1
|
Cheng J, Jia X, Yang L, Zhang S, Chen Z, Gui Q, Li T, Pu Z, Qi H, Zhang J. New therapeutic target NCF1-directed multi-bioactive conjugate therapies prevent preterm birth and adverse pregnancy outcomes. Sci Bull (Beijing) 2024; 69:2604-2621. [PMID: 39030102 DOI: 10.1016/j.scib.2024.03.064] [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: 11/08/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 07/21/2024]
Abstract
Preterm birth (PTB) is a leading cause of neonatal morbidity and mortality worldwide, yet the cellular and molecular mechanisms driving this condition remain undeciphered, thus limiting discovery of new therapies. In-depth analyses of human and mouse tissues associated with PTB, in combination with cellular studies, indicated that aberrantly high-expressed neutrophil cytoplasmic factor (NCF) 1 leads to oxidative distress, recruitment, and pro-inflammatory activation of neutrophils and macrophages, while sequentially overexpressed pro-inflammatory mediators induce contractions of uterine smooth muscle cells (USMCs) as well as apoptosis of USMCs and amniotic epithelial cells, thereby causing PTB. According to these new findings, we rationally engineered an amphiphilic macromolecular conjugate LPA by covalently integrating low-molecular-weight heparin, a reactive oxygen species-responsive/scavenging component, and an anti-inflammatory peptide. This bioengineered macromolecular conjugate can self-assemble into multi-bioactive nanoparticles (LPA NP). In a mouse model of PTB, LPA NP effectively delayed PTB and inhibited adverse pregnancy outcomes, by regulating NCF1-mediated oxidative-inflammatory cascades, i.e., attenuating oxidative stress, inhibiting inflammatory cell activation, reducing local inflammation, and decreasing contraction/apoptosis of myometrial cells. Packaging LPA NP into temperature-responsive, self-healing, and bioadhesive hydrogel further potentiated its in vivo efficacies after intravaginal delivery, by prolonging retention time, sustaining nanotherapy release, and increasing bioavailability in the placenta/uterus. Importantly, both the conjugate/nanotherapy and hydrogel formulations exhibited excellent safety profiles in pregnant mice, with negligible side effects on the mother and offspring.
Collapse
Affiliation(s)
- Juan Cheng
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoyan Jia
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Limei Yang
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Siqi Zhang
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhiyu Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qian Gui
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Ting Li
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Zedan Pu
- Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Hongbo Qi
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Yu-Yue Pathology Scientific Research Center, Chongqing 400039, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
2
|
Cocchi E, Bellisario V, Cresi F, Plazzotta C, Cassardo C, Siniscalco C, Peruzzi L, Bono R. Air Pollution and Aeroallergens as Possible Triggers in Preterm Birth Delivery. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1610. [PMID: 36674364 PMCID: PMC9860587 DOI: 10.3390/ijerph20021610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
Preterm birth (PTB) identifies infants prematurely born <37 weeks/gestation and is one of the main causes of infant mortality. PTB has been linked to air pollution exposure, but its timing is still unclear and neglects the acute nature of delivery and its association with short-term effects. We analyzed 3 years of birth data (2015−2017) in Turin (Italy) and the relationships with proinflammatory chemicals (PM2.5, O3, and NO2) and biological (aeroallergens) pollutants on PTB vs. at-term birth, in the narrow window of a week before delivery. A tailored non-stationary Poisson model correcting for seasonality and possible confounding variables was applied. Relative risk associated with each pollutant was assessed at any time lag between 0 and 7 days prior to delivery. PTB risk was significantly associated with increased levels of both chemical (PM2.5, RR = 1.023 (1.003−1.043), O3, 1.025 (1.001−1.048)) and biological (aeroallergens, RR ~ 1.01 (1.0002−1.016)) pollutants in the week prior to delivery. None of these, except for NO2 (RR = 1.01 (1.002−1.021)), appeared to play any role on at-term delivery. Pollutant-induced acute inflammation eliciting delivery in at-risk pregnancies may represent the pathophysiological link between air pollution and PTB, as testified by the different effects played on PTB revealed. Further studies are needed to better elucidate a possible exposure threshold to prevent PTB.
