1
|
Habelrih T, Augustin TL, Mauffette-Whyte F, Ferri B, Sawaya K, Côté F, Gallant M, Olson DM, Chemtob S. Inflammatory mechanisms of preterm labor and emerging anti-inflammatory interventions. Cytokine Growth Factor Rev 2024; 78:50-63. [PMID: 39048393 DOI: 10.1016/j.cytogfr.2024.07.007] [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: 07/04/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Preterm birth is a major public health concern, requiring a deeper understanding of its underlying inflammatory mechanisms and to develop effective therapeutic strategies. This review explores the complex interaction between inflammation and preterm labor, highlighting the pivotal role of the dysregulation of inflammation in triggering premature delivery. The immunological environment of pregnancy, characterized by a fragile balance of immune tolerance and resistance, is disrupted in preterm labor, leading to a pathological inflammatory response. Feto-maternal infections, among other pro-inflammatory stimuli, trigger the activation of toll-like receptors and the production of pro-inflammatory mediators, promoting uterine contractility and cervical ripening. Emerging anti-inflammatory therapeutics offer promising approaches for the prevention of preterm birth by targeting key inflammatory pathways. From TLR-4 antagonists to chemokine and interleukin receptor antagonists, these interventions aim to modulate the inflammatory environment and prevent adverse pregnancy outcomes. In conclusion, a comprehensive understanding of the inflammatory mechanisms leading to preterm labor is crucial for the development of targeted interventions in hope of reducing the incidence of preterm birth and improving neonatal health outcomes.
Collapse
Affiliation(s)
- Tiffany Habelrih
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Thalyssa-Lyn Augustin
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Félix Mauffette-Whyte
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Béatrice Ferri
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Kevin Sawaya
- Research Center, CHU Sainte-Justine, Montreal, QC, Canada; Programmes de cycles supérieurs en sciences biomédicales, Faculté de médecine, Université de Montréal, Montreal, QC, Canada
| | - France Côté
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Mathilde Gallant
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada.
| |
Collapse
|
2
|
Mosebarger A, Vidal MS, Bento GFC, Lintao RCV, Severino MEL, Kumar Kammala A, Menon R. Immune cells at the feto-maternal interface: Comprehensive characterization and insights into term labor. J Reprod Immunol 2024; 163:104239. [PMID: 38493591 DOI: 10.1016/j.jri.2024.104239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/05/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Immune cells at the feto-maternal interface play an important role in pregnancy; starting at implantation, maintenance of pregnancy, and parturition. The role of decidual immune cells in induction of labor still needs to be understood. Published reports on this topic show heterogeneity in methods of cell isolation, assay, analysis and cellular characterization making it difficult to collate available information in order to understand the contribution of immune cells at term leading to parturition. In the present study, available literature was reviewed to study the differences in immune cells between the decidua basalis and decidua parietalis, as well as between immune cells in term and preterm labor. Additionally, immune cells at the decidua parietalis were isolated from term not in labor (TNL) or term in labor (TL) samples and characterized via flow cytometry using a comprehensive, high-dimensional antibody panel. This allowed a full view of immune cell differences without combining multiple studies, which must include variation in isolation and analysis methods, for more conclusive data. The ratio of cells found in decidua parietalis in this study generally matched those reported in the literature, although we report a lower percentage of natural killer (NK) cells at term. We report that CD4 expression on CD8- NK cells decreased in term labor compared to not in labor samples, suggesting that natural killer cells may be migrating to other sites during labor. Also, we report a decrease in CD38 expression on CD8+ CD57+ T cells in labor, indicative of cytotoxic T cell senescence. Our study provides a comprehensive status of immune cells at the decidua-chorion interface at term.
Collapse
Affiliation(s)
- Angela Mosebarger
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Manuel S Vidal
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | | | - Ryan C V Lintao
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | - Mary Elise L Severino
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | - Ananth Kumar Kammala
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| |
Collapse
|
3
|
Xue L, Chen R, Liu Y, Niu P, Zhou J, Liu J, Zhang J, Chen H. Association of maternal blood high-mobility group box 1 levels and adverse pregnancy outcomes: A systematic review and meta-analysis. Reprod Biol 2024; 24:100859. [PMID: 38492434 DOI: 10.1016/j.repbio.2024.100859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 03/18/2024]
Abstract
Conflicting findings have emerged regarding the levels of high mobility group box 1 (HMGB1) in individuals experiencing adverse pregnancy outcomes. Here we conducted a meta-analysis to assess the association between maternal blood HMGB1 levels and adverse pregnancy outcomes. Utilizing databases such as PubMed, Cochrane Central Register of Controlled Trials, Web of Science, Embase and China National Knowledge Infrastructure (CNKI), a systematic literature search was conducted in January 2024. Eligible literature was screened according to inclusion and exclusion criteria. Quality assessment was evaluated using the Newcastle-Ottawa Scale (NOS). The extracted data were analyzed using Review Manager 5.4 and STATA 12.0 software. 21 observational studies with a total of 2471 participants were included in this meta-analysis. Significantly higher peripheral blood levels of HMGB1 were associated with preeclampsia (PE) (SMD=1.34; 95% CI: 0.72-1.95; P < 0.0001) and gestational diabetes mellitus (GDM) (SMD=1.20; 95% CI: 0.31-2.09; P = 0.009). Additionally, HMGB1 levels in peripheral blood were significantly elevated in patients with unexplained recurrent spontaneous abortion (URSA) than those in pregnancy controls (SMD=4.22; 95% CI: 1.64-6.80; P = 0.001) or non-pregnancy controls (SMD=3.87; 95% CI: 1.81-5.92; P = 0.0002). Interestingly, higher blood HMGB1 levels were observed in women with preterm birth (PTB), however, the results did not reach a statistical difference (SMD=0.54; 95% CI: -0.36-1.44; P = 0.24). In conclusion, overexpressed maternal blood HMGB1 levels were associated with adverse pregnancy outcomes, including PE, GDM and URSA. Further studies should be conducted to validate the efficacy of HMGB1 as a biomarker for assessing the risk of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Liping Xue
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruolin Chen
- College of Letter and Science, University of California Davis, CA, USA
| | - Ying Liu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Peiguang Niu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Jintuo Zhou
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Liu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Zhang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Huajiao Chen
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
4
|
Bonney EA, Lintao RCV, Zelop CM, Kammala AK, Menon R. Are fetal microchimerism and circulating fetal extracellular vesicles important links between spontaneous preterm delivery and maternal cardiovascular disease risk? Bioessays 2024; 46:e2300170. [PMID: 38359068 DOI: 10.1002/bies.202300170] [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: 09/05/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/17/2024]
Abstract
Trafficking and persistence of fetal microchimeric cells (fMCs) and circulating extracellular vesicles (EVs) have been observed in animals and humans, but their consequences in the maternal body and their mechanistic contributions to maternal physiology and pathophysiology are not yet fully defined. Fetal cells and EVs may help remodel maternal organs after pregnancy-associated changes, but the cell types and EV cargos reaching the mother in preterm pregnancies after exposure to various risk factors can be distinct from term pregnancies. As preterm delivery-associated maternal complications are rising, revisiting this topic and formulating scientific questions for future research to reduce the risk of maternal morbidities are timely. Epidemiological studies report maternal cardiovascular risk as one of the major complications after preterm delivery. This paper suggests a potential link between fMCs and circulating EVs and adverse maternal cardiovascular outcomes post-pregnancies, the underlying mechanisms, consequences, and methods for and how this link might be assessed.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, The University of Vermont, Burlington, Vermont, USA
| | - Ryan C V Lintao
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
- College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Carolyn M Zelop
- The Valley Hospital, Ridgewood, Paramus, New Jersey, USA
- Grossman School of Medicine, New York University, New York City, New York, USA
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
5
|
Jancsura MK, Schmella MJ, Helsabeck N, Gillespie SL, Roberts JM, Conley YP, Hubel CA. Inflammatory markers are elevated in early pregnancy, but not late pregnancy, in women with overweight and obesity that later develop preeclampsia. Am J Reprod Immunol 2023; 90:e13763. [PMID: 37641371 PMCID: PMC10465815 DOI: 10.1111/aji.13763] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
PROBLEM Obesity and preeclampsia both involve a pathological inflammatory response, which may be how obesity increases preeclampsia risk. Previous studies have failed to assess robust measurements of inflammatory markers across gestation, specifically in overweight/ obese women in the context of preeclampsia. METHOD OF STUDY We measured 20 inflammatory markers in plasma via multiplex assay (ThermoFisher Inflammation 20 plex Human ProcartaPlex Panel) across the three trimesters of pregnancy in an existing cohort of overweight and obese women who developed preeclampsia (n = 37) and without preeclampsia (n = 74). Mann-Whitney U tests examined differences in inflammatory marker concentrations between cases and controls. Repeated measures ANOVA tests were used to explore differences in inflammatory marker concentrations over time within cases and controls. RESULTS Pro-inflammatory markers (IL-1α, IL-1β, IL-6, IFN-α, IFN-γ, GM-CSF, IL-12p70, IL-17α, TNF-α, IL-8) and anti-inflammatory markers (IL-4, IL-10, IL-13) were higher in the first and second trimester in participants who later developed preeclampsia compared to those who did not (p < .05). Only TNF-α and IL-8 remained elevated in the third trimester. Inflammatory markers did not change across pregnancy in preeclampsia cases but did increase across pregnancy in controls. CONCLUSION Our findings diverge from prior studies, predominantly of non-obese women, that report lower circulating concentrations of anti-inflammatory cytokines in preeclampsia versus normotensive pregnancy, particularly by late pregnancy. We posit that women with overweight and obesity who develop preeclampsia entered pregnancy with a heightened pro-inflammatory state likely related to obesity, which increased risk for preeclampsia. Further studies are needed to investigate if inflammatory maker profiles differ between obese and non-obese women.
