12958-024-01303-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Implantation is the key initial complex stage of pregnancy. Several factors are involved in implantation, but acute and controlled inflammation has been shown to play as a key role. On the other hand, the role of viral infections in directly infecting blastocyst and trophoblast and inducing chronic and uncontrolled inflammation and disrupting microRNAs expression can make this review strongly attractive and practical. We aim to provide an overview of viral infections as the potential etiology of unsuccessful implantation pathophysiology through alteration of the cellular and molecular endometrial microenvironment. Based on our search, this is the first review to discuss the role of inflammation associated with viral infection in implantation failure.
Collapse
Affiliation(s)
- Marzieh Rezaei
- Department of Obstetrics and Gynecology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Moghoofei
- Infectious Diseases Research Center, Health Research Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
20
|
Chen S, Zhang J, Chen J, Ke J, Huang Y, Du X, Fu B, Wei H. Compromised C3b-VSIG4 axis between decidual NK cells and macrophages contributes to recurrent spontaneous abortion. J Transl Med 2024; 22:1017. [PMID: 39529122 PMCID: PMC11556194 DOI: 10.1186/s12967-024-05829-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
NK cells and macrophages constitute the predominant immune cell subsets in the decidua during the first trimester of pregnancy, with macrophages typically adopting an anti-inflammatory phenotype. Conversely, in the third trimester, macrophages undergo a shift towards a pro-inflammatory phenotype concurrent with a reduction in NK cell numbers. The direct regulatory impact of NK cells on macrophage phenotype remains poorly explored. In our investigation, we observed that ICAM1+ macrophages stimulate the expression of intracellular C3 in LFA1+ decidual NK cells. Notably, Cathepsin W within NK cells exhibit the potential to generate active C3b fragments, effectively inhibit the proinflammatory phenotype of macrophages by binding to VSIG4. Our study unveils a direct regulatory mechanism orchestrated by decidual NK cells over macrophages, providing a potential pathogenic explanation for recurrent spontaneous abortion.
Collapse
Affiliation(s)
- Siao Chen
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Jinghe Zhang
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Jian Chen
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China
- Department of Intensive Care Unit, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Jieqi Ke
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Yu Huang
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianghui Du
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Binqing Fu
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China.
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China.
| | - Haiming Wei
- Department of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, Anhui, China.
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
21
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [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: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Basanta S, Stadtmauer DJ, Maziarz JD, McDonough-Goldstein CE, Cole AG, Dagdas G, Wagner GP, Pavličev M. Hallmarks of uterine receptivity predate placental mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621939. [PMID: 39574771 PMCID: PMC11580939 DOI: 10.1101/2024.11.04.621939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Embryo implantation requires tightly coordinated signaling between the blastocyst and the endometrium, and is crucial for the establishment of a uteroplacental unit that persists until term in eutherian mammals. In contrast, marsupials, with a unique life cycle and short gestation, make only brief fetal-maternal contact and lack implantation. To better understand the evolutionary link between eutherian implantation and its ancestral equivalent in marsupials, we compare single-cell transcriptomes from the receptive and non-receptive endometrium of the mouse and guinea pig with that of the opossum, a marsupial. We identify substantial differences between rodent peri-implantation endometrium and opossum placental attachment, including differences in the diversity and abundance of stromal and epithelial cells which parallel the difference between histotrophic and hemotrophic provisioning strategies. We also identify a window of conserved epithelial gene expression between the opossum shelled blastocyst stage and rodent peri-implantation, including IHH and LIF . We find strong conservation of blastocyst proteases, steroid synthetases, Wnt and BMP signals between eutherians and the opossum despite its lack of implantation. Finally, we show that the signaling repertoire of the maternal uterine epithelium during implantation displays substantial overlap with that of the post-implantation placental trophoblast, suggesting that the fetal trophoblast can compensate for the loss of endometrial epithelium in eutherian invasive placentation. Together, our results suggest that eutherian implantation primarily involved the re-wiring of maternal signaling networks, some of which were already present in the therian ancestor, and points towards an essential role of maternal innovations in the evolution of invasive placentation.
Collapse
Affiliation(s)
- Silvia Basanta
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
| | - Daniel J. Stadtmauer
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Department of Ecology & Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Jamie D. Maziarz
- Department of Ecology & Evolutionary Biology, Yale University, New Haven, CT, USA
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Caitlin E. McDonough-Goldstein
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Department of Integrative Biology, University of Wisconsin-Madison, WI, USA
| | - Alison G. Cole
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Gülay Dagdas
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
| | - Günter P. Wagner
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Department of Ecology & Evolutionary Biology, Yale University, New Haven, CT, USA
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Mihaela Pavličev
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Complexity Science Hub Vienna, Vienna, Austria
| |
Collapse
|
23
|
Kiemde F, Sorgho H, Zango SH, Some GF, Rouamba T, Traore O, Kabore B, Natama HM, Hien YE, Valea I, Schallig H, Tinto H. Effects of gestational age on blood cortisol and prolactin levels during pregnancy in malaria endemic area. PLoS One 2024; 19:e0310372. [PMID: 39495748 PMCID: PMC11534236 DOI: 10.1371/journal.pone.0310372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/30/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND The hormonal shift occurring in pregnant women is crucial for the outcome of pregnancy. We conducted a study in pregnant women living in a malaria endemic area to determine the potential effect of gestational age on the modulation of the endocrine system by cortisol and prolactin production during pregnancy. METHODS Primigravidae and multigravidae with a gestational age between 16-20 weeks were included in the study and followed up to delivery and 6-7 weeks thereafter. Venous blood was collected at scheduled visit: Visit 1 (V1; 16-20 weeks of amenorrhea), Visit 2 (V2; 28 ±1 weeks of pregnancy), Visit 3 (V3; 32 ±1 weeks of pregnancy), Visit4 (V4; delivery) and Visit5 (V5; 6-7 weeks after delivery). In addition, a cord blood sample was also collected during labour at delivery. Nulliparous and primiparous/multiparous non-pregnant women were enrolled in the control group. Cortisol and prolactin plasma concentrations were measured using ichroma II and i-chamber apparatus. Light microscopy was used to detect Plasmodium falciparum infections. A linear mixed-effects regression (LMER) model was used to assess the association between the variation of cortisol titres and prolactin levels during the pregnancy and the post-partum. RESULTS Results showed that cortisol and prolactin levels in the peripheral blood were globally up-regulated during pregnancy. Concentrations of cortisol during follow-up was significantly higher in primigravidae than in multigravidae during the whole pregnancy (p<0.024). Moreover, the level of prolactin which was higher before delivery in primigravidae reversed at delivery and postpartum visit, but the difference was not statistically significant during the follow-up (V1 to V5) (p = 0.60). The cortisol level in peripheral blood at delivery was higher than that in the cord blood, and conversely for prolactin. Cortisol and prolactin levels decreased after delivery, though the level of prolactin was still higher than that at enrolment. An increase of one unit of prolactin was associated with the decrease of the average concentration of cortisol by 0.04 ng/ml (p = 0.009). However, when cortisol increases with one unit, the average concentration of prolactin decreases by 1.16 ng/ml (p = 0.013). CONCLUSION These results showed that the up-regulation effects of cortisol and prolactin are related to gestational age. A The downward regulation effect that both hormones have on each other during the pregnancy when each increase to 1 unit (1.0 ng/ml) was also reported.
Collapse
Affiliation(s)
- Francois Kiemde
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Hermann Sorgho
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Serge Henri Zango
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Gnohion Fabrice Some
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Ousmane Traore
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Berenger Kabore
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Hamtandi Magloire Natama
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Yeri Esther Hien
- Unité de Recherche et de Formation en Sciences de la Vie et de la Terre (URF-SVT), Université Joseph Ki-Zerbo Ouaga 1, Ouagadougou, Burkina Faso
| | - Innocent Valea
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Henk Schallig
- Department of Medical Microbiology and Infection Prevention, Laboratory for Experimental Parasitology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centres, Academic Medical Centre at the University of Amsterdam, Amsterdam, The Netherlands
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| |
Collapse
|
24
|
Miodownik S, Sheiner E. Stress and Pregnancy Outcomes: A Review of the Literature. Semin Thromb Hemost 2024. [PMID: 39496304 DOI: 10.1055/s-0044-1792002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
The human body has the ability to adapt to changing circumstances, and mobilizes various biological systems in order to do so. When exposed to stressful conditions, the endocrine, nervous, and immune systems come together to aid in maintaining homeostasis; however, during periods of chronic stress, these systems can become maladaptive and lead to long-term detrimental health outcomes. Amongst the lingering effects associated with chronic stress exposure, increasingly, studies are identifying a link to adverse pregnancy and neonatal outcomes. This review explores what has been uncovered in the field to date, and examines the effects of stress on fertility and gestation. Establishing additional factors which put women at risk for adverse pregnancy outcomes can aid in identifying a vulnerable population who could benefit from early stress-reducing interventions.