Collapse
Affiliation(s)
- Enrico Cocchi
- Department of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy
- Pediatric Nephrology Unit, Regina Margherita Children’s Hospital, 10126 Turin, Italy
- Pediatric Residency School, University of Turin, 10126 Turin, Italy
| | - Valeria Bellisario
- Department of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy
- Biostatistics Residency School, University of Turin, 10126 Turin, Italy
| | - Francesco Cresi
- Neonatal Intensive Care Unit, Sant’Anna Obstetric Gynecological Hospital, 10126 Turin, Italy
| | - Claudio Plazzotta
- Neonatal Intensive Care Unit, Sant’Anna Obstetric Gynecological Hospital, 10126 Turin, Italy
| | | | - Consolata Siniscalco
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Licia Peruzzi
- Pediatric Nephrology Unit, Regina Margherita Children’s Hospital, 10126 Turin, Italy
| | - Roberto Bono
- Department of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy
| |
Collapse
|
3
|
African American Women with Cardiometabolic Complications of Pregnancy Have Decreased Serum Abundance of Specialized Pro-Resolving Lipid Mediators and Endocannabinoids. Nutrients 2022; 15:nu15010140. [PMID: 36615797 PMCID: PMC9823622 DOI: 10.3390/nu15010140] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
African American (AA) women experience higher rates of maternal morbidity and mortality compared to US women of other racial/ ethnic groups. Cardiometabolic complications of pregnancy (including gestational diabetes, gestational hypertension, and preeclampsia) are leading contributors to maternal morbidity and mortality. Marked changes in circulating lipids are known to accompany cardiometabolic complications of pregnancy. Serum concentrations of docosahexaenoic acid (DHA) have been shown to be inversely correlated with risk for preeclampsia. DHA is a biosynthetic precursor of a class of specialized pro-resolving mediators (SPMs), resolvins, that have anti-inflammatory properties and are also associated with hypertensive disorders of pregnancy. We employed targeted lipidomics to characterize the distribution of DHA-containing phospholipids and SPMs in maternal serum collected in early and late pregnancy (8-14 weeks and 24-30 weeks gestation, respectively) to identify key lipids that are dysregulated during pregnancy in AA women who develop cardiometabolic complications. We identified a lipid signature in early pregnancy serum samples of AA women that is predictive of cardiometabolic complications of pregnancy with 74% accuracy. These are Resolvin D1, Resolvin E1, 2-AG, PGE2-glyerol ester, and 36:6 PC. These findings suggest that there are blood-based markers detectable in early pregnancy that can potentially identify persons at risk and tailor clinical interventions.
Collapse
|
4
|
Takahashi Y, Saito M, Usuda H, Takahashi T, Watanabe S, Hanita T, Sato S, Kumagai Y, Koshinami S, Ikeda H, Carter S, Fee EL, Furfaro L, Chemtob S, Keelan J, Olson D, Yaegashi N, Newnham JP, Jobe AH, Kemp MW. Direct administration of the non-competitive interleukin-1 receptor antagonist rytvela transiently reduced intrauterine inflammation in an extremely preterm sheep model of chorioamnionitis. PLoS One 2021; 16:e0257847. [PMID: 34559862 PMCID: PMC8462743 DOI: 10.1371/journal.pone.0257847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/10/2021] [Indexed: 11/21/2022] Open
Abstract
Background Intraamniotic inflammation is associated with up to 40% of preterm births, most notably in deliveries occurring prior to 32 weeks’ gestation. Despite this, there are few treatment options allowing the prevention of preterm birth and associated fetal injury. Recent studies have shown that the small, non-competitive allosteric interleukin (IL)-1 receptor inhibitor, rytvela, may be of use in resolving inflammation associated with preterm birth (PTB) and fetal injury. We aimed to use an extremely preterm sheep model of chorioamnionitis to investigate the anti-inflammatory efficacy of rytvela in response to established intra-amniotic (IA) lipopolysaccharide (LPS) exposure. We hypothesized that rytvela would reduce LPS-induced IA inflammation in amniotic fluid (AF) and fetal tissues. Methods Sheep with a single fetus at 95 days gestation (estimated fetal weight 1.0 kg) had surgery to place fetal jugular and IA catheters. Animals were recovered for 48 hours before being randomized to either: i) IA administration of 2 ml saline 24 hours before 2 ml IA and 2 ml fetal intravenous (IV) administration of saline (Saline Group, n = 7); ii) IA administration