Collapse
Affiliation(s)
- McKenzie K Jancsura
- College of Nursing Martha S. Pitzer Center for Women, Children and Youth, The Ohio State University, Columbus, USA
| | | | | | - Shannon L Gillespie
- College of Nursing Martha S. Pitzer Center for Women, Children and Youth, The Ohio State University, Columbus, USA
| | - James M Roberts
- Departments of Obstetrics Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Epidemiology and Clinical and Translational Research, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Magee-Womens Research Institute, Pittsburgh, United States
| | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, USA
| | - Carl A Hubel
- Departments of Obstetrics Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Magee-Womens Research Institute, Pittsburgh, United States
| |
Collapse
|
6
|
Morales-Vázquez MM, Meza-Serrano E, Lara-Pereyra I, Acuña-González RJ, Alonso-Morales R, Hayen-Valles S, Boeta AM, Zarco L, Lozano-Cuenca J, López-Canales JS, Flores-Herrera H. Equine Placentitis in Mares Induces the Secretion of Pro-Inflammatory Cytokine eIL-1β and the Active Extracellular Matrix Metalloproteinase (MMP)-9. Vet Sci 2023; 10:532. [PMID: 37756054 PMCID: PMC10536981 DOI: 10.3390/vetsci10090532] [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: 06/19/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
Equine placentitis is characterized by infection and inflammation of the placenta. Different biomarkers associated with this inflammatory response have been evaluated in experimentally induced equine placentitis, but not in pregnant mares with spontaneous placentitis. The aim of the current study was to determine the concentration of eIL-1β and the activity of proMMP-2 and proMMP-9 in the serum of healthy mares and mares with placentitis on days 240 and 320 of gestation to explore whether these biomarkers are associated with equine maternal placentitis and/or with the birth of an infected or inviable foals. Serum samples were collected from sixteen pregnant English Thoroughbred mares, retrospectively classified as follows: (1) healthy mares with full-term gestation; and (2) mares with ultrasonographic signs of placentitis. The health of each foal was examined at birth, and it was decided to classify the cases into four groups: (1) healthy mares delivering a healthy foals (HM-HF, n = 6); (2) mares with USP delivering a healthy foal (USP-HF, n = 3); (3) mares with USP delivering a live septic foal (USP-LSeF, n = 4); and (4) mares with USP delivering a dead foal (USP-DF, n = 3). eIL-1β was quantified by ELISA, and proMMP-2 and proMMP-9 activity by gelatin zymography electrophoresis. In healthy mares, the serum concentrations of eIL-1β underwent a significant 16.5-fold increase from day 240 to day 320 of gestation. Although similar results were found in the mares with ultrasonographic signs of placentitis that delivered a healthy foal, those delivering a live septic or nonviable foal exhibited much higher concentrations of eIL-1β. proMMP-2 and proMMP-9 activity was not associated with maternal placentitis, foal infection, or death. Hence, the presence of placentitis severe enough to affect the health of the foal can be confirmed or discarded by determining the eIL-1β concentration in mares that have shown ultrasonographic signs of placentitis.
Collapse
Affiliation(s)
- María Margarita Morales-Vázquez
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Europa Meza-Serrano
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Irlando Lara-Pereyra
- Departamento de Ginecología, Hospital General de Zona 252, Instituto Mexicano del Seguro Social, Atlacomulco 28984, Mexico
| | - Ricardo Josué Acuña-González
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
| | - Rogelio Alonso-Morales
- Genética, Laboratorio de Biotecnologías, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico;
| | - Sergio Hayen-Valles
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Ana Myriam Boeta
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, CP, Mexico; (E.M.-S.); (S.H.-V.); (A.M.B.)
| | - Luis Zarco
- Centro de Enseñanza, Investigación y Extensión en Producción Ovina, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Tres Marías, Ciudad de México 62515, Mexico;
| | - Jair Lozano-Cuenca
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, Mexico; (J.L.-C.); (J.S.L.-C.)
| | - Jorge Skiold López-Canales
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, Mexico; (J.L.-C.); (J.S.L.-C.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Héctor Flores-Herrera
- Departamento de Immunobioquímica, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes” INPerIER, Ciudad de México 11000, CP, Mexico; (M.M.M.-V.); (R.J.A.-G.)
| |
Collapse
|
7
|
Flores-Espinosa P, Méndez I, Irles C, Olmos-Ortiz A, Helguera-Repetto C, Mancilla-Herrera I, Ortuño-Sahagún D, Goffin V, Zaga-Clavellina V. Immunomodulatory role of decidual prolactin on the human fetal membranes and placenta. Front Immunol 2023; 14:1212736. [PMID: 37359537 PMCID: PMC10288977 DOI: 10.3389/fimmu.2023.1212736] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
The close interaction between fetal and maternal cells during pregnancy requires multiple immune-endocrine mechanisms to provide the fetus with a tolerogenic environment and protection against any infectious challenge. The fetal membranes and placenta create a hyperprolactinemic milieu in which prolactin (PRL) synthesized by the maternal decidua is transported through the amnion-chorion and accumulated into the amniotic cavity, where the fetus is bedded in high concentrations during pregnancy. PRL is a pleiotropic immune-neuroendocrine hormone with multiple immunomodulatory functions mainly related to reproduction. However, the biological role of PRL at the maternal-fetal interface has yet to be fully elucidated. In this review, we have summarized the current information on the multiple effects of PRL, focusing on its immunological effects and biological significance for the immune privilege of the maternal-fetal interface.
Collapse
Affiliation(s)
- Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Isabel Méndez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Claudine Irles
- Institut National de la Santé et de la Recherche Médicale (INSERM) U978, Université Sorbonne Paris Nord, Unité de Formation et de Recherche (UFR) Santé Médecine et Biologie Humaine (SMBH), Bobigny, France
| | - Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Cecilia Helguera-Repetto
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Ismael Mancilla-Herrera
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara, Mexico
| | - Vincent Goffin
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)-S1151, CNRS Unité Mixte de Recherche (UMR)-S8253, Institut Necker Enfants Malades, Paris, France
| | - Verónica Zaga-Clavellina
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| |
Collapse
|
8
|
Taylor BD, Adekanmbi V, Zhang Y, Berenson A. The Impact of Neisseria gonorrhoeae Mono- and Coinfection on Adverse Pregnancy Outcomes. Open Forum Infect Dis 2023; 10:ofad220. [PMID: 37250177 PMCID: PMC10220503 DOI: 10.1093/ofid/ofad220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Background Sexually transmitted infections (STIs) have recently been linked to hypertensive disorders of pregnancy (HDP). However, the impact of Neisseria gonorrhoeae on risk of HDP is not well understood. This study determined the impact of gonorrhea and gonorrhea coinfection on HDP and other adverse pregnancy outcomes in a population with a high screening rate and presumed treatment. Methods This retrospective study included 29 821 singleton births between 2016 and 2021. The STI testing results, demographic variables, and pregnancy outcomes were identified from electronic health records. The HDP were primary outcomes of interest including gestational hypertension, preeclampsia, and superimposed preeclampsia. We further examined preeclampsia subtypes defined by severe features and gestational age of delivery (term and preterm preeclampsia). Secondary outcomes included preterm premature rupture of membranes, chorioamnionitis, and preterm delivery. Logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs). Models were adjusted for maternal age, maternal race/ethnicity, and smoking. Results Gonorrhea screening occurred in 95% of the population. Gonorrhea increased the odds of preterm preeclampsia (adjusted OR [ORadj.], 1.95; 95% CI, 1.02-3.73) and preterm birth (ORadj., 1.78; 95% CI, 1.22-2.60). Furthermore, gonorrhea-chlamydia coinfection was associated with preterm birth (ORadj., 1.77; 95% CI, 1.03-3.04). However, results were similar when we examined gonorrhea monoinfection (ORadj., 1.76; 95% CI, 1.04-2.97). Conclusions Among a diverse population of pregnant individuals, gonorrhea increased odds of preterm preeclampsia and preterm delivery Further research is needed to determine the burden of STIs on HDP, including investigations into biological effects during pregnancy.
