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
|
Moore R, Spicer SK, Lu J, Chambers SA, Noble KN, Lochner J, Christofferson RC, Vasco KA, Manning SD, Townsend SD, Gaddy JA. The Utility of Human Milk Oligosaccharides against Group B Streptococcus Infections of Reproductive Tissues and Cognate Adverse Pregnancy Outcomes. ACS CENTRAL SCIENCE 2023; 9:1737-1749. [PMID: 37780357 PMCID: PMC10540283 DOI: 10.1021/acscentsci.3c00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Indexed: 10/03/2023]
Abstract
Preterm birth affects nearly 10% of all pregnancies in the United States, with 40% of those due, in part, to infections. Streptococcus agalactiae (Group B Streptococcus, GBS) is one of the most common perinatal pathogens responsible for these infections. Current therapeutic techniques aimed to ameliorate invasive GBS infections are less than desirable and can result in complications in both the neonate and the mother. To this end, the need for novel therapeutic options is urgent. Human milk oligosaccharides (HMOs), an integral component of human breast milk, have been previously shown to possess antiadhesive and antimicrobial properties. To interrogate these characteristics, we examined HMO-mediated outcomes in both in vivo and ex vivo models of GBS infection utilizing a murine model of ascending GBS infection, an EpiVaginal human organoid tissue model, and ex vivo human gestational membranes. Supplementation of HMOs resulted in diminished adverse pregnancy outcomes, decreased GBS adherence to gestational tissues, decreased colonization within the reproductive tract, and reduced proinflammatory immune responses to GBS infection. Taken together, these results highlight the potential of HMOs as promising therapeutic interventions in perinatal health.
Collapse
Affiliation(s)
- Rebecca
E. Moore
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Veterans Affairs, Tennessee
Valley Healthcare Systems, Nashville, Tennessee 37212, United States
| | - Sabrina K. Spicer
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Schuyler A. Chambers
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Kristen N. Noble
- Department
of Pediatrics, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Jonathan Lochner
- Department
of Pediatrics, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Rebecca C. Christofferson
- Department of Pathobiological
Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Karla A. Vasco
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Shannon D. Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Jennifer A. Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Veterans Affairs, Tennessee
Valley Healthcare Systems, Nashville, Tennessee 37212, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
3
|
Huang G, Yao D, Yan X, Zheng M, Yan P, Chen X, Wang D. Emerging role of toll-like receptors signaling and its regulators in preterm birth: a narrative review. Arch Gynecol Obstet 2023; 308:319-339. [PMID: 35916961 DOI: 10.1007/s00404-022-06701-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/03/2022] [Indexed: 11/02/2022]
Abstract
INTRODUCTION Despite intensive research, preterm birth (PTB) rates have not decreased significantly in recent years due to a lack of understanding of the underlying causes and insufficient treatment options for PTB. We are committed to finding promising biomarkers for the treatment of PTB. METHODS An extensive search of the literature was conducted with MEDLINE/PubMed, and in total, 151 studies were included and summarized in the present review. RESULTS Substantial evidence supports that the infection and/or inflammatory cascade associated with infection is an early event in PTB. Toll-like receptor (TLR) is a prominent pattern recognition receptor (PRR) found on both immune and non-immune cells, including fetal membrane cells. The activation of TLR downstream molecules, followed by TLR binding to its ligand, is critical for infection and inflammation, leading to the involvement of the TLR signaling pathway in PTB. TLR ligands are derived from microbial components and molecules released by damaged and dead cells. Particularly, TLR4 is an essential TLR because of its ability to recognize lipopolysaccharide (LPS). In this comprehensive overview, we discuss the role of TLR signaling in PTB, focus on numerous host-derived genetic and epigenetic regulators of the TLR signaling pathway, and cover ongoing research and prospective therapeutic options for treating PTB by inhibiting TLR signaling. CONCLUSION This is a critical topic because TLR-related molecules and mechanisms may enable obstetricians to better understand the physiological changes in PTB and develop new treatment and prevention strategies.