of 10 mg LPS in 2 ml saline 24 hours before 2 ml IA and 2 ml fetal IV saline (LPS Group, n = 10); 3) IA administration of 10 mg LPS in 2 ml saline 24 hours before 0.3 mg/fetal kg IA and 1 mg/fetal kg fetal IV rytvela in 2 ml saline, respectively (LPS + rytvela Group, n = 7). Serial AF samples were collected for 120 h. Inflammatory responses were characterized by quantitative polymerase chain reaction (qPCR), histology, fluorescent immunohistochemistry, enzyme-linked inmmunosorbent assay (ELISA), fluorescent western blotting and blood chemistry analysis. Results LPS-treated animals had endotoxin and AF monocyte chemoattractant protein (MCP)-1 concentrations that were significantly higher at 24 hours (immediately prior to rytvela administration) relative to values from Saline Group animals. Following rytvela administration, the average MCP-1 concentrations in the AF were significantly lower in the LPS + rytvela Group relative to in the LPS Group. In delivery samples, the expression of IL-1β in fetal skin was significantly lower in the LPS + rytvela Group compared to the LPS Group. Conclusion A single dose of rytvela was associated with partial, modest inhibition in the expression of a panel of cytokines/chemokines in fetal tissues undergoing an active inflammatory response.
Collapse
Affiliation(s)
- Yuki Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- * E-mail:
| | - Masatoshi Saito
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shimpei Watanabe
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Takushi Hanita
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shinichi Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yusaku Kumagai
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shota Koshinami
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hideyuki Ikeda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sean Carter
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Erin L. Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Lucy Furfaro
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Canada
| | - Jeffrey Keelan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - David Olson
- Department of Obstetrics and Gynaecology, University of Alberta, Alberta, Canada
| | - Nobuo Yaegashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - John P. Newnham
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Alan H. Jobe
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Cincinnati Children’s Hospital Medical Centre, Cincinnati, OH, United States of America
| | - Matthew W. Kemp
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
5
|
Floridia M, Pinnetti C, Masuelli G, Spinillo A, Savasi VM, Liuzzi G, Degli Antoni AM, Sansone M, Guaraldi G, Dalzero S, Maso G, Francisci D, Sterrantino G, Ravizza M, Tamburrini E. CD4/CD8 ratio in pregnant women with HIV and its association with pregnancy outcome: data from a national study in Italy. Infection 2021; 49:955-964. [PMID: 33963983 DOI: 10.1007/s15010-021-01619-4] [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: 02/08/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE To evaluate associations between CD4/CD8 ratio and pregnancy outcomes in women with HIV. METHODS We evaluated, in a national study of pregnant women with HIV receiving antiretroviral treatment (ART), values of CD4/CD8 ratio at entry in pregnancy, changes between first and third trimester, and possible associations with preterm delivery, low birthweight, and HIV-RNA < 50 copies/ml at third trimester in univariate and multivariate analyses. RESULTS Among 934 women, 536 (57.4%) were already on ART at conception. CD4/CD8 ratio (baseline value 0.570) increased significantly between the first and third trimesters, particularly in women who started ART in pregnancy (+ 0.163, vs. + 0.036 in women already on treatment). The rate of CD4/CD8 ratio normalization, defined by achieving a ratio ≥ 1 at the third trimester, was 13.2%. In multivariable analyses, women who entered pregnancy with a CD4/CD8 ratio < 0.3, compared to women with ratio ≥ 1, were almost four-times less likely to have third-trimester HIV-RNA < 50 copies/ml (AOR 0.258, 95%CI 0.111-0.601), and more than twice as likely to have preterm delivery (AOR 2.379, 95%CI 1.082-5.232). For preterm delivery, also a baseline CD4/CD8 ratio between 0.3 and 0.45 was significantly associated with an increased risk (AOR: 3.415, 95%CI 1.690-6.900). CONCLUSION We described for the first time independent associations of low CD4/CD8 ratio with preterm delivery and HIV-RNA suppression.