Collapse
Affiliation(s)
- Brandie DePaoli Taylor
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Population Health and Health Disparities, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Interdisciplinary Research in Women's Health, University of Texas Medical Branch, Galveston, Texas, USA
| | - Victor Adekanmbi
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Interdisciplinary Research in Women's Health, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuanyi Zhang
- Department of Biostatistics and Data Science, University of Texas Medical Branch, Galveston, Texas, USA
| | - Abbey Berenson
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Interdisciplinary Research in Women's Health, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
9
|
Dumitriu D, Baldwin E, Coenen RJ, Hammond LA, Peterka DS, Heilbrun L, Frye RE, Palmer R, Norrman HN, Fridell A, Remnelius KL, Isaksson J, Austin C, Curtin P, Bölte S, Arora M. Deciduous tooth biomarkers reveal atypical fetal inflammatory regulation in autism spectrum disorder. iScience 2023; 26:106247. [PMID: 36926653 PMCID: PMC10011823 DOI: 10.1016/j.isci.2023.106247] [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: 09/21/2022] [Revised: 01/17/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Atypical regulation of inflammation has been proposed in the etiology of autism spectrum disorder (ASD); however, measuring the temporal profile of fetal inflammation associated with future ASD diagnosis has not been possible. Here, we present a method to generate approximately daily profiles of prenatal and early childhood inflammation as measured by developmentally archived C-reactive protein (CRP) in incremental layers of deciduous tooth dentin. In our discovery population, a group of Swedish twins, we found heightened inflammation in the third trimester in children with future ASD diagnosis relative to controls (n = 66; 14 ASD cases; critical window: -90 to -50 days before birth). In our replication study, in the US, we observed a similar increase in CRP in ASD cases during the third trimester (n = 47; 23 ASD cases; -128 to -21 days before birth). Our results indicate that the third trimester is a critical period of atypical fetal inflammatory regulation in ASD.
Collapse
Affiliation(s)
- Dani Dumitriu
- Departments of Neuroscience and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Departments of Pediatrics and Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian Morgan Stanley Children’s Hospital, New York, NY 10032, USA
| | - Elena Baldwin
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Roozie J.J. Coenen
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luke A. Hammond
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Darcy S. Peterka
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Lynne Heilbrun
- Family and Community Medicine, School of Medicine, University of Texas Health Sciences Center, San Antonio, TX 78229, USA
| | - Richard E. Frye
- Department of Neurology, Barrow Neurological Institute, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
| | - Raymond Palmer
- Family and Community Medicine, School of Medicine, University of Texas Health Sciences Center, San Antonio, TX 78229, USA
| | - Hjalmar Nobel Norrman
- Center of Neurodevelopmental Disorder (KIND), Division of Neuropsychiatry, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Anna Fridell
- Center of Neurodevelopmental Disorder (KIND), Division of Neuropsychiatry, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm 171 77, Sweden
- Child and Adolescent Psychiatry Research Center, Center for Psychiatry Research, Region Stockholm, Stockholm 104 31, Sweden
| | - Karl Lundin Remnelius
- Center of Neurodevelopmental Disorder (KIND), Division of Neuropsychiatry, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Johan Isaksson
- Center of Neurodevelopmental Disorder (KIND), Division of Neuropsychiatry, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm 171 77, Sweden
- Department of Medical Sciences, Child and Adolescent Psychiatry Unit, Uppsala University, Uppsala 751 85, Sweden
| | - Christine Austin
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Linus Biotechnology Inc., New York, NY 10013, USA
| | - Paul Curtin
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Linus Biotechnology Inc., New York, NY 10013, USA
| | - Sven Bölte
- Center of Neurodevelopmental Disorder (KIND), Division of Neuropsychiatry, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm 171 77, Sweden
- Child and Adolescent Psychiatry Research Center, Center for Psychiatry Research, Region Stockholm, Stockholm 104 31, Sweden
- Curtin Autism Research Group, Curtin School of Allied Health, Curtin University, Perth, WA 6102, Australia
| | - Manish Arora
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Linus Biotechnology Inc., New York, NY 10013, USA
| |
Collapse
|
10
|
Kyathanahalli C, Snedden M, Hirsch E. Is human labor at term an inflammatory condition?†. Biol Reprod 2023; 108:23-40. [PMID: 36173900 PMCID: PMC10060716 DOI: 10.1093/biolre/ioac182] [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: 05/11/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 01/20/2023] Open
Abstract
Parturition at term in normal pregnancy follows a predictable sequence of events. There is some evidence that a state of inflammation prevails in the reproductive tissues during labor at term, but it is uncertain whether this phenomenon is the initiating signal for parturition. The absence of a clear temporal sequence of inflammatory events prior to labor casts doubt on the concept that normal human labor at term is primarily the result of an inflammatory cascade. This review examines evidence linking parturition and inflammation in order to address whether inflammation is a cause of labor, a consequence of labor, or a separate but related phenomenon. Finally, we identify and suggest ways to reconcile inconsistencies regarding definitions of labor onset in published research, which may contribute to the variability in conclusions regarding the genesis and maintenance of parturition. A more thorough understanding of the processes underlying normal parturition at term may lead to novel insights regarding abnormal labor, including spontaneous preterm labor, preterm premature rupture of the fetal membranes, and dysfunctional labor, and the role of inflammation in each.
Collapse
Affiliation(s)
- Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Madeline Snedden
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Emmet Hirsch
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
11
|
An Z, Zhao R, Han F, Sun Y, Liu Y, Liu L. Potential Serum Biomarkers Associated with Premature Rupture of Fetal Membranes in the First Trimester. Front Pharmacol 2022; 13:915935. [PMID: 35873552 PMCID: PMC9304655 DOI: 10.3389/fphar.2022.915935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Premature rupture of the fetal membranes (PROM) is a common and important obstetric complication with increased risk of adverse consequences for both mothers and fetuses. An accurate and timely method to predict the occurrence of PROM is needed for ensuring maternal and fetal safety. Untargeted metabolomics was applied to characterize metabolite profiles related to PROM in early pregnancy. 41 serum samples from pregnant women who developed PROM later in gestation and 106 from healthy pregnant women as a control group, were analyzed. Logistic regression analysis was adjusted to analyze a PROM prediction model in the first trimester. A WISH amniotic cell viability assay was applied to explore the underlying mechanisms involved in PROM, mediated by C8-dihydroceramide used to mimic a potential biomarker (Cer 40:0; O2). Compared with healthy controls, 13 serum metabolites were identified. The prediction model comprising four compounds (Cer 40:0; O2, sphingosine, isohexanal and PC O-38:4) had moderate accuracy to predict PROM events with the maximum area under the curve of a receiver operating characteristics curve of approximately 0.70. Of these four compounds, Cer 40:0; O2 with an 1.81-fold change between PROM and healthy control serum samples was defined as a potential biomarker and inhibited the viability of WISH cells. This study sheds light on predicting PROM in early pregnancy and on understanding the underlying mechanism of PROM.Trial Registration: This study protocol has been registered at www.ClinicalTrials.gov, CT03651934, on 29 August 2018 (prior to recruitment).