Collapse
Affiliation(s)
- Shayna Miodownik
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Eyal Sheiner
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
25
|
Cao XY, Li MY, Shao CX, Shi JL, Zhang T, Xie F, Peng T, Li MQ. Fatty Acid Metabolism Disruptions: A Subtle yet Critical Factor in Adverse Pregnancy Outcomes. Int J Biol Sci 2024; 20:6018-6037. [PMID: 39664564 PMCID: PMC11628336 DOI: 10.7150/ijbs.103404] [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: 09/08/2024] [Accepted: 10/26/2024] [Indexed: 12/13/2024] Open
Abstract
The establishment and maintenance of pregnancy encompass a series of complex and high-energy-consuming physiological processes, resulting in a significant energy demand. Fatty acids, one of the most essential nutrients, play a crucial role in energy supply via oxidation and perform critical biological functions such as anti-inflammatory and anti-oxidant effects, which substantially impact human health. Disordered fatty acid metabolism can cause anomalies in fetal growth and development, as well as a range of pregnancy problems, which can influence the health of both the mother and the fetus. In this review, we innovatively explore the relationship between fatty acid metabolism abnormalities and pregnancy complications, emphasizing the potential of dietary interventions with polyunsaturated fatty acids in improving pregnancy outcomes. These findings provide important evidence for clinical interventions and enhance the understanding and practical application of health management during pregnancy.
Collapse
Affiliation(s)
- Xiao-Yan Cao
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
| | - Meng-Ying Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
| | - Chang-Xiang Shao
- Department of Obstetrics and Gynecology, Shanghai Changning Maternity & Infant Health Hospital, East China Normal University, Shanghai 200051, People's Republic of China
| | - Jia-Lu Shi
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Feng Xie
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
| | - Ting Peng
- Department of Obstetrics and Gynecology, Shanghai Changning Maternity & Infant Health Hospital, East China Normal University, Shanghai 200051, People's Republic of China
| | - Ming-Qing Li
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, People's Republic of China
| |
Collapse
|
26
|
Suleri A, White T, de Witte L, Gigase F, Cecil CAM, Jaddoe VWV, Breen M, Hillegers MHJ, Muetzel RL, Bergink V. Maternal Immune Activation and Child Brain Development: A Longitudinal Population-Based Multimodal Neuroimaging Study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024:S2451-9022(24)00312-4. [PMID: 39491788 DOI: 10.1016/j.bpsc.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Maternal immune activation (MIA) has been hypothesized to have an adverse effect on child neurodevelopment, but only a few neuroimaging studies have been performed to date, mostly in neonates. In this population-based cohort study, we investigated the association between MIA and multiple neuroimaging modalities depicting brain development from childhood to adolescence. METHODS We used data of mother-child pairs from the Generation R Study. To define our exposure, we measured interleukin (IL) 1β, IL-6, IL-17a, IL-23, interferon gamma, and C-reactive protein at 2 time points during pregnancy. Because levels of these 5 cytokines were highly correlated, we were able to compute a cytokine index. We used multiple brain imaging modalities as outcomes, including global and regional measures of brain morphology (structural magnetic resonance imaging, volume; n = 3295), white matter microstructure (diffusion magnetic resonance imaging, fractional anisotropy and mean diffusivity; n = 3267), and functional connectivity (functional magnetic resonance imaging, graph theory measures, and network-level connectivity; n = 2914) in the children at ages 10 and 14 years. We performed mixed effects models using child's age as a continuous time variable. RESULTS We found no significant effect of time on any neuroimaging outcomes in children over time, and there was no time × MIA interaction. These associations were similar for the cytokine index, C-reactive protein, and individual cytokines. We observed no evidence for differential effects of timing of prenatal MIA or child sex after multiple testing correction. CONCLUSIONS In this longitudinal population-based study, we found no evidence for an association between MIA and child brain development in the general population. Our findings differ from previous research in neonates that have shown structural and functional brain abnormalities after MIA.
Collapse
Affiliation(s)
- Anna Suleri
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands; Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, National Institute of Mental Health, Bethesda, Maryland
| | - Lot de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Frederieke Gigase
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Biomedical Data Sciences, Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent W V Jaddoe
- Department of Pediatrics, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Michael Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Manon H J Hillegers
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
27
|
Xu G, Tian C, Li Y, Fang L, Wang J, Jing Z, Li S, Chen P. Inhibition of BCAT1 expression improves recurrent miscarriage by regulating cellular dysfunction and inflammation of trophoblasts. Cell Tissue Res 2024; 398:111-121. [PMID: 39356334 DOI: 10.1007/s00441-024-03921-7] [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: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Sustained or chronic inflammation in the placenta can result in placental insufficiency, leading to adverse reproductive outcomes such as pregnancy loss. Branched-chain amino acid transaminase 1 (BCAT1) expresses in the placenta and is involved in the pathological inflammatory response, but its role in recurrent miscarriage (RM) has not been fully investigated. In the present study, we delved into the effects of BCAT1 on trophoblast inflammation induced by lipopolysaccharide (LPS) and a mouse model of pregnancy loss induced by LPS. In vitro, after the HTR-8/SVneo cells were treated with LPS and BCATc inhibitor 2 (a selective BCAT inhibitor), the cell apoptosis was verified by TUNEL assay, and the activity of caspase-3 and caspase-9 was detected. Real-time PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence (IF) were used to determine the expression of inflammatory cytokines (TNF-α, IL-6, and IL-1β) and inflammasomes (NLRP3 and ASC) in LPS-treated trophoblast cells. Western blot analysis was performed to verify the expression of phospho-IκBα (p-IκBα) in cells and NF-κB p65 in the nuclei. IF staining was used to detect the nuclear translocation of NF-κB p65. The DNA binding activity of NF-κB was detected by an electrophoretic mobility shift assay (EMSA). The results demonstrated that inhibition of BCAT1 reduced trophoblast apoptosis, suppressed the release of proinflammatory cytokines, and prevented NLRP3 inflammasome activation in response to LPS. Additionally, BCAT1 inhibition blocked the activation of the NF-κB pathway in trophoblasts. This study highlights the potential therapeutic role of targeting BCAT1 in preventing adverse reproductive outcomes associated with chronic placental inflammation.
Collapse
Affiliation(s)
- Guangli Xu
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| | - Chao Tian
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Yanru Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Lei Fang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Jing Wang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Zhuqing Jing
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Simeng Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Ping Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| |
Collapse
|
28
|
Yount CS, Scheible K, Thurston SW, Qiu X, Ge Y, Hopke PK, Lin Y, Miller RK, Murphy SK, Brunner J, Barrett E, O'Connor TG, Zhang J, Rich DQ. Short term air pollution exposure during pregnancy and associations with maternal immune markers. ENVIRONMENTAL RESEARCH 2024; 260:119639. [PMID: 39034020 PMCID: PMC11421383 DOI: 10.1016/j.envres.2024.119639] [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: 04/02/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Air pollution exposure during pregnancy has been associated with numerous adverse pregnancy, birth, and child health outcomes. One proposed mechanism underlying these associations is maternal immune activation and dysregulation. We examined associations between PM2.5 and NO2 exposure during pregnancy and immune markers within immune function groups (TH1, TH2, TH17, Innate/Early Activation, Regulatory, Homeostatic, and Proinflammatory), and examined whether those associations changed across pregnancy. METHODS In a pregnancy cohort study (n = 290) in Rochester, New York, we measured immune markers (using Luminex) in maternal plasma up to 3 times during pregnancy. We estimated ambient PM2.5 and NO2 concentrations at participants' home addresses using a spatial-temporal model. Using mixed effects models, we estimated changes in immune marker concentrations associated with interquartile range increases in PM2.5 (2.88 μg/m3) and NO2 (7.82 ppb) 0-6 days before blood collection, and assessed whether associations were different in early, mid, and late pregnancy. RESULTS Increased NO2 concentrations were associated with higher maternal immune markers, with associations observed across TH1, TH2, TH17, Regulatory, and Homeostatic groups of immune markers. Furthermore, the largest increases in immune markers associated with each 7.82 ppb increase in NO2 concentration were in late pregnancy (e.g., IL-23 = 0.26 pg/ml, 95% CI = 0.07, 0.46) compared to early pregnancy (e.g., IL-23 = 0.08 pg/ml, 95% CI = -0.11, 0.26). CONCLUSIONS Results were suggestive of NO2-related immune activation. Increases in effect sizes from early to mid to late pregnancy may be due to changes in immune function over the course of pregnancy. These findings provide a basis for immune activation as a mechanism for previously observed associations between air pollution exposure during pregnancy and reduced birthweight, fetal growth restriction, and pregnancy complications.