Collapse
Affiliation(s)
- Marco Floridia
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | | | - Giulia Masuelli
- Department of Obstetrics and Neonatology, Città della Salute e della Scienza Hospital, and University of Turin, Turin, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynaecology, IRCCS S. Matteo, Pavia, Italy
| | - Valeria M Savasi
- Department of Obstetrics and Gynaecology, Luigi Sacco Hospital and University of Milan, Milan, Italy
| | | | - Anna M Degli Antoni
- Department of Infectious Diseases and Hepatology, Azienda Ospedaliera di Parma, Parma, Italy
| | - Matilde Sansone
- Department of Neurosciences, Reproductive and Dentistry Science, University Federico II, Naples, Italy
| | - Giovanni Guaraldi
- Department of Medical Specialties, Infectious Diseases Clinic, University of Modena and Reggio Emilia, Modena, Italy
| | - Serena Dalzero
- Department of Obstetrics and Gynaecology, DMSD San Paolo Hospital Medical School, University of Milan, Milan, Italy
| | - Gianpaolo Maso
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Daniela Francisci
- Clinic of Infectious Diseases, Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Gaetana Sterrantino
- SOD Malattie Infettive e Tropicali, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Marina Ravizza
- Department of Obstetrics and Gynaecology, DMSD San Paolo Hospital Medical School, University of Milan, Milan, Italy
| | - Enrica Tamburrini
- Department of Infectious Diseases, Catholic University and Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | |
Collapse
|
6
|
Huang HL, Ngam PI, Tan KM, Ng DCE, Lim ST, Chan JY. The exact Deauville score, NABS score and high SUVmax predicts outcome in extranodal natural killer/T-cell lymphoma. Ann Nucl Med 2021; 35:557-568. [PMID: 33683545 DOI: 10.1007/s12149-021-01598-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/14/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Natural killer T-cell lymphoma (NKTCL) is an aggressive type of non-Hodgkin's lymphoma. While FDG-PET/CT imaging has been increasingly utilized for disease assessment, its prognostic value and potential utility in NKTCL patient stratification remain controversial. We aim to investigate the prognostic utility of FDG-PET/CT and its role in complementing clinical indices. METHODS We conducted a retrospective review of 72 patients from a tertiary National Cancer Centre with biopsy-proven NKTCL and available FDG-PET/CT data (either baseline, end of treatment or both). Survival analysis was performed using the Kaplan-Meier method and multivariable Cox proportional regression. RESULTS High initial SUVmax was significantly associated with advanced Ann-Arbor stage (p = 0.0352), elevated LDH levels (p = 0.0059) and plasma EBV DNA detection (p = 0.0278). SUVmax correlated with worse progression-free survival (PFS) (HR 3.68, 95% CI 1.56-8.69, p = 0.0030) and a trend toward worse overall survival (OS) (HR 2.06, 95% CI 0.95-4.45, p = 0.0676). End of treatment Deauville scores of 4-5, as compared to scores of 1-3, was associated with worse PFS (HR 2.72, 95% CI 1.04-7.12, p = 0.0419). Notably, while all patients with scores of 5 developed progressive disease, only 2 of 5 patients with scores of 4 eventually relapsed. Clinical indices (NABS score) were still able to stratify survival outcomes regardless of end-of-treatment Deauville scores. CONCLUSIONS A Deauville score of 5 is more diagnostic of true disease progression than a score of 4, and NABS score may be used in patients who achieve Deauville scores of 1-3 for further risk stratification. A higher SUVmax at baseline portends a worse prognosis in NKTCL.