Collapse
Affiliation(s)
- Zhuoling An
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Rui Zhao
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Feifei Han
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yuan Sun
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yanping Liu
- Department of Clinical Nutrition, Peking Union Medical College Hospital, China Academic Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Yanping Liu, ; Lihong Liu,
| | - Lihong Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yanping Liu, ; Lihong Liu,
| |
Collapse
|
12
|
Menon R, Richardson L. Organ-on-a-chip for perinatal biology experiments. PLACENTA AND REPRODUCTIVE MEDICINE 2022; 1:98. [PMID: 36530581 PMCID: PMC9757604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Cell culture and organ explant systems have traditionally been used by scientists in the reproductive biology and perinatal medicine area to address various research questions. Although most are unrelated to human pregnancy, animal models were also extensively used to study various mechanisms associated with pregnancy and parturition. However, limitations of traditional approaches have shifted the attention to the use of organ on a chip (OOC) technology. OOC platform simulates an organ using cells, and OOCs are biomimetic microfluidic systems comprising multiple cell types from an organ that mimic the environment of a physiological organ. OOC maintains intercellular interactions and helps to recreate organ physiology as expected for utero in perinatal medicine research. This short review introduces some basic concepts of OOC, and its utility based on some published reports.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555 TX, USA
| | - Lauren Richardson
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555 TX, USA
| |
Collapse
|
13
|
Zhang Y, Feng W, Li J, Cui L, Chen ZJ. Periodontal Disease and Adverse Neonatal Outcomes: A Systematic Review and Meta-Analysis. Front Pediatr 2022; 10:799740. [PMID: 35601423 PMCID: PMC9114501 DOI: 10.3389/fped.2022.799740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Objective The aim of this study was to evaluate the association between maternal periodontal disease (PD) and three main adverse neonatal outcomes, namely, preterm birth (PTB), low birth weight (LBW), and small for gestational age (SGA). Methods The Ovid Medline, Web of Science, Embase, and Cochrane Library were searched up to 6 December 2020 for relevant observational studies on an association between PD and risk of PTB, LBW, and SGA. Eligibility criteria included observational studies which compared the prevalence of PTB and/or LBW and/or SGA between PD women and periodontal health controls. The exclusion criteria included incomplete data, animal research, and mixing up various pregnancy outcomes, such as "preterm low birth weight" and languages other than Chinese and English. Data were extracted and analyzed independently by two authors. The meta-analysis was performed using Stata Statistical Software, Release 12 (StataCorp LP, College Station, TX, USA). Odds ratio (OR), confidence intervals (CIs), and heterogeneity (I 2) were computed. Results Fourteen case-control studies and 10 prospective cohort studies, involving 15,278 participants, were identified. Based on fixed effect meta-analysis, PTB showed a significant association with PD (OR = 1.57, 95% CI: 1.39-1.77, P < 0.00001) and LBW also showed a significant association with PD (OR = 2.43, 95% CI: 1.75-3.37, P < 0.00001) in a random effect meta-analysis. However, a random effect meta-analysis showed no relationship between PD and SGA (OR = 1.62, 95% CI: 0.86-3.07, P = 0.136). Conclusion Our findings indicate that pregnant women with PD have a significantly higher risk of PTB and LBW. However, large prospective, blinded cohort studies with standardized diagnostic criteria of PD and adequate control of confounding factors are still required to confirm the relationship between PD and adverse neonatal outcomes.
Collapse
Affiliation(s)
- Youzhen Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Wanbing Feng
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Jingyu Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Linlin Cui
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Menon R. Fetal inflammatory response at the fetomaternal interface: A requirement for labor at term and preterm. Immunol Rev 2022; 308:149-167. [PMID: 35285967 DOI: 10.1111/imr.13075] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022]
Abstract
Human parturition at term and preterm is an inflammatory process synchronously executed by both fetomaternal tissues to transition them from a quiescent state t an active state of labor to ensure delivery. The initiators of the inflammatory signaling mechanism can be both maternal and fetal. The placental (fetal)-maternal immune and endocrine mediated homeostatic imbalances and inflammation are well reported. However, the fetal inflammatory response (FIR) theories initiated by the fetal membranes (amniochorion) at the choriodecidual interface are not well established. Although immune cell migration, activation, and production of proparturition cytokines to the fetal membranes are reported, cellular level events that can generate a unique set of inflammation are not well discussed. This review discusses derangements to fetal membrane cells (physiologically and pathologically at term and preterm, respectively) in response to both endogenous and exogenous factors to generate inflammatory signals. In addition, the mechanisms of inflammatory signal propagation (fetal signaling of parturition) and how these signals cause immune imbalances at the choriodecidual interface are discussed. In addition to maternal inflammation, this review projects FIR as an additional mediator of inflammatory overload required to promote parturition.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
15
|
Double Balloon Catheter (Plus Oxytocin) versus Dinoprostone Vaginal Insert for Term Rupture of Membranes: A Randomized Controlled Trial (RUBAPRO). J Clin Med 2022; 11:jcm11061525. [PMID: 35329852 PMCID: PMC8952372 DOI: 10.3390/jcm11061525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 01/27/2023] Open
Abstract
Background: The aim of this study is to demonstrate that a double balloon catheter combined with oxytocin decreases time between induction of labor and delivery (TID) as compared to a vaginal dinoprostone insert in cases of premature rupture of membranes at term. Methods: This is a prospective, randomized, controlled trial including patient undergoing labor induction for PROM at term with an unfavorable cervix in Clermont-Ferrand university hospital. We compared the double balloon catheter over a period of 12 h with adjunction of oxytocin 6 h after catheter insertion versus dinoprostone vaginal insert. After device ablation, cervical ripening continued only with oxytocin. The main outcome was TID. Secondary outcomes concerned delivery mode, as well as maternal and fetal outcome, and were adjusted for parity. Results: 40 patients per group were randomized. Each group had similar baseline characteristics. The study failed to demonstrate reduced TID (16.2 versus 20.2 h, ES = 0.16 (−0.27 to 0.60), p = 0.12) in the catheter group versus dinoprostone except in nulliparous women (17.0 versus 26.5 h, ES = 0.62 (0.10 to 1.14), p = 0.006). The rate of vaginal delivery <24 h significantly increased with combined induction (88.5% versus 66.6%, p = 0.03). No statistical difference was observed concerning caesarean rate (12.5% versus 17.5%, p > 0.05), chorioamnionitis (0% versus 2.5%, p = 1), postpartum endometritis, or maternal or neonatal outcomes. Procedure-related pain and tolerance to devices were found to be similar for the two methods. Interpretation: The double balloon catheter combined with oxytocin is an alternative for cervical ripening in case of PROM at term, and may reduce TID in nulliparous women.
Collapse
|
16
|
Fan M, Pan T, Jin W, Sun J, Zhang S, Du Y, Chen X, Chen Q, Xu W, Choo SW, Zhu G, Chen Y, Zhou J. FGF4, A New Potential Regulator in Gestational Diabetes Mellitus. Front Pharmacol 2022; 13:827617. [PMID: 35317005 PMCID: PMC8934430 DOI: 10.3389/fphar.2022.827617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Gestational diabetes mellitus (GDM) is associated with adverse maternal and neonatal outcomes, however the underlying mechanisms remain elusive. The aim of this study was to find efficient regulator of FGFs in response to the pathogenesis of GDM and explore the role of the FGFs in GDM.Methods: We performed a systematic screening of placental FGFs in GDM patients and further in two different GDM mouse models to investigate their expression changes. Significant changed FGF4 was selected, engineered, purified, and used to treat GDM mice in order to examine whether it can regulate the adverse metabolic phenotypes of the diabetic mice and protect their fetus.Results: We found FGF4 expression was elevated in GDM patients and its level was positively correlated to blood glucose, indicating a physiological relevance of FGF4 with respect to the development of GDM. Recombinant FGF4 (rFGF4) treatment could effectively normalize the adverse metabolic phenotypes in high fat diet induced GDM mice but not in STZ induced GDM mice. However, rFGF4 was highly effective in reduce of neural tube defects (NTDs) of embryos in both the two GDM models. Mechanistically, rFGF4 treatment inhibits pro-inflammatory signaling cascades and neuroepithelial cell apoptosis of both GDM models, which was independent of glucose regulation.Conclusions/interpretation: Our study provides novel insight into the important roles of placental FGF4 and suggests that it may serve as a promising diagnostic factor and therapeutic target for GDM.