Collapse
Affiliation(s)
- C S Yount
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - K Scheible
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - S W Thurston
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - X Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Y Ge
- Nicholas School of the Environment & Duke Global Health Institute, Duke University, Durham, NC, USA
| | - P K Hopke
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA; Center for Air and Aquatic Resources Engineering and Sciences, Clarkson University, Potsdam, NY, USA
| | - Y Lin
- Nicholas School of the Environment & Duke Global Health Institute, Duke University, Durham, NC, USA
| | - R K Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - S K Murphy
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - J Brunner
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - E Barrett
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA; Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA; Department of Biostatistics and Epidemiology, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - T G O'Connor
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA; Department of Psychology, University of Rochester, Rochester, NY, USA; Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - J Zhang
- Nicholas School of the Environment & Duke Global Health Institute, Duke University, Durham, NC, USA
| | - D Q Rich
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
29
|
Nowak AL, Saadat N, Sun J, Forsman AM, Liang X, Joyce C, Woo J, Engeland CG, Misra DP, Giurgescu C, Zhang W, Anderson CM. Preterm Birth in African American Women: A Multi-Omic Pilot Study in Early Pregnancy. Biol Res Nurs 2024:10998004241275049. [PMID: 39440846 DOI: 10.1177/10998004241275049] [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: 10/25/2024]
Abstract
Preterm birth (PTB; <37 weeks completed gestation) is a devastating problem affecting over 13 million live births worldwide. In the U.S., African Americans experience significantly higher rates of PTB compared to non-Hispanic Whites. PTB disparities have been linked to social determinants of health (e.g., socioeconomic status, discrimination). However, the biological underpinnings related to these associations are unclear. DNA methylation (DNAm) is subject to environmental influences, and DNAm modifications are known to affect gene expression. Using a multi-omic approach, we examined differences in combined DNA methylation (DNAm) and messenger RNA (mRNA) transcriptomic data from 20 pregnant African American women (12 PTB; 8 term birth) early in pregnancy (8-18 weeks gestation). We found that the HLA-DQB2 gene was both differentially methylated (cg12296550; p = .02) and differentially expressed (p = .014; log2FC = 2.5) between women with PTB and term birth. Gene expression analysis showed HLA-DQB2 and HLA-DRB4 (p = .028; log2FC = -3.6) were the two most highly expressed genes. HLA-DQB2 expressed higher in PTB and HLA-DRB4 expressed higher in term birth. However, no genes remained significant (p < .05) after Bonferroni correction. HLA-DRB4 and AKR1C1 were identified as a potential biomarkers in dimensionality reduction models and are also important to immune function and allogenic breakdown. Altered gene expression may lead to inflammatory imbalances or allogenic intolerance resulting in PTB. This study provides proof-of-concept evidence for the feasibility and importance of future multi-omics studies with larger populations to further explore the genes and pathways identified here.
Collapse
Affiliation(s)
- Alexandra L Nowak
- Marcella Niehoff School of Nursing, Loyola University at Chicago, Maywood, IL, USA
| | - Nadia Saadat
- Department of Paediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Jiao Sun
- Department of Computer Science, University of Central Florida, Orlando, FL, USA
| | - Anna M Forsman
- Department of Biology, Colby College, Waterville, ME, USA
| | - Xiaoyu Liang
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Cara Joyce
- Biostatistics Core, Department of Medicine, Center for Translational Research and Education, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Jennifer Woo
- University of Texas at Arlington, Arlington, TX, USA
| | - Christopher G Engeland
- Biobehavioral Health, College of Health and Human Development, Ross and Carol Ness College of Nursing, The Pennsylvania State University, University Park, PA, USA
| | - Dawn P Misra
- Department of Epidemiology and Biostatistics, MSU College of Human Medicine, East Lansing, MI, USA
| | - Carmen Giurgescu
- Chatlos Foundation Endowed Chair in Nursing, University of Central Florida College of Nursing, Orlando, FL, USA
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, Orlando, FL, USA
| | - Cindy M Anderson
- Maternal Infant Health, Martha S. Pitzer Center for Women, Children and Youth, The Ohio State University College of Nursing, Columbus, OH, USA
| |
Collapse
|
30
|
Dong S, Fu C, Shu C, Xie M, Li Y, Zou J, Meng YZ, Xu P, Shan YH, Tian HM, He J, Yang YG, Hu Z. Development of a humanized mouse model with functional human materno-fetal interface immunity. JCI Insight 2024; 9:e176527. [PMID: 39435662 PMCID: PMC11529984 DOI: 10.1172/jci.insight.176527] [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: 10/18/2023] [Accepted: 09/04/2024] [Indexed: 10/23/2024] Open
Abstract
Materno-fetal immunity possesses specialized characteristics to ensure pathogen clearance while maintaining tolerance to the semiallogeneic fetus. Most of our understanding on human materno-fetal immunity is based on conventional rodent models that may not precisely represent human immunological processes owing to the huge evolutionary divergence. Herein, we developed a pregnant human immune system (HIS) mouse model through busulfan preconditioning, which hosts multilineage human immune subset reconstitution at the materno-fetal interface. Human materno-fetal immunity exhibits a tolerogenic feature at the midgestation stage (embryonic day [E] 14.5), and human immune regulatory subsets were detected in the decidua. However, the immune system switches to an inflammatory profile at the late gestation stage (E19). A cell-cell interaction network contributing to the alternations in the human materno-fetal immune atmosphere was revealed based on single-cell RNA-Seq analysis, wherein human macrophages played crucial roles by secreting several immune regulatory mediators. Furthermore, depletion of Treg cells at E2.5 and E5.5 resulted in severe inflammation and fetus rejection. Collectively, these results demonstrate that the pregnant HIS mouse model permits the development of functional human materno-fetal immunity and offers a tool for human materno-fetal immunity investigation to facilitate drug discovery for reproductive disorders.
Collapse
Affiliation(s)
- Shuai Dong
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Chang Shu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
| | - Min Xie
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yan Li
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Jun Zou
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yi-Zi Meng
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Peng Xu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yan-Hong Shan
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
| | - Hui-Min Tian
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Jin He
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| |
Collapse
|
31
|
Dudley JS, Renfree MB, Wagner GP, Griffith OW. The extension of mammalian pregnancy required taming inflammation: Independent evolution of extended placentation in the tammar wallaby. Proc Natl Acad Sci U S A 2024; 121:e2310047121. [PMID: 39378090 PMCID: PMC11494332 DOI: 10.1073/pnas.2310047121] [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: 07/03/2023] [Accepted: 08/24/2024] [Indexed: 10/10/2024] Open
Abstract
In the first live-bearing mammals, pregnancy was likely short and ended with a brief period of inflammatory maternal-fetal interaction. This mode of reproduction has been retained in many marsupials. While inflammation is key to successful implantation in eutherians, a key innovation in eutherians is the ability to switch off this inflammation after it has been initiated. This extended period, in which inflammation is suppressed, likely allowed for an extended period of placentation. Extended placentation has evolved independently in one lineage of marsupials, the macropodids (wallabies and kangaroos), with placentation lasting beyond the 2 to 4 d seen in other marsupial taxa, which allows us to investigate the role of inflammation response after attachment in the extension of placentation in mammals. By comparing gene expression changes at attachment in three marsupial species, the tammar wallaby, opossum, and fat-tailed dunnart, we show that inflammatory attachment is an ancestral feature of marsupial implantation. In contrast to eutherians, where attachment-related (quasi-) inflammatory reaction is even involved in epitheliochorial placentation (e.g., pig), this study found no evidence of a distinct attachment-related reaction in wallabies. Instead, only a small number of inflammatory genes are expressed at distinct points of gestation, including IL6 before attachment, LIF throughout placentation, and prostaglandins before birth. During parturition, a more distinct inflammatory reaction is detectable, likely involved in precipitating the parturition cascade similar to eutherians. We suggest that in wallaby, extended gestation became possible by avoiding an inflammatory attachment reaction, which is a different strategy than seen in eutherians.