Collapse
Affiliation(s)
- Hian Liang Huang
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
| | - Pei Ing Ngam
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - Khee Ming Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Outram Road, Singapore, 169608, Singapore
| | - David Chee Eng Ng
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Soon Thye Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Outram Road, Singapore, 169608, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Outram Road, Singapore, 169608, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
7
|
Triggs T, Kumar S, Mitchell M. Experimental drugs for the inhibition of preterm labor. Expert Opin Investig Drugs 2020; 29:507-523. [PMID: 32290715 DOI: 10.1080/13543784.2020.1752661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Preterm birth is the leading cause of neonatal morbidity and mortality globally and poses a substantial economic burden. Consequently, there is a need for the identification of therapeutic targets and novel experimental drugs for the inhibition of preterm labor to improve neonatal outcomes. AREAS COVERED The authors review the pathophysiology of labor and the inflammatory pathways underpinning it. The interruption of these pathways forms the basis of therapeutic targets to inhibit preterm labor. Current drugs available for the treatment of preterm labor are reviewed, followed by experimental drugs including toll-like receptor 4 (TLR-4) antagonists, cytokine suppressive anti-inflammatory drugs (CSAIDs), N-acetyl cysteine (NAC), Sulfasalazine (SSZ), tumor necrosis factor-alpha (TNF-α) antagonists, interleukin-1 receptor (IL-1) inhibitors, omega-3 polyunsaturated fatty acids and lipid metabolites, and the polyphenols. EXPERT OPINION A number of new therapeutic strategies for the prevention of preterm labor are being investigated. These have the potential to improve neurodevelopmental outcomes and survival in babies born preterm, reducing the economic and healthcare costs of caring for the complex needs of these children in the immediate and long term. It is likely that over the next decade there will be a new treatment option that targets the pathological inflammatory processes involved in preterm labor.
Collapse
Affiliation(s)
- Tegan Triggs
- Women's & Newborn Services, Royal Brisbane and Women's Hospital , Herston, Queensland, Australia
| | - Sailesh Kumar
- Women's & Newborn Services, Royal Brisbane and Women's Hospital , Herston, Queensland, Australia
| | - Murray Mitchell
- Women's & Newborn Services, Royal Brisbane and Women's Hospital , Herston, Queensland, Australia
| |
Collapse
|
8
|
Chin PY, Dorian C, Sharkey DJ, Hutchinson MR, Rice KC, Moldenhauer LM, Robertson SA. Toll-Like Receptor-4 Antagonist (+)-Naloxone Confers Sexually Dimorphic Protection From Inflammation-Induced Fetal Programming in Mice. Endocrinology 2019; 160:2646-2662. [PMID: 31504393 PMCID: PMC6936318 DOI: 10.1210/en.2019-00493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022]
Abstract
Inflammation elicited by infection or noninfectious insults during gestation induces proinflammatory cytokines that can shift the trajectory of development to alter offspring phenotype, promote adiposity, and increase susceptibility to metabolic disease in later life. In this study, we use mice to investigate the utility of a small molecule Toll-like receptor (TLR)4 antagonist (+)-naloxone, the nonopioid isomer of the opioid receptor antagonist (-)-naloxone, for mitigating altered fetal metabolic programming induced by a modest systemic inflammatory challenge in late gestation. In adult progeny exposed to lipopolysaccharide (LPS) challenge in utero, male but not female offspring exhibited elevated adipose tissue, reduced muscle mass, and elevated plasma leptin at 20 weeks of age. Effects were largely reversed by coadministration of (+)-naloxone following LPS. When given alone without LPS, (+)-naloxone elicited accelerated postweaning growth and elevated muscle and fat mass in adult male but not female offspring. LPS induced expression of inflammatory cytokines Il1a, Il1b, Il6, Tnf, and Il10 in fetal brain, placental, and uterine tissues, and (+)-naloxone suppressed LPS-induced cytokine expression. Fetal sex-specific regulation of cytokine expression was evident, with higher Il1a, Il1b, Il6, and Il10 induced by LPS in tissues associated with male fetuses, and greater suppression by (+)-naloxone of Il6 in females. These data demonstrate that modulating TLR4 signaling with (+)-naloxone provides protection from inflammatory diversion of fetal developmental programming in utero, associated with attenuation of gestational tissue cytokine expression in a fetal sex-specific manner. The results suggest that pharmacologic interventions targeting TLR4 warrant evaluation for attenuating developmental programming effects of fetal exposure to maternal inflammatory mediators.