Collapse
Affiliation(s)
- Miaojuan Fan
- Department of Infectious Diseases & Zhejiang Provincial Key laboratory of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Baoji Maternal and Child Health Hospital, Baoji, China
| | - Tongtong Pan
- Department of Infectious Diseases & Zhejiang Provincial Key laboratory of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Jin
- Department of Infectious Diseases & Zhejiang Provincial Key laboratory of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jian Sun
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shujun Zhang
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yali Du
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinwei Chen
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiong Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenxin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Siew Woh Choo
- College of Science and Technology, Wenzhou-Kean University, Wenzhou, China
| | - Guanghui Zhu
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Guanghui Zhu, ; Yongping Chen, ; Jie Zhou,
| | - Yongping Chen
- Department of Infectious Diseases & Zhejiang Provincial Key laboratory of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Guanghui Zhu, ; Yongping Chen, ; Jie Zhou,
| | - Jie Zhou
- Department of Infectious Diseases & Zhejiang Provincial Key laboratory of Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Guanghui Zhu, ; Yongping Chen, ; Jie Zhou,
| |
Collapse
|
17
|
Menon R, Dixon CL, Cayne S, Radnaa E, Salomon C, Sheller-Miller S. Differences in cord blood extracellular vesicle cargo in preterm and term births. Am J Reprod Immunol 2022; 87:e13521. [PMID: 35007379 DOI: 10.1111/aji.13521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE This study determined the cord plasma-derived extracellular vesicle (exosomes; 30-160 nm particles) proteomic profile in patients who had spontaneous preterm birth (PTB) or preterm premature rupture of membranes (pPROM), compared to those who delivered at term regardless of labor status. METHODS This is a cross-sectional analysis of a retrospective cohort that quantified and determined the proteomic cargo content of exosomes present in cord blood plasma samples in PTB or pPROM, and normal term in labor (TL) or term not in labor (TNIL) pregnancies. Exosomes were isolated by differential centrifugation followed by size exclusion chromatography. Exosomes were characterized by nanoparticle tracking analysis (quantity and size) and markers (dot blots for exosome markers). The exosomal proteomic profile was identified by liquid chromatography-mass spectrometry (LC-MS/MS). Ingenuity pathway analysis determined canonical pathways and biofunctions associated with dysregulated proteins. RESULTS Cord plasma exosomes have similar quantity and exhibit both tetraspanin and ESCRT protein markers specific of exosomes regardless of the conditions. Proteomics analysis exhibited several similar markers as well as very unique markers in exosomes from each condition; however, bioinformatics analysis revealed a generalized and non-specific inflammatory condition represented in exosomes from different condition that is not indicative of any specific underlying biological functions indicative of an underlying pathology. CONCLUSIONS Compared to maternal plasma and amniotic fluid exosomes, the value of cord plasma derived exosomes is limited. Quantity, character, and proteomic cargo contents in exosomes or the pathways and functions represented by differentially expressed proteins do not distinguish specific conditions regarding normal and abnormal parturition. The value of cord plasma exosome proteomic cargo has limited value as an indicator of an underlying physiology or as a biomarker of fetal well-being.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Christopher Luke Dixon
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Samir Cayne
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Enkhtuya Radnaa
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, Australia.,Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
| | - Samantha Sheller-Miller
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
18
|
Secomandi L, Borghesan M, Velarde M, Demaria M. The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions. Hum Reprod Update 2022; 28:172-189. [PMID: 34918084 PMCID: PMC8888999 DOI: 10.1093/humupd/dmab038] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/28/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Advanced maternal age is associated with decreased oocyte quantity and quality as well as uterine and placental dysfunctions. These changes lead to infertility, pregnancy complications and birth defects in the offspring. As the mean age of giving birth is increasing worldwide, prevention of age-associated infertility and pregnancy complications, along with the more frequent use of ART, become extremely important. Currently, significant research is being conducted to unravel the mechanisms underlying female reproductive aging. Among the potential mechanisms involved, recent evidence has suggested a contributing role for cellular senescence, a cellular state of irreversible growth arrest characterized by a hypersecretory and pro-inflammatory phenotype. Elucidating the role of senescence in female reproductive aging holds the potential for developing novel and less invasive therapeutic measures to prevent or even reverse female reproductive aging and increase offspring wellbeing. OBJECTIVE AND RATIONALE The review will summarize the positive and negative implications of cellular senescence in the pathophysiology of the female reproductive organs during aging and critically explore the use of novel senotherapeutics aiming to reverse and/or eliminate their detrimental effects. The focus will be on major senescence mechanisms of the ovaries, the uterus, and the placenta, as well as the potential and risks of using senotherapies that have been discovered in recent years. SEARCH METHODS Data for this review were identified by searches of MEDLINE, PubMed and Google Scholar. References from relevant articles using the search terms 'Cellular Senescence', 'Aging', 'Gestational age', 'Maternal Age', 'Anti-aging', 'Uterus', 'Pregnancy', 'Fertility', 'Infertility', 'Reproduction', 'Implant', 'Senolytic', 'Senostatic', 'Senotherapy' and 'Senotherapeutic' where selected. A total of 182 articles published in English between 2005 and 2020 were included, 27 of which focus on potential senotherapies for reproductive aging. Exclusion criteria were inclusion of the terms 'male' and 'plants'. OUTCOMES Aging is a major determinant of reproductive wellbeing. Cellular senescence is a basic aging mechanism, which can be exploited for therapeutic interventions. Within the last decade, several new strategies for the development and repurposing of drugs targeting senescent cells have emerged, such as modulators of the anti-inflammatory response, oxidative stress, DNA damage, and mitochondria and protein dysfunctions. Several studies of female reproductive aging and senotherapies have been discussed that show promising results for future interventions. WIDER IMPLICATIONS In most countries of the Organization for Economic Co-operation and Development, the average age at which women give birth is above 30 years. Currently, in countries such as the Netherlands, Australia, Spain, Finland, Germany and the UK, birth rates among 30- to 34-year-olds are now higher than in any other age groups. This review will provide new knowledge and scientific advancement on the senescence mechanisms during female reproductive aging, and benefit fundamental and clinical scientists and professionals in the areas of reproduction, cancer, immunobiology and fibrosis.
Collapse
Affiliation(s)
- Laura Secomandi
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| | - Michela Borghesan
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| | - Michael Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, PH 1101, Philippines
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| |
Collapse
|
19
|
Preterm Labor, a Syndrome Attributed to the Combination of External and Internal Factors. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
20
|
Geng Y, Zhao W, Liu W, Tang J, Zhang H, Ke W, Yao R, Xu J, Lin Q, Li Y, Huang J. Term Neonatal Complications During the Second Localized COVID-19 Lockdown and Prolonged Premature Rupture of Membranes at Home Among Nulliparas With Reference Interval for Maternal C-Reactive Protein: A Retrospective Cohort Study. Front Pediatr 2022; 10:787947. [PMID: 35463873 PMCID: PMC9024135 DOI: 10.3389/fped.2022.787947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/21/2022] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE The COVID-19 lockdown extended premature rupture of membranes (PROM) expectant time among nulliparas and increased the risk of term neonatal complications. This study investigated the impact of term nulliparas with PROM delays at home on neonatal outcomes during the COVID-19 lockdown period, considering the clinical diagnostic application of maternal C-reactive protein (CRP). METHODS This study collected 505 term nulliparous women who underwent PROM at home from five provinces in a non-designated hospital of China in 2020. We analyzed PROM maternal information at home and neonatal complications in the COVID-19 regional lockdown and compared related information in the national lockdown. Poisson regression models estimated the correlation of PROM management at home, maternal CRP, and neonatal morbidity. We constructed two diagnostic models: the CRP univariate model, and an assessed cut-off value of CRP in the combined model (CRP with PROM waiting time at home). RESULTS In the regional lockdown, PROM latency at home and the severity of neonatal complications were extended and increased lower than in the nationwide lockdown, but term neonatal morbidity was not reduced in the COVID-19 localized lockdown. Prolonged waiting time at home (≥8.17 h) was associated with increasing maternal CRP values and neonatal morbidity (adjusted risk ratio 2.53, 95% CI, 1.43 to 4.50, p for trend <0.001) in the regional lockdown period. In the combined model, CRP ≥7 mg/L with PROM latency ≥8.17 h at home showed higher diagnostic sensitivity and AUC than only CRP for initial assessing the risk of adverse neonatal complications in COVID-19 regional lockdowns (AUC, 0.714 vs. 0.534; sensitivity, 0.631 vs. 0.156). CONCLUSION The impact of the acute COVID-19 national blockade on the PROM newborns' health could continue to the COVID-19 easing period. Maternal CRP reference interval (≥7 mg/L) would effectively assess the risk of term neonatal morbidity when nulliparas underwent prolonged PROM expectant at home (≥8.17 h) during the second COVID-19 lockdown.