Collapse
Affiliation(s)
- Ge Huang
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dan Yao
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoli Yan
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mingyu Zheng
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ping Yan
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoxia Chen
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dan Wang
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
4
|
Bhattacharjee E, Thiruvengadam R, Ayushi, Das C, Wadhwa N, Natchu UCM, Kshetrapal P, Bhatnagar S, Majumder PP, Maitra A. Genetic variants associated with spontaneous preterm birth in women from India: a prospective cohort study. THE LANCET REGIONAL HEALTH. SOUTHEAST ASIA 2023; 14:100190. [PMID: 37492417 PMCID: PMC10363490 DOI: 10.1016/j.lansea.2023.100190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/28/2022] [Accepted: 03/23/2023] [Indexed: 07/27/2023]
Abstract
Background Despite having the highest number of preterm births globally, no genomic study on preterm birth was previously published from India or other South-Asian countries. Methods We conducted a genome-wide association (GWA) study of spontaneous preterm birth (sPTB) on 6211 women from India. We used a novel resampling procedure to identify the associated single nucleotide polymorphisms (SNPs) followed by haplotype association analysis and imputation. Findings We found that 512 maternal SNPs were associated with sPTB (p < 2.51e-3), of which minor allele at 19 SNPs (after Bonferroni correction) had increased genotype relative risk. Haplotypes containing six of the 19 SNPs (rs13011430, rs8179838, rs2327290, rs4798499, rs7629800, and rs13180906) were associated with sPTB (p < 9.9e-4; Bonferroni adjusted p-value <0.05). After imputation in regions around the 19 SNPs, 15 imputed SNPs were found to be associated with sPTB (Bonferroni adjusted p-value <0.05). One of these imputed SNPs, rs35760881, and three other SNPs (rs17307697, rs4308815, and rs10983507) were also reported to be associated with sPTB in women belonging to European ancestry. Moreover, we found that GG genotype at rs1152954, one of the associated SNPs, enhanced risk of sPTB and reduced telomere length. Interpretation This is the first study from South Asia on the genome-wide identification of maternal SNPs associated with sPTB. These SNPs are known to alter the expression of genes associated with major pathways in sPTB viz. inflammation, apoptosis, cervical ripening, telomere maintenance, selenocysteine biosynthesis, myometrial contraction, and innate immunity. From a public health perspective, the trans-ethnic association of four SNPs identified in our study may help to stratify women with risk of sPTB in most populations. Funding Department of Biotechnology (India), Grand Challenges India - All Children Thriving Program and Biotechnology Industry Research Assistance Council (BIRAC).
Collapse
Affiliation(s)
- Esha Bhattacharjee
- National Institute of Biomedical Genomics, PO: NSS, Kalyani, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
| | - Ramachandran Thiruvengadam
- Translational Health Science and Technology Institute, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
- Pondicherry Institute of Medical Sciences, Ganapathichettikulam, Kalapet, Puducherry, India
| | - Ayushi
- Translational Health Science and Technology Institute, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
| | - Chitrarpita Das
- National Institute of Biomedical Genomics, PO: NSS, Kalyani, India
| | | | - Nitya Wadhwa
- Translational Health Science and Technology Institute, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
| | - Uma Chandra Mouli Natchu
- Translational Health Science and Technology Institute, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
- Division of Infectious Diseases, St. John's Research Institute, 100 Feet Road, John Nagar, Koramangala, Bengaluru, India
| | - Pallavi Kshetrapal
- Translational Health Science and Technology Institute, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
| | - Shinjini Bhatnagar
- Translational Health Science and Technology Institute, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, India
| | - Partha Pratim Majumder
- National Institute of Biomedical Genomics, PO: NSS, Kalyani, India
- Indian Statistical Institute, Barrackpore Trunk Road, Kolkata, India
| | - Arindam Maitra
- National Institute of Biomedical Genomics, PO: NSS, Kalyani, India
| |
Collapse
|
5
|