Collapse
Affiliation(s)
- Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Camilla Dorian
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Mark R Hutchinson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, South Australia, Australia
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Correspondence: Sarah A. Robertson, PhD, Robinson Research Institute and the Adelaide Medical School, University of Adelaide, North Terrace, Adelaide, South Australia 5005, Australia. E-mail:
| |
Collapse
|
9
|
O'Brien JM, Santolaya JL, Palomares K, Blitzer D, Santolaya-Forgas J. Association of histological chorioamnionitis and magnesium sulfate treatment in singleton and dichorionic twin pregnancies with preterm premature rupture of membranes: preliminary observations. J Perinat Med 2018; 46:839-844. [PMID: 28873067 DOI: 10.1515/jpm-2017-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 07/25/2017] [Indexed: 12/30/2022]
Abstract
Objective To evaluate the possible association between antenatal magnesium sulfate treatment with histological chorioamnionitis in patients with singleton or dichorionic twins that had preterm premature rupture of the membranes. Methods This was an observational study performed in patients admitted to the hospital with rupture of membranes before 34 weeks' gestation. The primary outcome was histological chorioamnionitis and the primary predictor was antenatal magnesium sulfate treatment. A logistic regression model was used without consideration of other antenatal medical treatments. Results Among 107 patients with preterm deliveries, 57 were admitted to the hospital before 34 weeks' gestation with preterm premature rupture of membranes. Fifty-cases were excluded from the analysis because they were admitted after 34 weeks' gestation, delivered before 24 weeks' gestation or had intrauterine fetal demise or monochorionic twins. The logistic regression analysis adjusting for maternal age, gravidity, parity, multiple gestation, gestational age at delivery, and birthweight, indicated that patients with singleton pregnancies and histological chorioamnionitis had received magnesium sulfate antenatally more frequently (χ2=6.46; P=0.01). The association between histological chorioamnionitis and magnesium sulfate treatment was not found among patients with dichorionic twin pregnancies with one intact gestational sac. Conclusions In this cohort of patients with preterm premature rupture of membranes admitted to the hospital before 34 week's gestation, those with singleton pregnancies treated antenatally with magnesium sulfate for neonatal neuroprotection had a greater rate of histological chorioamnionitis.
Collapse
Affiliation(s)
- James M O'Brien
- Penn State Milton S. Hershey Medical Center, Division of Maternal Fetal Medicine, Hershey, PA, USA
| | - Jacobo L Santolaya
- Children's Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA, USA
| | - Kristy Palomares
- Saint Peters University Hospital, Division of Maternal Fetal Medicine, New Brunswick, NJ, USA
| | - David Blitzer
- MedStar Union Memorial Hospital, Department of Surgery, Baltimore, MD, USA
| | - Joaquin Santolaya-Forgas
- University of Florida, College of Medicine, Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, PO Box 100294, Gainesville, FL 100294, USA, Tel.: +(352) 273-7562, Fax: +(352) 294-5094
- Jersey Shore University Medical Center, Perinatal Institute, Neptune, NJ, USA
| |
Collapse
|
10
|
Maduro MR. A Potentially New Tocolytic Agent. Reprod Sci 2018; 25:1529-1530. [PMID: 30326820 DOI: 10.1177/1933719118802730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Maduro MR. In the Spotlight. Reprod Sci 2018. [DOI: 10.1177/1933719118785520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
12
|
Garcia-Flores V, Romero R, Miller D, Xu Y, Done B, Veerapaneni C, Leng Y, Arenas-Hernandez M, Khan N, Panaitescu B, Hassan SS, Alvarez-Salas LM, Gomez-Lopez N. Inflammation-Induced Adverse Pregnancy and Neonatal Outcomes Can Be Improved by the Immunomodulatory Peptide Exendin-4. Front Immunol 2018; 9:1291. [PMID: 29967606 PMCID: PMC6015905 DOI: 10.3389/fimmu.2018.01291] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/23/2018] [Indexed: 12/14/2022] Open
Abstract