Collapse
Affiliation(s)
- Yang Geng
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Weihua Zhao
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Wenlan Liu
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jie Tang
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Hui Zhang
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Weilin Ke
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Runsi Yao
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Ji Xu
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Qing Lin
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yun Li
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jianlin Huang
- Department of Obstetrics and Gynecology and Center for Perinatal Medical Health, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
21
|
Spencer NR, Radnaa E, Baljinnyam T, Kechichian T, Tantengco OAG, Bonney E, Kammala AK, Sheller-Miller S, Menon R. Development of a mouse model of ascending infection and preterm birth. PLoS One 2021; 16:e0260370. [PMID: 34855804 PMCID: PMC8638907 DOI: 10.1371/journal.pone.0260370] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/08/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Microbial invasion of the intraamniotic cavity and intraamniotic inflammation are factors associated with spontaneous preterm birth. Understanding the route and kinetics of infection, sites of colonization, and mechanisms of host inflammatory response is critical to reducing preterm birth risk. OBJECTIVES This study developed an animal model of ascending infection and preterm birth with live bacteria (E. coli) in pregnant CD-1 mice with the goal of better understanding the process of microbial invasion of the intraamniotic cavity and intraamniotic inflammation. STUDY DESIGN Multiple experiments were conducted in this study. To determine the dose of E. coli required to induce preterm birth, CD-1 mice were injected vaginally with four different doses of E. coli (103, 106, 1010, or 1011 colony forming units [CFU]) in 40 μL of nutrient broth or broth alone (control) on an embryonic day (E)15. Preterm birth (defined as delivery before E18.5) was monitored using live video. E. coli ascent kinetics were measured by staining the E. coli with lipophilic tracer DiD for visualization through intact tissue with an in vivo imaging system (IVIS) after inoculation. The E. coli were also directly visualized in reproductive tissues by staining the bacteria with carboxyfluorescein succinimidyl ester (CFSE) prior to administration and via immunohistochemistry (IHC) by staining tissues with anti-E. coli antibody. Each pup's amniotic fluid was cultured separately to determine the extent of microbial invasion of the intraamniotic cavity at different time points. Intraamniotic inflammation resulting from E. coli invasion was assessed with IHC for inflammatory markers (TLR-4, P-NF-κB) and neutrophil marker (Ly-6G) for chorioamnionitis at 6- and 24-h post-inoculation. RESULTS Vaginally administered E. coli resulted in preterm birth in a dose-dependent manner with higher doses causing earlier births. In ex vivo imaging and IHC detected uterine horns proximal to the cervix had increased E. coli compared to the distal uterine horns. E. coli were detected in the uterus, fetal membranes (FM), and placenta in a time-dependent manner with 6 hr having increased intensity of E. coli positive signals in pups near the cervix and in all pups at 24 hr. Similarly, E. coli grew from the cultures of amniotic fluid collected nearest to the cervix, but not from the more distal samples at 6 hr post-inoculation. At 24 hr, all amniotic fluid cultures regardless of distance from the cervix, were positive for E. coli. TLR-4 and P-NF-κB signals were more intense in the tissues where E. coli was present (placenta, FM and uterus), displaying a similar trend toward increased signal in proximal gestational sacs compared to distal at 6 hr. Ly-6G+ cells, used to confirm chorioamnionitis, were increased at 24 hr compared to 6 hr post-inoculation and control. CONCLUSION We report the development of mouse model of ascending infection and the associated inflammation of preterm birth. Clinically, these models can help to understand mechanisms of infection associated preterm birth, determine targets for intervention, or identify potential biomarkers that can predict a high-risk pregnancy status early in pregnancy.
Collapse
Affiliation(s)
- Nicholas R. Spencer
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Enkhtuya Radnaa
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Tuvshintugs Baljinnyam
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Talar Kechichian
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Ourlad Alzeus G. Tantengco
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Elizabeth Bonney
- Department of Obstetrics and Gynecology, University of Vermont, Burlington, VT, United States of America
| | - Ananth Kumar Kammala
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| |
Collapse
|
22
|
Zhang X, Wei H. Role of Decidual Natural Killer Cells in Human Pregnancy and Related Pregnancy Complications. Front Immunol 2021; 12:728291. [PMID: 34512661 PMCID: PMC8426434 DOI: 10.3389/fimmu.2021.728291] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022] Open
Abstract
Pregnancy is a unique type of immunological process. Healthy pregnancy is associated with a series of inflammatory events: implantation (inflammation), gestation (anti-inflammation), and parturition (inflammation). As the most abundant leukocytes during pregnancy, natural killer (NK) cells are recruited and activated by ovarian hormones and have pivotal roles throughout pregnancy. During the first trimester, NK cells represent up to 50–70% of decidua lymphocytes. Differently from peripheral-blood NK cells, decidual natural killer (dNK) cells are poorly cytolytic, and they release cytokines/chemokines that induce trophoblast invasion, tissue remodeling, embryonic development, and placentation. NK cells can also shift to a cytotoxic identity and carry out immune defense if infected in utero by pathogens. At late gestation, premature activation of NK cells can lead to a breakdown of tolerance of the maternal–fetal interface and, subsequently, can result in preterm birth. This review is focused on the role of dNK cells in normal pregnancy and pathological pregnancy, including preeclampsia, recurrent spontaneous abortion, endometriosis, and recurrent implantation failure. dNK cells could be targets for the treatment of pregnancy complications.
Collapse
Affiliation(s)
- Xiuhong Zhang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, Division of Molecular Medicine, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
23
|
Radnaa E, Richardson LS, Sheller-Miller S, Baljinnyam T, de Castro Silva M, Kumar Kammala A, Urrabaz-Garza R, Kechichian T, Kim S, Han A, Menon R. Extracellular vesicle mediated feto-maternal HMGB1 signaling induces preterm birth. LAB ON A CHIP 2021; 21:1956-1973. [PMID: 34008619 PMCID: PMC8162392 DOI: 10.1039/d0lc01323d] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Preterm birth (PTB; <37 weeks of gestation) impacts ∼11% of all pregnancies and contributes to 1 million neonatal deaths worldwide annually. An understanding of the feto-maternal (F-M) signals that initiate birthing (parturition) at term is critical to design strategies to prevent their premature activation, resulting in PTB. Although endocrine and immune cell signaling are well-reported, fetal-derived paracrine signals capable of transitioning quiescent uterus to an active state of labor are poorly studied. Recent reports have suggested that senescence of the fetal amnion membrane coinciding with fetal growth and maturation generates inflammatory signals capable of triggering parturition. This is by increasing the inflammatory load at the feto-maternal interface (FMi) tissues (i.e., amniochorion-decidua). High mobility group box 1 protein (HMGB1), an alarmin, is one of the inflammatory signals released by senescent amnion cells via extracellular vesicles (exosomes; 40-160 nm). Increased levels of HMGB1 in the amniotic fluid, cord and maternal blood are associated with term and PTB. This study tested the hypothesis that senescent amnion cells release HMGB1, which is fetal signaling capable of increasing FMi inflammation, predisposing them to parturition. To test this hypothesis, exosomes from amnion epithelial cells (AECs) grown under normal conditions were engineered to contain HMGB1 by electroporation (eHMGB1). eHMGB1 was characterized (quantity, size, shape, markers and loading efficiency), and its propagation through FMi was tested using a four-chamber microfluidic organ-on-a-chip device (FMi-OOC) that contained four distinct cell types (amnion and chorion mesenchymal, chorion trophoblast and decidual cells) connected through microchannels. eHMGB1 propagated through the fetal cells and matrix to the maternal decidua and increased inflammation (receptor expression [RAGE and TLR4] and cytokines). Furthermore, intra-amniotic injection of eHMGB1 (containing 10 ng) into pregnant CD-1 mice on embryonic day 17 led to PTB. Injecting carboxyfluorescein succinimidyl ester (CFSE)-labeled eHMGB1, we determined in vivo kinetics and report that eHMGB1 trafficking resulting in PTB was associated with increased FMi inflammation. This study determined that fetal exosome mediated paracrine signaling can generate inflammation and induce parturition. Besides, in vivo functional validation of FMi-OOC experiments strengthens the reliability of such devices to test physiologic and pathologic systems.
Collapse
Affiliation(s)
- Enkhtuya Radnaa
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Lauren S Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA. and Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Tuvshintugs Baljinnyam
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Mariana de Castro Silva
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Ananth Kumar Kammala
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Rheanna Urrabaz-Garza
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Talar Kechichian
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| |
Collapse
|
24
|
Guo M, Lu J, Yu X, Hu X, Hou W, Pang S. The protective role of serum uric acid against premature membrane rupture in gestational diabetes: a cross-sectional study. BMC Endocr Disord 2021; 21:95. [PMID: 33957911 PMCID: PMC8101033 DOI: 10.1186/s12902-021-00736-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/07/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Uric acid has strong antioxidant activity, whereas its oxidative damage is closely related to many diseases. We assessed the association between serum uric acid (SUA) levels and premature rupture of membranes (PROM) in pregnant women with gestational diabetes (GDM) in China. METHODS In this cross-sectional study, a total of 456 pregnant women were enrolled. Anthropometric parameters for pregnant women were collected within 12 weeks of gestation. Weight gain during pregnancy was obtained from the patients' records. GDM was diagnosed according to 75-g oral glucose tolerance tests at the 24-28th week of gestation, and SUA was determined simultaneously. PROM was identified as the natural rupture of foetal membranes before the first stage of labour. Logistic models were fitted to identify the presence of PROM using clinical characteristics with (Model 2) or without serum uric acid (Model 1). RESULTS There were differences in BMI, haemoglobin A1c, fasting blood glucose, 1-h postprandial glucose (PG), 2-h PG, insulin levels, triglycerides,weight gain during pregnancy, the rate of macrosomia, fetus birth weight and PROM between women with and without GDM (all P < 0.05). Furthermore, GDM women with PROM had lower levels of SUA compared to those without PROM (P = 0.030). The odds ratio of PROM decreased with increasing SUA levels. The area under the receiver operating characteristic curves for PROM based on Model 2 was larger than that in Model 1 (0.86 versus 0.71, P < 0.05). CONCLUSION Relatively elevated SUA levels at the 24-28th weeks of gestation were associated with a lower risk of PROM in women with GDM. Therefore, SUA may be a protective factor for PROM in GDM patients. The optimal concentration of uric acid in different diseases and different populations needs to be further studied.