Phung J, Wang C, Reeders J, Zakar T, Paul JW, Tyagi S, Pennell CE, Smith R. Preterm labor with and without chorioamnionitis is associated with activation of myometrial inflammatory networks: a comprehensive transcriptomic analysis. Am J Obstet Gynecol 2023; 228:330.e1-330.e18. [PMID: 36002050 DOI: 10.1016/j.ajog.2022.08.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The onset of preterm labor is associated with inflammation. Previous studies suggested that this is distinct from the inflammation observed during term labor. Our previous work on 44 genes differentially expressed in myometria in term labor demonstrated a different pattern of gene expression from that observed in preterm laboring and nonlaboring myometria. We found increased expression of inflammatory genes in preterm labor associated with chorioamnionitis, but in the absence of chorioamnionitis observed no difference in gene expression in preterm myometria regardless of laboring status, suggesting that preterm labor is associated with different myometrial genes or signals originating from outside the myometrium. Given that a small subset of genes were assessed, this study aimed to use RNA sequencing and bioinformatics to assess the myometrial transcriptome during preterm labor in the presence and absence of chorioamnionitis. OBJECTIVE This study aimed to comprehensively determine protein-coding transcriptomic differences between preterm nonlaboring and preterm laboring myometria with and without chorioamnionitis. STUDY DESIGN Myometria were collected at cesarean delivery from preterm patients not in labor (n=16) and preterm patients in labor with chorioamnionitis (n=8) or without chorioamnionitis (n=6). Extracted RNA from myometrial tissue was prepared and sequenced using Illumina NovaSeq. Gene expression was quantified by mapping the sequence reads to the human reference genome (hg38). Differential gene expression analysis, gene set enrichment analysis, and weighted gene coexpression network analysis were used to comprehensively interrogate transcriptomic differences and their associated biology. RESULTS Differential gene expression analysis comparing preterm patients in labor with chorioamnionitis with preterm patients not in labor identified 931 differentially expressed genes, whereas comparing preterm patients in labor without chorioamnionitis with preterm patients not in labor identified no statistically significant gene expression changes. In contrast, gene set enrichment analysis and weighted gene coexpression network analysis demonstrated that preterm labor with and without chorioamnionitis was associated with enrichment of pathways involved in activation of the innate immune system and inflammation, and activation of G protein-coupled receptors. Key genes identified included chemotactic CYP4F3, CXCL8, DOCK2, and IRF1 in preterm labor with chorioamnionitis and CYP4F3, FCAR, CHUK, and IL13RA2 in preterm labor without chorioamnionitis. There was marked overlap in the pathways enriched in both preterm labor subtypes. CONCLUSION Differential gene expression analysis demonstrated that myometria from preterm patients in labor without chorioamnionitis and preterm patients not in labor were transcriptionally similar, whereas the presence of chorioamnionitis was associated with marked gene changes. In contrast, comprehensive bioinformatic analysis indicated that preterm labor with or without chorioamnionitis was associated with innate immune activation. All causes of preterm labor were associated with activation of the innate immune system, but this was more marked in the presence of chorioamnionitis. These data suggest that anti-inflammatory therapy may be relevant in managing preterm labor of all etiologies.
Collapse
Affiliation(s)
- Jason Phung
- Department of Maternity and Gynaecology, John Hunter Hospital, Newcastle, Australia; Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, Australia.
| | - Carol Wang
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, Australia
| | - Jocelyn Reeders
- Department of Anatomical Pathology, John Hunter Hospital, Newcastle, Australia
| | - Tamas Zakar
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, Australia
| | - Jonathan W Paul
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, Australia
| | - Sonika Tyagi
- Central Clinical School, Monash University, Clayton, Australia
| | - Craig E Pennell
- Department of Maternity and Gynaecology, John Hunter Hospital, Newcastle, Australia; Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, Australia
| |
Collapse
|
6
|
Liu Y, Liu J, Liu A, Yin H, Burd I, Lei J. Maternal siRNA silencing of placental SAA2 mitigates preterm birth following intrauterine inflammation. Front Immunol 2022; 13:902096. [PMID: 36211368 PMCID: PMC9539923 DOI: 10.3389/fimmu.2022.902096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
The placental inflammatory processes induced maternally result in preterm birth (PTB). Serum amyloid A (SAA) is a well-known biomarker of inflammation. The objective of this study was to investigate whether murine placental SAA isoforms (SAA1–4) participate in the mechanism of spontaneous PTB and whether maternal regulation of SAA production may serve as a therapeutic approach. During the gestation, all isoforms of SAA were detectable except SAA2. The mouse model of intrauterine inflammation was established using LPS infusion to the uterus. Following intrauterine inflammation, placental SAA2 increased significantly. Inhibition of Saa2, using siSaa2, markedly decreased PTB. The increased placental expression of pro-inflammatory cytokines Il1β, Il6, and Tnfα were downregulated by siSaa2 treatment. Maternal inhibition of Saa2 did not change the expression of Saa1–4 in the fetal brain. Explant inflammatory culture of placentas with siSaa2 showed similar results to our in vivo experiments. This study demonstrates the highly expressed placental SAA2 as a novel therapeutic target, and maternal administration of siRNA as a promising approach to alleviate PTB.