Collapse
Affiliation(s)
- Jessica S. Dudley
- Department of Biological Sciences, School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, NSW2109, Australia
| | - Marilyn B. Renfree
- School of BioSciences, University of Melbourne, Melbourne, VIC3010, Australia
| | - Günter P. Wagner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT06520
- Yale Systems Biology Institute, Yale University, West Haven, CT06520
- Department of Evolutionary Biology, University of Vienna, ViennaA-1030, Austria
| | - Oliver W. Griffith
- Department of Biological Sciences, School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, NSW2109, Australia
| |
Collapse
|
32
|
Park JS, Kim E, Kwon SJ, Heo JS, Ahn KH. Differences in maternal subgingival microbiome between preterm and term births: The MOHEPI study. J Periodontal Res 2024; 59:939-950. [PMID: 38808521 DOI: 10.1111/jre.13292] [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: 12/23/2023] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
AIM Periodontitis is a potential risk factor for preterm birth (PTB) in women; however, the causal relationship or the exact mechanism remain unknown. This study aimed to compare the oral microbiome features of mothers with full-term birth (FTB) with those who had preterm delivery. METHODS This study prospectively enrolled 60 women (30 mothers with PTB and 30 mothers with FTB), and subgingival plaque samples were collected and analysed by metagenomic 16S rDNA sequencing. Clinical measurements, including periodontal probing depth, clinical attachment level, modified gingival index (mGI) and plaque index, were performed to determine the periodontal state of the participants. Medical and obstetric data were collected as well. RESULTS Among the periodontal measurements, mGI score, reflecting the level of gingival inflammation, exhibited a statistically significant association with PTB (adjusted odds ratio 2.705, 95% confidence interval 1.074-6.811, p = .035). When subgroup analysis was conducted based on mean mGI scores (mGI ≥ 2, high inflammation [HI] versus mGI < 2, low inflammation [LI]), microbiome analysis revealed clear distinctions in microbial compositions between PTB and FTB mothers in both the HI and LI groups. Especially in the HI group, alpha diversity exhibited a decreasing trend in PTB mothers compared to FTB mothers. Beta diversity also revealed significant differences between the two groups. In Linear Discriminant Analysis Effect Size analysis, certain anaerobic taxa, including the genera Spirochaetes, Treponema and Porphyromonas, were relatively abundant in the FTB/HI group, whereas the PTB/HI group showed a high abundance of the order Actinomycetales. Network analysis showed that the FTB/HI had relatively stronger connectivity in microbial composition than the PTB/HI group. Dysbiosis ratio of plaque microbiome, in terms of periodontitis, was significantly lower in PTB/HI group compared to FTB/HI group. CONCLUSION The compositions of maternal subgingival microbiomes differed between PTB and FTB mothers in both the high and low levels of gingival inflammation groups. In the presence of high level of gingival inflammation, dysbiosis in plaque microbiome, in terms of periodontitis, was decreased in PTB mothers compared to FTB mothers.
Collapse
Affiliation(s)
- Jung Soo Park
- Department of Periodontology, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Eunha Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | | | - Ju Sun Heo
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ki Hoon Ahn
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Singh S, Goel I, Tripathi S, Ahirwar A, Kumar M, Rana A, Dhar R, Karmakar S. Effect of environmental air pollutants on placental function and pregnancy outcomes: a molecular insight. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:59819-59851. [PMID: 39388084 DOI: 10.1007/s11356-024-35016-9] [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: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024]
Abstract
Air pollution has become a major health concern, particularly for vulnerable populations such as the elderly, children, and pregnant women. Studies have reported a strong association between prenatal exposure to air pollutants and adverse pregnancy outcomes, including lower birth weight, reduced fetal growth, and an increased frequency of preterm births. This review summarizes the harmful effects of air pollutants, such as particulate matter, on pregnancy and outlines the mechanistic details associated with these adverse outcomes. Particulate pollutant matter may be able to cross the placenta barrier, and alterations in placental functions are central to the detrimental effects of these pollutants. In addition to associations with preeclampsia and gestational hypertension, air pollutants also induce oxidative stress, inflammation, and epigenetic alteration in the placenta. These pollutants can also affect placental homeostasis and endocrine function, contributing to pregnancy complications and possible transgenerational effects. Prenatal air pollution exposure has been linked to reduced cognitive and motor function in infants and newborns, increasing the predisposition to autism spectrum disorders and other neuropsychiatric disorders. This review also summarizes the use of various animal models to study the harmful effects of air pollution on pregnancy and postnatal outcomes. These findings provide valuable insight into the molecular events associated with the process and can aid in risk mitigation and adopting safety measures. Implementing effective environmental protocols and taking appropriate steps may reduce the global disease burden, particularly for developing nations with poor regulatory compliance and large populations of pregnant women.
Collapse
Affiliation(s)
- Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Room 3020, New Delhi, 110029, India
| | - Isha Goel
- Department of Psychiatry, All India Institute of Medical Sciences, New Delhi, India
| | - Smita Tripathi
- Department of Biochemistry, Lady Harding Medical College, New Delhi, India
| | - Ashok Ahirwar
- Department of Lab Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Megha Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Habsiguda, Hyderabad, India
| | - Anubhuti Rana
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, Room 3020, New Delhi, 110029, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, Room 3020, New Delhi, 110029, India.
| |
Collapse
|
34
|
Zhou J, Teng Y, Ouyang J, Wu P, Tong J, Gao G, Yan S, Tao F, Huang K. Associations of Placental Inflammation and Oxidative Stress Biomarkers with Glucolipid Metabolism in Children: A Birth Cohort Study in China. J Am Heart Assoc 2024; 13:e035754. [PMID: 39206740 PMCID: PMC11646502 DOI: 10.1161/jaha.124.035754] [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: 03/26/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The maternal intrauterine immune environment may affect offspring long-term health. We aimed to investigate the association between the intrauterine placental immunological milieu and glycolipid metabolic health in children. METHODS AND RESULTS This study enrolled 1803 mother-child pairs from the Ma'anshan birth cohort (2013-2014). Placental mRNA expression of inflammatory cytokines (interleukin-1β [IL-1β], IL-10, monocyte chemoattractant protein-1, tumor necrosis factor-α, IL-4, IL-6, IL-8, C-reactive protein, and interferon-γ) and oxidative stress biomarkers (heme oxygenase-1, hypoxia-inducible factor-1alpha, and glucose-related protein 78) was quantified using real-time quantitative polymerase chain reaction. Fasting blood glucose, insulin, triglycerides, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and total cholesterol were assessed at 5 to 6 years old. Statistical analyses included multiple linear regression, binary logistic regression, restricted cubic spline model, and the Bayesian kernel machine regression model. Placental inflammatory cytokines (IL-1β, monocyte chemoattractant protein-1, C-reactive protein, IL-6, IL-8, IL-10) and oxidative stress biomarkers (heme oxygenase-1, hypoxia-inducible factor-1alpha, glucose-related protein 78) showed positive associations with children's fasting blood glucose levels. Heme oxygenase-1 and glucose-related protein 78 exhibited negative correlations with children's fasting insulin levels. Elevated IL-6, heme oxygenase-1, hypoxia-inducible factor-1alpha, and glucose-related protein 78 were associated with increased risk of prediabetes in children. Overall upregulation of placental proinflammatory cytokines and oxidative stress factors mRNA expression correlated with higher prediabetes risk in children. Bayesian kernel machine regression analysis indicated a joint positive effect of the 12 placental inflammation and oxidative stress mixtures on children's risk of high fasting blood glucose. CONCLUSIONS This exploratory study underscores significant correlations between maternal intrauterine placental inflammation, oxidative stress markers, and offspring fasting blood glucose and insulin levels. These findings highlight the potential role of intrauterine holistic immunity in shaping offspring glucose metabolism health.
Collapse
Affiliation(s)
- Jixing Zhou
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Yuzhu Teng
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Jiajun Ouyang
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Penggui Wu
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Guopeng Gao
- Maternal and Child Health Care Center of Ma’anshanAnhuiChina
| | - Shuangqin Yan
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Maternal and Child Health Care Center of Ma’anshanAnhuiChina
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public HealthAnhui Medical UniversityHefeiChina
- Key Laboratory of Population Health Across Life Cycle (AHMU), MOEHefeiChina
- NHC Key Laboratory of study on abnormal gametes and reproductive tractHefeiChina
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life CourseHefeiChina
| |
Collapse
|
35
|
Gao J, Jiang N, Chen Q, Zhao M, Tang Y. Systemic Immune-Inflammation Indices Could Be Additional Predictive Markers for Cesarean Scar Pregnancy. Am J Reprod Immunol 2024; 92:e13924. [PMID: 39221973 DOI: 10.1111/aji.13924] [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: 04/25/2024] [Revised: 06/20/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
PROBLEM Cesarean scar pregnancy (CSP) is characterized by a gestational sac fully or partially implanted in the scar from a previous cesarean section. Systemic immune-inflammation indices (SIIs) have recently been discussed as additional diagnostic markers in placenta accreta and preeclampsia. CSP shares a similar pathogenesis with these diseases, suggesting that assessing the SIIs and neutrophil-to-lymphocyte ratio (NLR) could enhance additional predictability in diagnosing CSP. METHOD OF STUDY In this study, we analyzed the complete blood counts between 264 women who were confirmed with CSP by ultrasound and 295 women who underwent elective termination. RESULTS The mean counts of total white cells and neutrophils were significantly higher, whereas the counts of monocytes, lymphocytes, and platelets were significantly lower in the CSP group compared to the control group (p < 0.001). Additionally, the SII, systemic inflammation response index (SIRI), or NLR was significantly higher in the CSP group compared to the control group (p < 0.0001). Given the limited effect of SII and SIRI on the increased risk of developing CSP, the optimal cut-off value for NLR in predicting CSP was 2.87 (area under the curve [AUC] 0.656, 68% sensitivity). The optimal cut-off value for NLR in predicting type 2 CSP was 2.91 (AUC 0.690, 71% sensitivity). CONCLUSIONS Although ultrasound or magnetic resonance imaging images are a gold standard for visualizing the gestational sac's location in the diagnosis of CSP, assessing peripheral blood tests is cost-effective, and NLR may provide additional diagnosis value for CSP.