Collapse
Affiliation(s)
- Meixiang Guo
- Department of Endocrinology and Metabolism, Institute/University/Hospital: Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105 Jiefang Road, Jinan City, 250013, Shandong Province, China
- Departments of Endocrinology and Metabolism, Fengxian District Central Hospital, Shanghai, China
| | - Jun Lu
- Departments of Endocrinology and Metabolism, Fengxian District Central Hospital, Shanghai, China
- Department of Endocrinology and Metabolism, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuemei Yu
- Departments of Endocrinology and Metabolism, Fengxian District Central Hospital, Shanghai, China
| | - Xiaowen Hu
- Department of Endocrinology and Metabolism, Institute/University/Hospital: Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105 Jiefang Road, Jinan City, 250013, Shandong Province, China
| | - Wenjing Hou
- Departments of Obstetrics and Gynaecology, Fengxian District Central Hospital, Shanghai, China
| | - Shuguang Pang
- Department of Endocrinology and Metabolism, Institute/University/Hospital: Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105 Jiefang Road, Jinan City, 250013, Shandong Province, China.
- Department of Endocrinology Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
25
|
Prairie E, Côté F, Tsakpinoglou M, Mina M, Quiniou C, Leimert K, Olson D, Chemtob S. The determinant role of IL-6 in the establishment of inflammation leading to spontaneous preterm birth. Cytokine Growth Factor Rev 2021; 59:118-130. [PMID: 33551331 DOI: 10.1016/j.cytogfr.2020.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022]
Abstract
Preterm birth (PTB) and its consequences are a major public health concern as preterm delivery is the main cause of mortality and morbidity at birth. There are many causes of PTB, but inflammation is undeniably associated with the process of premature childbirth and fetal injury. At present, treatments clinically available mostly involve attempt to arrest contractions (tocolytics) but do not directly address upstream maternal inflammation on development of the fetus. One of the possible solutions may lie in the modulation of inflammatory mediators. Of the many pro-inflammatory cytokines involved in the induction of PTB, IL-6 stands out for its pleiotropic effects and its involvement in both acute and chronic inflammation. Here, we provide a detailed review of the effects of IL-6 on the timing of childbirth, its occurrence during PTB and its indissociable roles with associated fetal tissue damage.
Collapse
Affiliation(s)
- Elizabeth Prairie
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - France Côté
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Marika Tsakpinoglou
- Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Michael Mina
- Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada.
| | - Kelycia Leimert
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - David Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada.
| |
Collapse
|
26
|
Richardson L, Kim S, Han A, Menon R. Modeling ascending infection with a feto-maternal interface organ-on-chip. LAB ON A CHIP 2020; 20:4486-4501. [PMID: 33112317 PMCID: PMC7815379 DOI: 10.1039/d0lc00875c] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Maternal infection (i.e., ascending infection) and the resulting host inflammatory response are risk factors associated with spontaneous preterm birth (PTB), a major pregnancy complication. However, the path of infection and its propagation from the maternal side to the fetal side have been difficult to study due to the lack of appropriate in vitro models and limitations of animal models. A better understanding of the propagation kinetics of infectious agents and development of the host inflammatory response at the feto-maternal (amniochorion-decidua, respectively) interface (FMi) is critical in curtailing host inflammatory responses that can lead to PTB. To model ascending infection and determine inflammatory responses at the FMi, we developed a microfluidic organ-on-chip (OOC) device containing primary cells from the FMi (decidua, chorion, and amnion [mesenchyme and epithelium]) and collagen matrix harvested from primary tissue. The FMi-OOC is composed of four concentric circular cell/collagen chambers designed to mimic the thickness and cell density of the FMi in vivo. Each layer is connected by arrays of microchannels filled with type IV collagen to recreate the basement membrane of the amniochorion. Cellular characteristics (viability, morphology, production of nascent collagen, cellular transitions, and migration) in the OOC were similar to those seen in utero, validating the physiological relevance and utility of the developed FMi-OOC. The ascending infection model of the FMi-OOC, triggered by exposing the maternal (decidua) side of the OOC to lipopolysaccharide (LPS, 100 ng mL-1), shows that LPS propagated through the chorion, amnion mesenchyme, and reached the fetal amnion within 72 h. LPS induced time-dependent and cell-type-specific pro-inflammatory cytokine production (24 h decidua: IL-6, 48 h chorion: GM-CSF and IL-6, and 72 h amnion mesenchyme and epithelium: GM-CSF and IL-6). Collectively, this OOC model and study successfully modeled ascending infection, its propagation, and distinct inflammatory response at the FMi indicative of pathologic pathways of PTB. This OOC model provides a novel platform to study physiological and pathological cell status at the FMi, and is expected to have broad utility in the field of obstetrics.
Collapse
Affiliation(s)
- Lauren Richardson
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA
| |
Collapse
|
27
|
Jacobs SO, Sheller-Miller S, Richardson LS, Urrabaz-Garza R, Radnaa E, Menon R. Characterizing the immune cell population in the human fetal membrane. Am J Reprod Immunol 2020; 85:e13368. [PMID: 33145922 DOI: 10.1111/aji.13368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 12/21/2022] Open
Abstract
PROBLEM This study localized CD45+ immune cells and compared changes in their numbers between term, not in labor (TNIL) and term, labor (TL) human fetal membranes. METHOD OF STUDY Fetal membranes (amniochorion) from normal TNIL and TL subjects were analyzed by immunohistochemistry (IHC), immunofluorescence (IF), and flow cytometry for evidence of total (CD45+ ) immune cells as well as innate immune cells (neutrophils, macrophages and NK cells) using specific markers. Fetal origin of immune cells was determined using polymerase chain reaction (PCR) for SRY gene in Y chromosome. RESULTS CD45+ cells were localized in human fetal membranes for both TNIL and TL. A threefold increase in CD45+ cells was seen in TL fetal membranes of (7.73% ± 2.35) compared to TNIL (2.36% ± 0.78). This increase is primarily contributed by neutrophils. Macrophages and NK cells did not change in the membranes between TNIL and TL. Leukocytes of fetal origin are present in the fetal membranes. CONCLUSION The fetal membranes without decidua contain a small proportion of immune cells. Some of these immune cells in the fetal membrane are fetal in origin. There is a moderate increase of immune cells in the fetal membranes at term labor; however, it is unclear whether this is a cause or consequence of labor. Further functional studies are needed to determine their contribution to membrane inflammation associated with parturition.
Collapse
Affiliation(s)
- Sara O Jacobs
- The Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Samantha Sheller-Miller
- The Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Lauren S Richardson
- The Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Rheanna Urrabaz-Garza
- The Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Enkhtuya Radnaa
- The Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ramkumar Menon
- The Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| |
Collapse
|
28
|
Saito Reis CA, Padron JG, Norman Ing ND, Kendal-Wright CE. High-mobility group box 1 is a driver of inflammation throughout pregnancy. Am J Reprod Immunol 2020; 85:e13328. [PMID: 32851715 DOI: 10.1111/aji.13328] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
A proinflammatory response driven by high-mobility group box 1 (HMGB1) is important for the success of both the early stages of pregnancy and parturition initiation. However, the tight regulation of HMGB1 within these two stages is critical, as increased HMGB1 can manifest into pregnancy-related pathologies. Although during the early stages of pregnancy HMGB1 is critical for the development and implantation of the embryo, and uterine decidualization, high levels within the uterine cavity have been linked to pregnancy failure. In addition, chronic inflammation, resultant from increased HMGB1 within the maternal circulation and gestational tissues, also increases the risk for preterm labor, preterm birth, or infant mortality. Due to the link between HMGB1 and several pregnancy pathologies, the possibility of leveraging HMGB1 as a biomarker has been assessed. However, data are limited that demonstrate how known HMGB1 inhibitors could reduce inflammation within pregnancy. Thus, further research is warranted to improve our understanding of the potential of HMGB1 as a therapeutic target to reduce inflammation within pregnancy. This review aims to describe what is understood about the role of HMGB1 that drives inflammation throughout pregnancy and highlight its potential as a biomarker and therapeutic target within this context.