Collapse
Affiliation(s)
- Yang Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jin Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anguo Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hillary Yin
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Irina Burd, ; Jun Lei,
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Irina Burd, ; Jun Lei,
| |
Collapse
|
7
|
Galaz J, Romero R, Arenas-Hernandez M, Farias-Jofre M, Motomura K, Liu Z, Kawahara N, Demery-Poulos C, Liu TN, Padron J, Panaitescu B, Gomez-Lopez N. Clarithromycin prevents preterm birth and neonatal mortality by dampening alarmin-induced maternal–fetal inflammation in mice. BMC Pregnancy Childbirth 2022; 22:503. [PMID: 35725425 PMCID: PMC9210693 DOI: 10.1186/s12884-022-04764-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background One of every four preterm neonates is born to a woman with sterile intra-amniotic inflammation (inflammatory process induced by alarmins); yet, this clinical condition still lacks treatment. Herein, we utilized an established murine model of sterile intra-amniotic inflammation induced by the alarmin high-mobility group box-1 (HMGB1) to evaluate whether treatment with clarithromycin prevents preterm birth and adverse neonatal outcomes by dampening maternal and fetal inflammatory responses. Methods Pregnant mice were intra-amniotically injected with HMGB1 under ultrasound guidance and treated with clarithromycin or vehicle control, and pregnancy and neonatal outcomes were recorded (n = 15 dams each). Additionally, amniotic fluid, placenta, uterine decidua, cervix, and fetal tissues were collected prior to preterm birth for determination of the inflammatory status (n = 7–8 dams each). Results Clarithromycin extended the gestational length, reduced the rate of preterm birth, and improved neonatal mortality induced by HMGB1. Clarithromycin prevented preterm birth by interfering with the common cascade of parturition as evidenced by dysregulated expression of contractility-associated proteins and inflammatory mediators in the intra-uterine tissues. Notably, clarithromycin improved neonatal survival by dampening inflammation in the placenta as well as in the fetal lung, intestine, liver, and spleen. Conclusions Clarithromycin prevents preterm birth and improves neonatal survival in an animal model of sterile intra-amniotic inflammation, demonstrating the potential utility of this macrolide for treating women with this clinical condition, which currently lacks a therapeutic intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04764-2.
Collapse
|
8
|
Gomez-Lopez N, Garcia-Flores V, Chin PY, Groome HM, Bijland MT, Diener KR, Romero R, Robertson SA. Macrophages exert homeostatic actions in pregnancy to protect against preterm birth and fetal inflammatory injury. JCI Insight 2021; 6:146089. [PMID: 34622802 PMCID: PMC8525593 DOI: 10.1172/jci.insight.146089] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/20/2021] [Indexed: 01/08/2023] Open
Abstract
Macrophages are commonly thought to contribute to the pathophysiology of preterm labor by amplifying inflammation — but a protective role has not previously been considered to our knowledge. We hypothesized that given their antiinflammatory capability in early pregnancy, macrophages exert essential roles in maintenance of late gestation and that insufficient macrophages may predispose individuals to spontaneous preterm labor and adverse neonatal outcomes. Here, we showed that women with spontaneous preterm birth had reduced CD209+CD206+ expression in alternatively activated CD45+CD14+ICAM3– macrophages and increased TNF expression in proinflammatory CD45+CD14+CD80+HLA-DR+ macrophages in the uterine decidua at the materno-fetal interface. In Cd11bDTR/DTR mice, depletion of maternal CD11b+ myeloid cells caused preterm birth, neonatal death, and postnatal growth impairment, accompanied by uterine cytokine and leukocyte changes indicative of a proinflammatory response, while adoptive transfer of WT macrophages prevented preterm birth and partially rescued neonatal loss. In a model of intra-amniotic inflammation–induced preterm birth, macrophages polarized in vitro to an M2 phenotype showed superior capacity over nonpolarized macrophages to reduce uterine and fetal inflammation, prevent preterm birth, and improve neonatal survival. We conclude that macrophages exert a critical homeostatic regulatory role in late gestation and are implicated as a determinant of susceptibility to spontaneous preterm birth and fetal inflammatory injury.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, US Department of Health and Human Services; Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, US Department of Health and Human Services; Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Holly M Groome