Collapse
Affiliation(s)
- Jing Gao
- Department of Medical Laboratory, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Nanyan Jiang
- Department of Medical Laboratory, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Qi Chen
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Min Zhao
- Department of Gynaecology, Wuxi Maternity and Child Health Hospital, Jiangnan University, Wuxi, China
| | - Yunhui Tang
- Department of Family Planning, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
36
|
Peng D, Zhong W, Wang Y, Fu Y, Shang W. The Causal Relationship Between Immune Cells and Infertility: A Mendelian Randomisation Study. Am J Reprod Immunol 2024; 92:e13932. [PMID: 39320012 DOI: 10.1111/aji.13932] [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: 04/06/2024] [Revised: 07/20/2024] [Accepted: 09/04/2024] [Indexed: 09/26/2024] Open
Abstract
OBJECTIVE Infertility has emerged as a significant global public health concern, with a multitude of complex underlying causes. Epidemiological evidence indicates that immunological factors are significant contributors to the aetiology of infertility. However, previous studies on the relationship between immune inflammation and infertility have yielded inconclusive results. METHODS Mendelian randomisation (MR) is an emerging statistical method that employs exposure-related genetic variation as an instrumental variable (IV) to infer causal relationships between immune cells and infertility by modelling the principle of random assignment in Mendelian genetics. In this study, MR was employed to assess the causal relationship between 731 immune cell signatures and infertility. The data utilized in this study were obtained from publicly available genome-wide association studies (GWAS) and validated IVs, which were employed to fulfil the essential assumptions of MR analysis. RESULTS The Mendelian randomisation analysis revealed a total of 27 statistically significant immune cell phenotypes out of 731. The risk factor with the largest odds ratio (OR) was CD28- CD25++ CD8+ %T cell [OR, 1.21; 95% confidence interval (CI), 1.04-1.42], while the protective factor with the largest OR was activated and resting Treg AC (OR, 0.89; 95% CI, 0.82-0.97). CONCLUSION The present study has demonstrated a correlation between certain characteristics of immune cells and female infertility. These results provide clues for further research into the immune mechanisms of infertility and may inform the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Dingchuan Peng
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Wei Zhong
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yiran Wang
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yiyao Fu
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Wei Shang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
37
|
Naidoo SJ, Naicker T. The Enigmatic Interplay of Interleukin-10 in the Synergy of HIV Infection Comorbid with Preeclampsia. Int J Mol Sci 2024; 25:9434. [PMID: 39273381 PMCID: PMC11395227 DOI: 10.3390/ijms25179434] [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: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Cytokines coordinate the intricate choreography of the immune system, directing cellular activities that mediate inflammation, pathogen defense, pathology and tissue repair. Within this spectrum, the anti-inflammatory prowess of interleukin-10 (IL-10) predominates in immune homeostasis. In normal pregnancy, the dynamic shift of IL-10 across trimesters maintains maternal immune tolerance ensuring fetal development and pregnancy success. Unravelling the dysregulation of IL-10 in pregnancy complications is vital, particularly in the heightened inflammatory condition of preeclampsia. Of note, a reduction in IL-10 levels contributes to endothelial dysfunction. In human immunodeficiency virus (HIV) infection, a complex interplay of IL-10 occurs, displaying a paradoxical paradigm of being immune-protective yet aiding viral persistence. Genetic variations in the IL-10 gene further modulate susceptibility to HIV infection and preeclampsia, albeit with nuanced effects across populations. This review outlines the conceptual framework underlying the role of IL-10 in the duality of normal pregnancy and preeclampsia together with HIV infection, thus highlighting its regulatory mechanisms and genetic influences. Synthesizing these findings in immune modulation presents avenues for therapeutic interventions in pregnancy complications comorbid with HIV infection.
Collapse
Affiliation(s)
| | - Thajasvarie Naicker
- Department of Optics and Imaging, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
38
|
Jia R, Ma H, Hao H, Wang F, Yang H. Metformin inhibits activation of NLRP3 inflammasome and inflammatory response in preeclamptic rats. Gene 2024; 919:148509. [PMID: 38677349 DOI: 10.1016/j.gene.2024.148509] [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: 01/15/2024] [Revised: 04/02/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUD It is widely acknowledged that Metformin (MET), an established medication for managing type 2 diabetes, possesses diverse pharmacological effects. This study aims to investigate the protective effects of MET against Nω-Nitro-L-arginine methyl ester (L-NAME)-induced preeclampsia (PE). METHODS Sprague Dawley (SD) rats were exposed to 200 mg/kg L-NAME with or without prior MET treatment. Histopathological analysis was performed using Hematoxylin and Eosin staining. Serum levels of inflammatory, antiangiogenic, and angiogenic factors were quantified using ELISA kits. Immunohistochemistry (IHC) staining was employed to observe NLRP3 and IL-1β expressions in placental tissues. Western blot and Quantitative Real-Time PCR (q-PCR) analyses were conducted to assess protein and mRNA expressions of NLRP3, caspase-1, ASC, and IL-1β. RESULTS We found that MET could mitigate placental histopathological deterioration and improve pregnancy outcomes in L-NAME-induced PE rat models. MET not only suppressed L-NAME-induced elevation of antiangiogenic factors but also stimulated the production of pro-angiogenic factors. Additionally, MET treatment reversed the excessive inflammatory response induced by L-NAME. Furthermore, MET inhibited the activation of the NLRP3 inflammasome triggered by L-NAME, evidenced by the downregulation of NLRP3 expression, caspase-1, and IL-1β. CONCLUSIONS MET demonstrates a protective effect against L-NAME-induced PE rats, potentially mediated through inhibition of the inflammatory response, downregulation of NLRP3 inflammasome expression in the placenta, and regulation of the balance between anti-angiogenic and pro-angiogenic factors.
Collapse
Affiliation(s)
- Ran Jia
- Department of Gynecology and Obstetrics, the First Hospital of Shanxi Medical University (First Clinical Medicine College), Taiyuan City, Shanxi Province 030001, China
| | - Huijing Ma
- Department of Gynecology and Obstetrics, the First Hospital of Shanxi Medical University (First Clinical Medicine College), Taiyuan City, Shanxi Province 030001, China
| | - Huiniu Hao
- Department of Gynecology and Obstetrics, the First Hospital of Shanxi Medical University (First Clinical Medicine College), Taiyuan City, Shanxi Province 030001, China
| | - Fang Wang
- Department of Gynecology and Obstetrics, the First Hospital of Shanxi Medical University (First Clinical Medicine College), Taiyuan City, Shanxi Province 030001, China
| | - Hailan Yang
- Department of Gynecology and Obstetrics, the First Hospital of Shanxi Medical University (First Clinical Medicine College), Taiyuan City, Shanxi Province 030001, China.
| |
Collapse
|
39
|
Einenkel R, Ehrhardt J, Zygmunt M, Muzzio DO. Less is more! Low amount of Fusobacterium nucleatum supports macrophage-mediated trophoblast functions in vitro. Front Immunol 2024; 15:1447190. [PMID: 39176096 PMCID: PMC11338817 DOI: 10.3389/fimmu.2024.1447190] [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: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
F. nucleatum, involved in carcinogenesis of colon carcinomas, has been described as part of the commensal flora of the female upper reproductive tract. Although its contribution to destructive inflammatory processes is well described, its role as commensal uterine bacteria has not been thoroughly investigated. Since carcinogenesis shares similar mechanisms with early pregnancy development (including proliferation, invasion, blood supply and the induction of tolerance), these mechanisms induced by F. nucleatum could play a role in early pregnancy. Additionally, implantation and placentation require a well-balanced immune activation, which might be suitably managed by the presence of a limited amount of bacteria or bacterial residues. We assessed the effect of inactivated F. nucleatum on macrophage-trophoblast interactions. Monocytic cells (THP-1) were polarized into M1, M2a or M2c macrophages by IFN-γ, IL-4 or TGF-β, respectively, and subsequently treated with inactivated fusobacteria (bacteria:macrophage ratio of 0.1 and 1). Direct effects on macrophages were assessed by viability assay, flow cytometry (antigen presentation molecules and cytokines), qPCR (cytokine expression), in-cell Western (HIF and P-NF-κB) and ELISA (VEGF secretion). The function of first trimester extravillous trophoblast cells (HTR-8/SVneo) in response to macrophage-conditioned medium was microscopically assessed by migration (scratch assay), invasion (sprouting assay) and tube formation. Underlying molecular changes were investigated by ELISA (VEGF secretion) and qPCR (matrix-degrading factors and regulators). Inflammation-primed macrophages (M1) as well as high bacterial amounts increased pro-inflammatory NF-κB expression and inflammatory responses. Subsequently, trophoblast functions were impaired. In contrast, low bacterial stimulation caused an increased HIF activation and subsequent VEGF-A secretion in M2c macrophages. Accordingly, there was an increase of trophoblast tube formation. Our results suggest that a low-mass endometrial/decidual microbiome can be tolerated and while it supports implantation and further pregnancy processes.