Preterm birth is the leading cause of neonatal morbidity and mortality worldwide. Inflammation is causally linked to preterm birth; therefore, finding an intervention that dampens maternal and fetal inflammatory responses may provide a new strategy to prevent adverse pregnancy and neonatal outcomes. Using animal models of systemic maternal inflammation [intraperitoneal injection of lipopolysaccharide (LPS)] and fetal inflammation (intra-amniotic administration of LPS), we found that (1) systemic inflammation induced adverse pregnancy and neonatal outcomes by causing a severe maternal cytokine storm and a mild fetal cytokine response; (2) fetal inflammation induced adverse pregnancy and neonatal outcomes by causing a mild maternal cytokine response and a severe fetal cytokine storm; (3) exendin-4 (Ex4) treatment of dams with systemic inflammation or fetal inflammation improved adverse pregnancy outcomes by modestly reducing the rate of preterm birth; (4) Ex4 treatment of dams with systemic, but not local, inflammation considerably improved neonatal outcomes, and such neonates continued to thrive; (5) systemic inflammation facilitated the diffusion of Ex4 through the uterus and the maternal-fetal interface; (6) neonates born to Ex4-treated dams with systemic inflammation displayed a similar cytokine profile to healthy control neonates; and (7) treatment with Ex4 had immunomodulatory effects by inducing an M2 macrophage polarization and increasing anti-inflammatory neutrophils, as well as suppressing the expansion of CD8+ regulatory T cells, in neonates born to dams with systemic inflammation. Collectively, these results provide evidence that dampening maternal systemic inflammation through novel interventions, such as Ex4, can improve the quality of life for neonates born to women with this clinical condition.
Collapse
Affiliation(s)
- Valeria Garcia-Flores
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States.,Departamento de Genética y Biología Molecular, Cinvestav, Mexico City, Mexico
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Derek Miller
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yi Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bogdan Done
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Chharitha Veerapaneni
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yaozhu Leng
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States.,Departamento de Biomedicina Molecular, Cinvestav, Mexico City, Mexico
| | - Nabila Khan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bogdan Panaitescu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sonia S Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | | | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Detroit, MI, United States.,Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
13
|
Gomez-Lopez N, Romero R, Xu Y, Plazyo O, Unkel R, Leng Y, Than NG, Chaiworapongsa T, Panaitescu B, Dong Z, Tarca AL, Abrahams VM, Yeo L, Hassan SS. A Role for the Inflammasome in Spontaneous Preterm Labor With Acute Histologic Chorioamnionitis. Reprod Sci 2017; 24:1382-1401. [PMID: 28122480 PMCID: PMC5933090 DOI: 10.1177/1933719116687656] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inflammasomes are cytosolic multiprotein complexes that orchestrate inflammation in response to pathogens and endogenous danger signals. Herein, we determined whether the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis (1) express major inflammasome components; (2) express caspase (CASP)-1 and CASP-4 as well as their active forms; (3) exhibit apoptosis-associated speck-like protein containing a CARD (ASC)/CASP-1 complex formation; and (4) release the mature forms of interleukin (IL)-1β and IL-18. We utilized quantitative reverse transcription polymerase chain reaction, enzyme-linked immunosorbent assay, immunoblotting, and immunohistochemistry to determine the messenger RNA (mRNA) and protein expression of major inflammasome components, nucleotide-binding oligomerization domain (NOD) proteins, and the pro- and mature/active forms of CASP-1, CASP-4, IL-1β, and IL-18. The ASC/CASP-1 complex formation was determined using an in situ proximity ligation assay. When comparing the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis to those without this placental lesion, we found that (1) the mRNA of NLR family pyrin domain-containing protein ( NLRP) 1, NLRP3, NLR family CARD domain-containing protein 4 ( NLRC4), and NOD2 were higher; (2) the NLRP3 protein was increased; (3) the mRNA and active form (p10) of CASP-1 were greater; (4) the mRNA and active form of CASP-4 were increased; (5) the mRNA and mature form of IL-1β were higher; (6) the mature form of IL-18 was elevated; and (7) ASC/CASP-1 complex formation was increased. In conclusion, spontaneous preterm labor with acute histologic chorioamnionitis is characterized by an upregulation of NLRP3 and the active form of CASP-4, as well as increased ASC/CASP-1 complex formation, which may participate in the activation of CASP-1 and the maturation of IL-1β and IL-18 in the chorioamniotic membranes. These findings provide the first evidence that supports a role for the inflammasome in the pathological inflammation implicated in spontaneous preterm labor with acute histologic chorioamnionitis.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Yi Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Olesya Plazyo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ronald Unkel