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Melanie T Bijland
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kerrilyn R Diener
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, US Department of Health and Human Services; Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA.,Detroit Medical Center, Detroit, Michigan, USA
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
9
|
Jing X, Min C, Qi Yun L, Shun Qin H, Li Rui L, Jia L, Run Mei M. Toll-like receptor 2/4 inhibitors can reduce preterm birth in mice. J Int Med Res 2021; 48:300060520933795. [PMID: 33100071 PMCID: PMC7604950 DOI: 10.1177/0300060520933795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Objectives Preterm birth (PTB) occurs in 5% to 18% of newborns. However, the underlying inflammatory mechanisms have not been elucidated. Methods We established a mouse model of infection-associated PTB. Physical signs in pregnant mice with or without lipopolysaccharide (LPS) treatment were observed, and the frequencies of Toll-like receptor (TLR)2- and TLR4-positive CD11b+ cells were analyzed. Cytokine levels in plasma and pathological changes were assessed following LPS treatment. A rescue experiment was used to probe potential immunologic mechanisms underlying PTB. Results Lymphocyte infiltration could be observed in the placentas of mice following intrauterine injection with LPS. The percentage of inflammatory cells decreased 12 hours after treatment. Moreover, TLR2 and TLR4 expression in peripheral blood cells was significantly increased 4 hours after intraperitoneal injection of LPS. Peak TLR2 and TLR4 expression in peripheral blood cells occurred 8 hours post-treatment. TLR4 and TLR-2/4 inhibitors reduced levels of interleukin-10, interferon-γ, and tumor necrosis factor-α in peripheral blood and delayed PTB. Conclusions TLR2 and TLR4 inhibition could play important roles in PTB.
Collapse
Affiliation(s)
- Xu Jing
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chen Min
- Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liu Qi Yun
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hu Shun Qin
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Li Rui
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Jia
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ma Run Mei
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
10
|
Sheller-Miller S, Radnaa E, Yoo JK, Kim E, Choi K, Kim Y, Kim YN, Richardson L, Choi C, Menon R. Exosomal delivery of NF-κB inhibitor delays LPS-induced preterm birth and modulates fetal immune cell profile in mouse models. SCIENCE ADVANCES 2021; 7:eabd3865. [PMID: 33523942 PMCID: PMC10671068 DOI: 10.1126/sciadv.abd3865] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/04/2020] [Indexed: 06/12/2023]
Abstract
Accumulation of immune cells and activation of the pro-inflammatory transcription factor NF-κB in feto-maternal uterine tissues is a key feature of preterm birth (PTB) pathophysiology. Reduction of the fetal inflammatory response and NF-κB activation are key strategies to minimize infection-associated PTB. Therefore, we engineered extracellular vesicles (exosomes) to contain an NF-κB inhibitor, termed super-repressor (SR) IκBα. Treatment with SR exosomes (1 × 1010 per intraperitoneal injection) after lipopolysaccharide (LPS) challenge on gestation day 15 (E15) prolonged gestation by over 24 hours (PTB ≤ E18.5) and reduced maternal inflammation (n ≥ 4). Furthermore, using a transgenic model in which fetal tissues express the red fluorescent protein tdTomato while maternal tissues do not, we report that LPS-induced PTB in mice is associated with influx of fetal innate immune cells, not maternal, into feto-maternal uterine tissues. SR packaged in exosomes provides a stable and specific intervention for reducing the inflammatory response associated with PTB.