Collapse
Affiliation(s)
| | | | | | - Damián Oscar Muzzio
- Department of Gynecology and Obstetrics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
40
|
Zhu AZ, Ma Z, Wolff EV, Lin Z, Gao ZJ, Li X, Du W. HES1 is required for mouse fetal hematopoiesis. Stem Cell Res Ther 2024; 15:235. [PMID: 39075526 PMCID: PMC11287931 DOI: 10.1186/s13287-024-03836-8] [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: 05/14/2024] [Accepted: 07/06/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Hematopoiesis in mammal is a complex and highly regulated process in which hematopoietic stem cells (HSCs) give rise to all types of differentiated blood cells. Previous studies have shown that hairy and enhancer of split (HES) repressors are essential regulators of adult HSC development downstream of Notch signaling. METHODS In this study, we investigated the role of HES1, a member of HES family, in fetal hematopoiesis using an embryonic hematopoietic specific Hes1 conditional knockout mouse model by using phenotypic flow cytometry, histopathology analysis, and functional in vitro colony forming unit (CFU) assay and in vivo bone marrow transplant (BMT) assay. RESULTS We found that loss of Hes1 in early embryonic stage leads to smaller embryos and fetal livers, decreases hematopoietic stem progenitor cell (HSPC) pool, results in defective multi-lineage differentiation. Functionally, fetal hematopoietic cells deficient for Hes1 exhibit reduced in vitro progenitor activity and compromised in vivo repopulation capacity in the transplanted recipients. Further analysis shows that fetal hematopoiesis defects in Hes1fl/flFlt3Cre embryos are resulted from decreased proliferation and elevated apoptosis, associated with de-repressed HES1 targets, p27 and PTEN in Hes1-KO fetal HSPCs. Finally, pharmacological inhibition of p27 or PTEN improves fetal HSPCs function both in vitro and in vivo. CONCLUSION Together, our findings reveal a previously unappreciated role for HES1 in regulating fetal hematopoiesis, and provide new insight into the differences between fetal and adult HSC maintenance.
Collapse
Affiliation(s)
- Anthony Z Zhu
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Zhilin Ma
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Emily V Wolff
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Zichen Lin
- Master of Science in Medical Science, Boston University School of Medicine Graduate Master Program, Boston, MA, USA
| | - Zhenxia J Gao
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Xue Li
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Wei Du
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
41
|
Chico-Barba G, Sámano R, Martínez-Rojano H, Morales-Hernández RM, Barrientos-Galeana E, Luna-Hidalgo A, Kaufer-Horwitz M, Obrador GT, Villa-Romero AR. Total Gestational Weight Gain Is Explained by Leptin and Body Fat, Regardless of Pre-Pregnancy Body Mass Index and Other Adipokines, in Mexican Adolescents. Nutrients 2024; 16:2147. [PMID: 38999894 PMCID: PMC11242962 DOI: 10.3390/nu16132147] [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: 06/12/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Pre-pregnancy body mass index (pBMI) is a predictor of gestational weight gain (GWG). However, other factors, such as adipokines and inflammation markers, may also be associated with GWG. The aim of the study was to determine the association of leptin, adiponectin, irisin, and C-reactive protein, with GWG in adolescents. A longitudinal study was conducted from 2018 to 2023 in adolescents with a clinically healthy pregnancy. The assessments included sociodemographic and clinical data, pBMI, percent of body fat, serum concentrations of leptin, adiponectin, irisin, and high-sensitivity C-reactive protein (hsCRP), and total GWG adequacy. Cox regression models were performed, the outcome variables were inadequate and excessive GWG. In 198 participants, being overweight/obesity was marginally associated with a protective effect against inadequate GWG (HR = 0.44, 95%CI = 0.18-1.06), regardless of maternal characteristics and adipokines. Leptin (HR = 1.014, 95%CI = 1.008-1.021), and body fat percent (HR = 1.11, 95%CI = 1.05-1.17) were associated with a higher risk of excessive GWG, independent of other maternal variables such as pBMI, while adiponectin was associated with a lower risk. These findings suggest that, in Mexican adolescents, adipose tissue and its adipokines during pregnancy may play a more significant role in the final GWG than body weight.
Collapse
Affiliation(s)
- Gabriela Chico-Barba
- Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Reyna Sámano
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Hugo Martínez-Rojano
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | | | - Edgar Barrientos-Galeana
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Andrea Luna-Hidalgo
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Martha Kaufer-Horwitz
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | | | - Antonio Rafael Villa-Romero
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
42
|
Borowczak J, Gąsiorek-Kwiatkowska A, Szczerbowski K, Maniewski M, Zdrenka M, Szadurska-Noga M, Gostomczyk K, Rutkiewicz P, Olejnik K, Cnota W, Karpów-Greiner M, Knypiński W, Sekielska-Domanowska M, Ludwikowski G, Dubiel M, Szylberg Ł, Bodnar M. SARS-CoV-2 Infection during Delivery Causes Histopathological Changes in the Placenta. Diseases 2024; 12:142. [PMID: 39057113 PMCID: PMC11276080 DOI: 10.3390/diseases12070142] [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: 05/29/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND SARS-CoV-2 can damage human placentas, leading to pregnancy complications, such as preeclampsia and premature birth. This study investigates the histopathological changes found in COVID-19-affected placentas. MATERIALS AND METHODS This study included 23 placentas from patients with active COVID-19 during delivery and 22 samples from patients without COVID-19 infection in their medical history. The samples underwent histopathological examination for pathology, such as trophoblast necrosis, signs of vessel damage, or fetal vascular malperfusion. RESULTS Newborns from the research group have lower weights and Apgar scores than healthy newborns. In the COVID-19 group, calcifications and collapsed intervillous space were more frequent, and inflammation was more severe than in the healthy group. At the same time, the placenta of SARS-CoV-2-positive patients showed signs of accelerated vascular maturation. Trophoblast necrosis was found only in the placentas of the research group. The expression of CD68+ was elevated in the COVID-19 cohort, suggesting that macrophages constituted a significant part of the inflammatory infiltrate. The increase in lymphocyte B markers was associated with placental infarctions, while high levels of CD3+, specific for cytotoxic T lymphocytes, correlated with vascular injury. CONCLUSIONS SARS-CoV-2 is associated with pathological changes in the placenta, including trophoblast necrosis, calcification, and accelerated villous maturation. Those changes appear to be driven by T cells and macrophages, whose increased expression reflects ongoing histiocytic intervillositis in the placenta.
Collapse
Affiliation(s)
- Jędrzej Borowczak
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Agnieszka Gąsiorek-Kwiatkowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Krzysztof Szczerbowski
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Mateusz Maniewski
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Doctoral School of Medical and Health Sciences, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland
| | - Marek Zdrenka
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Marta Szadurska-Noga
- Department of Pathomorphology and Forensic Medicine, Faculty of Medical Sciences, University of Warmia and Mazury, 10-561 Olsztyn, Poland;
| | - Karol Gostomczyk
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Paula Rutkiewicz
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Katarzyna Olejnik
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Wojciech Cnota
- Chair and Department of Gynaecology and Obstetrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, 41-703 Ruda Śląska, Poland
| | - Magdalena Karpów-Greiner
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Wojciech Knypiński
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Marta Sekielska-Domanowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Grzegorz Ludwikowski
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Mariusz Dubiel
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Łukasz Szylberg
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Magdalena Bodnar
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| |
Collapse
|
43
|
Sha Q, Escobar Galvis ML, Madaj ZB, Keaton SA, Smart L, Edgerly YM, Anis E, Leach R, Osborne LM, Achtyes E, Brundin L. Dysregulated placental expression of kynurenine pathway enzymes is associated with inflammation and depression in pregnancy. Brain Behav Immun 2024; 119:146-153. [PMID: 38555986 PMCID: PMC11210184 DOI: 10.1016/j.bbi.2024.03.042] [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: 11/21/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Perinatal depression (including antenatal-, postnatal-, and depression that spans both timepoints) is a prevalent disorder with high morbidity that affects both mother and child. Even though the full biological blueprints of perinatal depression remain incomplete, multiple studies indicate that, at least for antenatal depression, the disorder has an inflammatory component likely linked to a dysregulation of the enzymatic kynurenine pathway. The production of neuroactive metabolites in this pathway, including quinolinic acid (QUIN), is upregulated in the placenta due to the multiple immunological roles of the metabolites during pregnancy. Since neuroactive metabolites produced by the pathway also may affect mood by directly affecting glutamate neurotransmission, we sought to investigate whether the placental expression of kynurenine pathway enzymes controlling QUIN production was associated with both peripheral inflammation and depressive symptoms during pregnancy. METHODS 68 placentas obtained at birth were analyzed using qPCR to determine the expression of kynurenine pathway enzymes. Cytokines and metabolites were quantified in plasma using high-sensitivity electroluminescence and ultra-performance liquid chromatography, respectively. Maternal depressive symptoms were assessed using the Edinburgh Postnatal Depression Scale (EPDS) throughout pregnancy and the post-partum. Associations between these factors were assessed using robust linear regression with ranked enzymes. RESULTS Low placental quinolinate phosphoribosyl transferase (QPRT), the enzyme responsible for degrading QUIN, was associated with higher IL-6 and higher QUIN/kynurenic acid ratios at the 3rd trimester. Moreover, women with severe depressive symptoms in the 3rd trimester had significantly lower placental expression of both QPRT and 2-amino-3-carboxymuconate-6-semialdehyde decarboxylase (ACMSD); impaired activity of these two enzymes leads to QUIN accumulation. CONCLUSION Overall, our data support that a compromised placental environment, featuring low expression of critical kynurenine pathway enzymes is associated with increased levels of plasma cytokines and the dysregulated kynurenine metabolite pattern observed in depressed women during pregnancy.