Collapse
Affiliation(s)
- Chelsea A Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA
| | - Justin G Padron
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoā, Honolulu, HI, USA
| | - Nainoa D Norman Ing
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA
| | - Claire E Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA.,Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoā, Honolulu, HI, USA.,Obstetrics, Gynecology and Women's Health, John A. Burns School of Medicine, University of Hawai'I at Manoā, Honolulu, HI, USA
| |
Collapse
|
29
|
Menon R, Peltier MR. Novel Insights into the Regulatory Role of Nuclear Factor (Erythroid-Derived 2)-Like 2 in Oxidative Stress and Inflammation of Human Fetal Membranes. Int J Mol Sci 2020; 21:E6139. [PMID: 32858866 PMCID: PMC7503839 DOI: 10.3390/ijms21176139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/16/2022] Open
Abstract
Fetal membrane dysfunction in response to oxidative stress (OS) is associated with adverse pregnancy outcomes. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is one of the regulators of innate OS response. This study evaluated changes in Nrf2 expression and its downstream targets heme oxygenase (HO-1) and peroxisome proliferator-activated receptor gamma (PPARγ) in fetal membranes during OS and infection in vitro. Furthermore, we tested the roles of sulforaphane (SFN; an extract from cruciferous vegetables) and trigonelline (TRN; an aromatic compound in coffee) in regulating Nrf2 and its targets. Fetal membranes (n = 6) collected at term were placed in an organ explant system were treated with water-soluble cigarette smoke extract (CSE), an OS inducer (1:10), and lipopolysaccharide (LPS; 100 ng/mL). Nrf2 expression, expression, its enhancement by sulforaphane (SFN, 10 µM/mL) and down regulation by TRN (10uM/mL) was determined by western blots. Expression of Nrf2 response elements PPARγ (western) heme oxygenase (HO-1), and IL-6 were quantified by ELISA. CSE and LPS treatment of fetal membranes increased nrf2, but reduced HO-1 and PPARγ and increased IL-6. Co-treatment of SFN, but not with TRN, with CSE and LPS increased Nrf2 substantially, as well as increased HO-1 and PPARγ and reduced IL-6 expression. Risk factor-induced Nrf2 increase is insufficient to generate an antioxidant response in fetal membranes. Sulforaphane may enhance innate antioxidant and anti-inflammatory capacity by increasing NRF-2 expression.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Morgan R Peltier
- Department of Foundations of Medicine, New York University-Long Island School of Medicine, Mineola, NY 11501, USA;
- Department of Obstetrics and Gynecology, New York University-Long Island School of Medicine, Mineola, NY 11501, USA
| |
Collapse
|
30
|
Menon R, Behnia F, Polettini J, Richardson LS. Novel pathways of inflammation in human fetal membranes associated with preterm birth and preterm pre-labor rupture of the membranes. Semin Immunopathol 2020; 42:431-450. [PMID: 32785751 DOI: 10.1007/s00281-020-00808-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Spontaneous preterm birth (PTB) and preterm pre-labor rupture of the membranes (pPROM) are major pregnancy complications. Although PTB and pPROM have common etiologies, they arise from distinct pathophysiologic pathways. Inflammation is a common underlying mechanism in both conditions. Balanced inflammation is required for fetoplacental growth; however, overwhelming inflammation (physiologic at term and pathologic at preterm) can lead to term and preterm parturition. A lack of effective strategies to control inflammation and reduce the risk of PTB and pPROM suggests that there are several modes of the generation of inflammation which may be dependent on the type of uterine tissue. The avascular fetal membrane (amniochorion), which provides structure, support, and protection to the intrauterine cavity, is one of the key contributors of inflammation. Localized membrane inflammation helps tissue remodeling during pregnancy. Two unique mechanisms that generate balanced inflammation are the progressive development of senescence (aging) and cyclic cellular transitions: epithelial to mesenchymal (EMT) and mesenchymal to epithelial (MET). The intrauterine build-up of oxidative stress at term or in response to risk factors (preterm) can accelerate senescence and promote a terminal state of EMT, resulting in the accumulation of inflammation. Inflammation degrades the matrix and destabilizes membrane function. Inflammatory mediators from damaged membranes are propagated via extracellular vesicles (EV) to maternal uterine tissues and transition quiescent maternal uterine tissues into an active state of labor. Membrane inflammation and its propagation are fetal signals that may promote parturition. This review summarizes the mechanisms of fetal membrane cellular senescence, transitions, and the generation of inflammation that contributes to term and preterm parturitions.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA.
| | - Faranak Behnia
- Department of Obstetrics, Gynecology, and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, UT Health, Houston, Texas, USA
| | - Jossimara Polettini
- Universidade Federal da Fronteira Sul, Campus Passo Fundo, Rua Capitão Araujo, 20, Centro, Passo Fundo, Rio Grande do Sul, Brazil
| | - Lauren S Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA
| |
Collapse
|
31
|
Fedorka CE, Murase H, Loux SC, Loynachan AT, Walker OF, Squires EL, Ball BA, Troedsson MHT. The Effect of Mycobacterium Cell Wall Fraction on Histologic, Immunologic, and Clinical Parameters of Postpartum Involution in the Mare. J Equine Vet Sci 2020; 90:103013. [PMID: 32534779 DOI: 10.1016/j.jevs.2020.103013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 11/13/2022]
Abstract
Maintaining yearly foal production is important for the economic success of the broodmare, and this requires breeding to occur as quickly postpartum as possible. The initial postpartum estrus occurs within 5-20 days postpartum, whereas the uterus is still undergoing repair from tissue alterations during pregnancy and parturition, a process known as involution. Attempts have been made to hasten this process, but with minimal success. Mycobacterium cell wall fraction (MCWF) is an immunomodulator that has been shown to reduce bacterial growth and alter aspects of the immune response to breeding, but it is unknown if MCWF hastens the process of involution. Therefore, the objectives of this study were to (1) investigate the effect of MCWF on tissue remodeling, (2) assess the effect of MCWF on the local immune system of the uterus, and (3) determine the optimal treatment interval needed for these processes to occur. We hypothesize that repeated treatments of MCWF postpartum will hasten the process of involution. To study this, 16 pregnant mares of mixed breeds were evaluated postpartum. Control mares (n = 4) received 1.5 mL lactated Ringer's solution intravenously on Day 1 (Day 0 = day of parturition) postpartum and again on Day 7, whereas treated mares either received 1.5 mL Settle intravenously on Day 1 and Day 7 (TX1; n = 6) or 1.5 mL Settle intravenously on Day 1 and then every 3 days until ovulation was detected (TX2; n = 6) and then evaluated until 15 days postpartum. Mares were assessed every 3 days for clinical, immunologic, and histologic parameters. Clinical parameters were assessed with transrectal ultrasonography and included ovarian activity, uterine fluid retention, and measurement of the uterine diameter, in addition to endometrial culture. Immunologic parameters included endometrial biopsies for quantitative polymerase chain reaction for expression of various cytokines (interleukin [IL]-1β, IL-1RN, IL-4, IL-6, IL-8, IL-10, tumor necrosis factor [TNF], interferon [IFN]-γ, and granulocyte-macrophage colony-stimulating factor) in addition to endometrial cytology. Formalin-fixed endometrial biopsies were histologically assessed for the retention of microcaruncles, dilation of endometrial glands, and inflammation of the mucosa, stratum compactum, and spongiosum. Statistics were performed using SAS 9.4, using a mixed model for repeated measures with mare and treatment as a random effect. All post-hoc analysis was done using a Tukey's honestly significant difference test. Involution was considered complete by Day 15 postpartum in all mares, and the day postpartum had a significant effect on almost all parameters investigated, indicating the immunologic process of involution. Treatment with MCWF decreased the magnitude of bacterial growth in addition to time to negative culture. In addition, MCWF increased the expression of IL-1β, IFNγ, and TNF. Although minimal treatment effect was noted histologically, a decrease in mucosal inflammation was seen in MCWF-treated mares. In conclusion, involution appears to be influenced by the immune system. In addition, MCWF appears to have a bactericidal effect on the postpartum mare, and this may be because of an increase in proinflammatory cytokines. It is unknown if this bactericidal property will improve fertility on the first estrous cycle postpartum, and future studies are needed to determine this.
Collapse
Affiliation(s)
- Carleigh E Fedorka
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY.
| | - Harutake Murase
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY; Equine Science Division, Hidaka Training and Research Center, Japan Racing Association, Hokkaido, Japan
| | - Shavahn C Loux
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY
| | - Alan T Loynachan
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY
| | - Olivia F Walker
- College of Veterinary Medicine, Lincoln Memorial University, Harrogate, TN
| | - Edward L Squires
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY
| | - Barry A Ball
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY
| | - Mats H T Troedsson
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY
| |
Collapse
|