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yaozhu Leng
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhong Dong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
14
|
Gomez-Lopez N, Romero R, Xu Y, Miller D, Unkel R, Shaman M, Jacques SM, Panaitescu B, Garcia-Flores V, Hassan SS. Neutrophil Extracellular Traps in the Amniotic Cavity of Women with Intra-Amniotic Infection: A New Mechanism of Host Defense. Reprod Sci 2017; 24:1139-1153. [PMID: 27884950 PMCID: PMC6343453 DOI: 10.1177/1933719116678690] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) control microbial infections through their antimicrobial activities attributed to DNA, histones, granules, and cytoplasmic proteins (eg, elastase). Intra-amniotic infection is characterized by the influx of neutrophils into the amniotic cavity; therefore, the aim of this study was to determine whether amniotic fluid neutrophils form NETs in this inflammatory process. METHODS Amniotic fluid samples from women with intra-amniotic infection (n = 15) were stained for bacteria detection using fluorescent dyes. Amniotic fluid neutrophils were purified by filtration. As controls, neutrophils from maternal blood samples (n = 3) were isolated by density gradients. Isolated neutrophils were plated onto glass cover slips for culture with and without 100 nM of phorbol-12-myristate-13-acetate (PMA). NET formation was assessed by 4',6-diamidino-2-phenylindole (DAPI) staining and scanning electron microscopy. Different stages of NET formation were visualized using antibodies against elastase and histone H3, in combination with DAPI staining, by confocal microscopy. Finally, maternal or neonatal neutrophils were added to amniotic fluid samples from women without intra-amniotic infection (n = 4), and NET formation was evaluated by DAPI staining. RESULTS (1) NETs were present in the amniotic fluid of women with intra-amniotic infection; (2) all of the amniotic fluid samples had detectable live and dead bacteria associated with the presence of NETs; (3) in contrast to neutrophils from the maternal circulation, amniotic fluid neutrophils did not require PMA stimulation to form NETs; (4) different stages of NET formation were observed by co-localizing elastase, histone H3, and DNA in amniotic fluid neutrophils; and (5) neither maternal nor neonatal neutrophils form NETs in the amniotic fluid of women without intra-amniotic infection. CONCLUSION NETs are detectable in the amniotic fluid of women with intra-amniotic infection.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor,
MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University,
East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit,
MI, USA
| | - Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Derek Miller
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Ronald Unkel
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Majid Shaman
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Suzanne M. Jacques
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Pathology, Hutzel Women’s Hospital/Harper University Hospital,
Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| |
Collapse
|
15
|
Abstract
Preterm birth (PTB) remains a major obstetric healthcare problem and a significant contributor to perinatal morbidity, mortality, and long-term disability. Over the past few decades, the perinatal outcomes of preterm neonates have improved markedly through research and advances in neonatal care, whereas rates of spontaneous PTB have essentially remained static. However, research into causal pathways and new diagnostic and treatment modalities is now bearing fruit and translational initiatives are beginning to impact upon PTB rates. Successful PTB prevention requires a multifaceted approach, combining public health and educational programs, lifestyle modification, access to/optimisation of obstetric healthcare, effective prediction and diagnostic modalities, and the application of effective, targeted interventions. Progress has been made in some of these areas, although there remain areas of controversy and uncertainty. Attention is now being directed to areas where greater gains can be achieved. In this mini-review, we will briefly and selectively review a range of PTB prevention strategies and initiatives where progress has been made and where exciting opportunities await exploitation, evaluation, and implementation.
Collapse
Affiliation(s)
- Jeff A Keelan
- Division of Obstetrics & Gynaecology, School of Medicine, University of Western Australia King Edward Memorial Hospital, Perth, Australia
| | - John P Newnham
- Division of Obstetrics & Gynaecology, School of Medicine, University of Western Australia King Edward Memorial Hospital, Perth, Australia
| |
Collapse
|