Collapse
Affiliation(s)
- Qiong Sha
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Zachary B Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Sarah A Keaton
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - LeAnn Smart
- Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| | | | - Ehraz Anis
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Richard Leach
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
| | - Lauren M Osborne
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Eric Achtyes
- Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA; Department of Psychiatry, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Lena Brundin
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
44
|
Gay L, Madariaga Zarza S, Abou Atmeh P, Rouvière MS, Andrieu J, Richaud M, Boumaza A, Miquel L, Diallo AB, Bechah Y, Otmani Idrissi M, La Scola B, Olive D, Resseguier N, Bretelle F, Mezouar S, Mege JL. Protective role of macrophages from maternal-fetal interface in unvaccinated coronavirus disease 2019 pregnant women. J Med Virol 2024; 96:e29819. [PMID: 39030992 DOI: 10.1002/jmv.29819] [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: 12/21/2023] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024]
Abstract
Pregnant women represent a high-risk population for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection. The presence of SARS-CoV-2 has been reported in placenta from infected pregnant women, but whether the virus influences placenta immune response remains unclear. We investigated the properties of maternal-fetal interface macrophages (MFMs) in a cohort of unvaccinated women who contracted coronavirus disease 2019 (COVID-19) during their pregnancy. We reported an infiltration of CD163+ macrophages in placenta from COVID-19 women 19 whereas lymphoid compartment was not affected. Isolated MFMs exhibited nonpolarized activated signature (NOS2, IDO1, IFNG, TNF, TGFB) mainly in women infected during the second trimester of pregnancy. COVID-19 during pregnancy primed MFM to produce type I and III interferon response to SARS-CoV-2 (Wuhan and δ strains), that were unable to elicit this in MFMs from healthy pregnant women. COVID-19 also primed SARS-CoV-2 internalization by MFM in an angiotensin-converting enzyme 2-dependent manner. Activation and recall responses of MFMs were influenced by fetal sex. Collectively, these findings support a role for MFMs in the local immune response to SARS-CoV-2 infection, provide a basis for protective placental immunity in COVID-19, and highlight the interest of vaccination in pregnant women.
Collapse
Affiliation(s)
- Laetitia Gay
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Sandra Madariaga Zarza
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Perla Abou Atmeh
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Marie-Sarah Rouvière
- Institut Paoli-Calmettes, UM105, Centre National de la Recherche Scientifique, Aix-Marseille University, Marseille, France
| | - Jonatane Andrieu
- Centre National de la Recherche Scientifique, Etablissement Français du Sang, Anthropologie bio-culturelle, Droit, Ethique et Santé, Aix-Marseille University, Marseille, France
| | - Manon Richaud
- Institut Paoli-Calmettes, UM105, Centre National de la Recherche Scientifique, Aix-Marseille University, Marseille, France
| | - Asma Boumaza
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Laura Miquel
- Department of Gynaecology-Obstetrics, La Conception Hospital, Marseille, France
| | - Aïssatou Bailo Diallo
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Yassina Bechah
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Myriem Otmani Idrissi
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Bernard La Scola
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Daniel Olive
- Institut Paoli-Calmettes, UM105, Centre National de la Recherche Scientifique, Aix-Marseille University, Marseille, France
| | - Noémie Resseguier
- Assistance Publique-Hôpitaux de Marseille, La Timone Hospital, Department of Epidemiology and Health Economics, Clinical Research Unit, Direction of Health Research, Aix Marseille University, Marseille, France
| | - Florence Bretelle
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
- Department of Gynaecology-Obstetrics, La Conception Hospital, Marseille, France
| | - Soraya Mezouar
- Centre National de la Recherche Scientifique, Etablissement Français du Sang, Anthropologie bio-culturelle, Droit, Ethique et Santé, Aix-Marseille University, Marseille, France
| | - Jean-Louis Mege
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
- Department of Immunology, Timone Hospital, Marseille, France
| |
Collapse
|
45
|
Cuadrado-Torroglosa I, García-Velasco JA, Alecsandru D. The Impacts of Inflammatory and Autoimmune Conditions on the Endometrium and Reproductive Outcomes. J Clin Med 2024; 13:3724. [PMID: 38999290 PMCID: PMC11242609 DOI: 10.3390/jcm13133724] [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: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/14/2024] Open
Abstract
Background: A healthy pregnancy begins with an adequate endometrial state, even before the arrival of a blastocyst. Proper endometrial priming and the development of a tolerogenic decidua are key steps in creating the perfect environment for implantation and pregnancy. In these processes, the involvement of the maternal immune system seems to be of great relevance, modulating the different decidual immune populations to prepare the endometrium for a potential pregnancy. However, certain local pathologies of an inflammatory and autoimmune nature appear to have a direct impact on these phenomena, thus altering patients' reproductive outcomes. Methods: This literature review analyzes original articles, reviews, systematic reviews, and meta-analyses published between 1990 and 2024, concerning the impact of different inflammatory and autoimmune conditions on endometrial status and fertility. The included papers were obtained from Medline (Pubmed) and the Cochrane library. Results: There is evidence that endometriosis, adenomyosis, and chronic endometritis, through the promotion of a chronic inflammatory environment, are capable of altering endometrial immune populations, and, thus, processes essential for early pregnancy. Among other effects, these conditions have been linked to impaired decidualization, alterations in progesterone responsiveness, and hindered placentation. Similarly, antiphospholipid syndrome (APS), thyroid dysfunction, diabetes, and other pathologies related to glucose and gluten metabolism, due to their autoimmune nature, also appear to have a local impact on the uterine environment, affecting reproductive success through different mechanisms, including altered hormonal response and, again, impaired decidualization. Conclusions: The management of inflammatory and autoimmune diseases in assisted reproduction patients is gaining importance due to their direct impact on the endometrium. It is necessary to follow current expert recommendations and established therapeutic approaches in order to improve patients' prospects, ranging from antibiotic treatment in chronic endometritis to heparin and aspirin in APS, as well as hormonal treatments for endometriosis/adenomyosis or a gluten-free diet in celiac disease. All of them and the rest of the therapeutic perspectives, both current and under investigation, are presented throughout this work, assessing the possible improvements for reproductive outcomes.
Collapse
Affiliation(s)
- Isabel Cuadrado-Torroglosa
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1a, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
| | - Juan A. García-Velasco
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1a, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
- IVIRMA Global Research Alliance, IVIRMA Madrid, Av. del Talgo, 68, 28023 Madrid, Spain
- Department of Obstetrics and Gynaecology, Rey Juan Carlos University, Av. de Atenas, s/n, 28922 Alcorcón, Spain
| | - Diana Alecsandru
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1a, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
- IVIRMA Global Research Alliance, IVIRMA Madrid, Av. del Talgo, 68, 28023 Madrid, Spain
| |
Collapse
|
46
|
Kim H, Bedsaul-Fryer JR, Schulze KJ, Sincerbeaux G, Baker S, Rebholz CM, Wu LSF, Gogain J, Cuddeback L, Yager JD, De Luca LM, Siddiqua TJ, West KP. An Early Gestation Plasma Inflammasome in Rural Bangladeshi Women. Biomolecules 2024; 14:736. [PMID: 39062451 PMCID: PMC11274825 DOI: 10.3390/biom14070736] [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: 05/29/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Circulating α1-acid glycoprotein (AGP) and C-reactive protein (CRP) are commonly measured to assess inflammation, but these biomarkers fail to reveal the complex molecular biology of inflammation. We mined the maternal plasma proteome to detect proteins that covary with AGP and CRP. In 435 gravida predominantly in <12-week gestation, we correlated the relative quantification of plasma proteins assessed via a multiplexed aptamer assay (SOMAScan®) with AGP and CRP, quantified by immunoassay. We defined a plasma inflammasome as protein correlates meeting a false discovery rate <0.05. We examined potential pathways using principal component analysis. A total of 147 and 879 of 6431 detected plasma proteins correlated with AGP and CRP, respectively, of which 61 overlapped with both biomarkers. Positive correlates included serum amyloid, complement, interferon-induced, and immunoregulatory proteins. Negative correlates were micronutrient and lipid transporters and pregnancy-related anabolic proteins. The principal components (PCs) of AGP were dominated by negatively correlated anabolic proteins associated with gestational homeostasis, angiogenesis, and neurogenesis. The PCs of CRP were more diverse in function, reflecting cell surface and adhesion, embryogenic, and intracellular and extra-hepatic tissue leakage proteins. The plasma proteome of AGP or CRP reveals wide proteomic variation associated with early gestational inflammation, suggesting mechanisms and pathways that merit future research.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA 98105, USA
| | - Jacquelyn R. Bedsaul-Fryer
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kerry J. Schulze
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gwen Sincerbeaux
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Sarah Baker
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Lee SF Wu
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | - James D. Yager
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Luigi M. De Luca
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Keith P. West
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
47
|
Khoshkerdar A, Eid N, Batra V, Baker N, Holmes N, Henson S, Sang F, Wright V, McLaren J, Shakesheff K, Woad KJ, Morgan HL, Watkins AJ. Sub-Optimal Paternal Diet at the Time of Mating Disrupts Maternal Adaptations to Pregnancy in the Late Gestation Mouse. Nutrients 2024; 16:1879. [PMID: 38931234 PMCID: PMC11206308 DOI: 10.3390/nu16121879] [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: 04/24/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Pregnancy represents a stage during which maternal physiology and homeostatic regulation undergo dramatic change and adaptation. The fundamental purpose of these adaptations is to ensure the survival of her offspring through adequate nutrient provision and an environment that is tolerant to the semi-allogenic foetus. While poor maternal diet during pregnancy is associated with perturbed maternal adaptations during pregnancy, the influence of paternal diet on maternal well-being is less clearly defined. We fed C57BL/6 male mice either a control (CD), low protein diet (LPD), a high fat/sugar Western diet (WD) or the LPD or WD supplemented with methyl donors (MD-LPD and MD-WD, respectively) for a minimum of 8 weeks prior to mating with C57BL/6 females. Mated females were culled at day 17 of gestation for the analysis of maternal metabolic, gut, cardiac and bone health. Paternal diet had minimal influences on maternal serum and hepatic metabolite levels or gut microbiota diversity. However, analysis of the maternal hepatic transcriptome revealed distinct profiles of differential gene expression in response to the diet of the father. Paternal LPD and MD-LPD resulted in differential expression of genes associated with lipid metabolism, transcription, ubiquitin conjugation and immunity in dams, while paternal WD and MD-WD modified the expression of genes associated with ubiquitin conjugation and cardiac morphology. Finally, we observed changes in maternal femur length, volume of trabecular bone, trabecular connectivity, volume of the cortical medullar cavity and thickness of the cortical bone in response to the father's diets. Our current study demonstrates that poor paternal diet at the time of mating can influence the patterns of maternal metabolism and gestation-associated adaptations to her physiology.
Collapse
Affiliation(s)
- Afsaneh Khoshkerdar
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Nader Eid
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Vipul Batra
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Nichola Baker
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Nadine Holmes
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Sonal Henson
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Fei Sang
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Victoria Wright
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Jane McLaren
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham NG7 2UH, UK; (J.M.)
| | - Kevin Shakesheff
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham NG7 2UH, UK; (J.M.)
| | - Kathryn J. Woad
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough LE12 5RD, UK;
| | - Hannah L. Morgan
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Adam J. Watkins
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| |
Collapse
|
48
|
Creisher PS, Klein SL. Pathogenesis of viral infections during pregnancy. Clin Microbiol Rev 2024; 37:e0007323. [PMID: 38421182 PMCID: PMC11237665 DOI: 10.1128/cmr.00073-23] [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: 03/02/2024] Open
Abstract
SUMMARYViral infections during pregnancy are associated with significant adverse perinatal and fetal outcomes. Pregnancy is a unique immunologic and physiologic state, which can influence control of virus replication, severity of disease, and vertical transmission. The placenta is the organ of the maternal-fetal interface and provides defense against microbial infection while supporting the semi-allogeneic fetus via tolerogenic immune responses. Some viruses, such as cytomegalovirus, Zika virus, and rubella virus, can breach these defenses, directly infecting the fetus and having long-lasting consequences. Even without direct placental infection, other viruses, including respiratory viruses like influenza viruses and severe acute respiratory syndrome coronavirus 2, still cause placental damage and inflammation. Concentrations of progesterone and estrogens rise during pregnancy and contribute to immunological adaptations, placentation, and placental development and play a pivotal role in creating a tolerogenic environment at the maternal-fetal interface. Animal models, including mice, nonhuman primates, rabbits, and guinea pigs, are instrumental for mechanistic insights into the pathogenesis of viral infections during pregnancy and identification of targetable treatments to improve health outcomes of pregnant individuals and offspring.
Collapse
Affiliation(s)
- Patrick S Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
49
|
Torelli FR, Rodrigues-Peres RM, Lopes-Cendes I, Bahamondes L, Juliato CRT. Gene expression associated with vaginal bleeding in women using the 52-mg levonorgestrel hormonal intrauterine device: A prospective study. Int J Gynaecol Obstet 2024; 165:1199-1209. [PMID: 38299835 DOI: 10.1002/ijgo.15357] [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: 05/05/2023] [Revised: 09/27/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024]
Abstract
OBJECTIVE To evaluate gene expression associated with vaginal bleeding in the 52-mg hormonal intrauterine device (IUD) users. MATERIALS AND METHODS We conducted a prospective study involving 100 women seeking to use the 52-mg hormonal IUD for contraception. We excluded women with a history or current condition of abnormal uterine bleeding and who were unable to attend a 1-year follow up. Women who expelled the device, removed it for reasons unrelated to vaginal bleeding, or were lost to follow up were discontinued. We collected endometrial biopsies immediately before IUD placement and assessed 20 selected genes using reverse transcription quantitative polymerase chain reaction. Users maintained a uterine bleeding diary for 12 months following IUD insertion. For statistical analysis, participants were categorized into groups with or without vaginal bleeding at 3 and 12 months. RESULTS Women with elevated CXCL9 expression had an 8.15-fold higher likelihood of experiencing vaginal bleeding at 3 months (odds ratio [OR] 8.15, 95% confidence interval [CI] 2.24-29.61, P = 0.001). At 12 months of follow up, women with increased TIMP1 expression had a 2.74-fold higher chance of experiencing vaginal bleeding (OR 2.74, 95% CI 1.08-6.95, P = 0.033). CXCL9 ≥ 1.5 and IL17A ≥ 0.68 were associated with a higher probability of vaginal bleeding at 3 months, while TIMP1 levels ≥0.943 were linked to an increased risk of bleeding at 12 months. CONCLUSION Users of the 52-mg hormonal IUD with elevated relative CXCL9 expression face an increased risk of vaginal bleeding at 3-month follow up, whereas those with heightened TIMP1 expression are more likely to experience vaginal bleeding at 12 months.
Collapse
Affiliation(s)
- Flávia R Torelli
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Raquel M Rodrigues-Peres
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Luis Bahamondes
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Cássia R T Juliato
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
50
|
Adamo KB, Goudreau AD, Corson AE, MacDonald ML, O'Rourke N, Tzaneva V. Physically active pregnancies: Insights from the placenta. Physiol Rep 2024; 12:e16104. [PMID: 38872466 PMCID: PMC11176744 DOI: 10.14814/phy2.16104] [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: 02/01/2024] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Physical activity (PA) positively influences pregnancy, a critical period for health promotion, and affects placental structure and function in ways previously overlooked. Here, we summarize the current body of literature examining the association between PA, placenta biology, and physiology while also highlighting areas where gaps in knowledge exist. PA during pregnancy induces metabolic changes, influencing nutrient availability and transporter expression in the placenta. Hormones and cytokines secreted during PA contribute to health benefits, with intricate interactions in pro- and anti-inflammatory markers. Extracellular vesicles and placental "-omics" data suggest that gestational PA can shape placental biology, affecting gene expression, DNA methylation, metabolite profiles, and protein regulation. However, whether cytokines that respond to PA alter placental proteomic profiles during pregnancy remains to be elucidated. The limited research on placenta mitochondria of physically active gestational parents (gesP), has shown improvements in mitochondrial DNA and antioxidant capacity, but the relationship between PA, placental mitochondrial dynamics, and lipid metabolism remains unexplored. Additionally, PA influences the placenta-immune microenvironment, angiogenesis, and may confer positive effects on neurodevelopment and mental health through placental changes, vascularization, and modulation of brain-derived neurotrophic factor. Ongoing exploration is crucial for unraveling the multifaceted impact of PA on the intricate placental environment.
Collapse
Affiliation(s)
- Kristi B Adamo
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandra D Goudreau
- Department of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Abbey E Corson
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Meaghan L MacDonald
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Nicholas O'Rourke
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Velislava Tzaneva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|