Collapse
Affiliation(s)
- Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Enkhtuya Radnaa
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | | | - Eunsoo Kim
- ILIAS Biologics, Incorporated, Daejeon, South Korea
| | | | - Youngeun Kim
- ILIAS Biologics, Incorporated, Daejeon, South Korea
| | - Yu Na Kim
- ILIAS Biologics, Incorporated, Daejeon, South Korea
| | - Lauren Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Chulhee Choi
- ILIAS Biologics, Incorporated, Daejeon, South Korea
- Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
11
|
Ophelders DRMG, Boots AW, Hütten MC, Al-Nasiry S, Jellema RK, Spiller OB, van Schooten FJ, Smolinska A, Wolfs TGAM. Screening of Chorioamnionitis Using Volatile Organic Compound Detection in Exhaled Breath: A Pre-clinical Proof of Concept Study. Front Pediatr 2021; 9:617906. [PMID: 34123958 PMCID: PMC8187797 DOI: 10.3389/fped.2021.617906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/29/2021] [Indexed: 11/13/2022] Open
Abstract
Chorioamnionitis is a major risk factor for preterm birth and an independent risk factor for postnatal morbidity for which currently successful therapies are lacking. Emerging evidence indicates that the timing and duration of intra-amniotic infections are crucial determinants for the stage of developmental injury at birth. Insight into the dynamical changes of organ injury after the onset of chorioamnionitis revealed novel therapeutic windows of opportunity. Importantly, successful development and implementation of therapies in clinical care is currently impeded by a lack of diagnostic tools for early (prenatal) detection and surveillance of intra-amniotic infections. In the current study we questioned whether an intra-amniotic infection could be accurately diagnosed by a specific volatile organic compound (VOC) profile in exhaled breath of pregnant sheep. For this purpose pregnant Texel ewes were inoculated intra-amniotically with Ureaplasma parvum and serial collections of exhaled breath were performed for 6 days. Ureaplasma parvum infection induced a distinct VOC-signature in expired breath of pregnant sheep that was significantly different between day 0 and 1 vs. day 5 and 6. Based on a profile of only 15 discriminatory volatiles, animals could correctly be classified as either infected (day 5 and 6) or not (day 0 and 1) with a sensitivity of 83% and a specificity of 71% and an area under the curve of 0.93. Chemical identification of these distinct VOCs revealed the presence of a lipid peroxidation marker nonanal and various hydrocarbons including n-undecane and n-dodecane. These data indicate that intra-amniotic infections can be detected by VOC analyses of exhaled breath and might provide insight into temporal dynamics of intra-amniotic infection and its underlying pathways. In particular, several of these volatiles are associated with enhanced oxidative stress and undecane and dodecane have been reported as predictive biomarker of spontaneous preterm birth in humans. Applying VOC analysis for the early detection of intra-amniotic infections will lead to appropriate surveillance of these high-risk pregnancies, thereby facilitating appropriate clinical course of action including early treatment of preventative measures for pre-maturity-associated morbidities.
Collapse
Affiliation(s)
- Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Center+, Maastricht, Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Agnes W Boots
- Department Pharmacology and Toxicology, Maastricht University, Maastricht, Netherlands.,NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Matthias C Hütten
- Department of Pediatrics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Salwan Al-Nasiry
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Reint K Jellema
- Department of Pediatrics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Owen B Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Frederik-Jan van Schooten
- Department Pharmacology and Toxicology, Maastricht University, Maastricht, Netherlands.,NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Agnieszka Smolinska
- Department Pharmacology and Toxicology, Maastricht University, Maastricht, Netherlands.,NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Center+, Maastricht, Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
12
|
Potential anti-neuroinflammatory compounds from Australian plants - A review. Neurochem Int 2020; 142:104897. [PMID: 33186611 DOI: 10.1016/j.neuint.2020.104897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
Neuroinflammation is a complex response to brain injury involving the activation of glia, release of inflammatory mediators, such as cytokines and chemokines, and generation of reactive oxygen and nitrogen species. Even though it is considered an event secondary to neuronal death or dysfunction, neuro-inflammation comprises a majority of the non-neuronal contributors to the cause and progression of neurodegenerative diseases like Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple Sclerosis (MS), Chronic Traumatic Encephalopathy (CTE) and others. As a result of the lack of effectiveness of current treatments for neurodegenerative diseases, neuroinflammation has become a legitimate therapeutic target for drug discovery, leading to the study of various in vivo and in vitro models of neuroinflammation. Several molecules sourced from plants have displayed anti-inflammatory properties in the study of neurodegenerative diseases. A group of these anti-inflammatory compounds has been classified as cytokine-suppressive anti-inflammatory drugs (CSAIDs), which target the pro-inflammatory AP1 and nuclear factor-κB signaling pathways and inhibit the expression of many pro-inflammatory cytokines, such as interleukin IL-1, IL-6, TNF-α, or nitric oxide. Australian plants, thriving amid the driest inhabited continent of the world, are an untapped source of chemical diversity in the form of secondary metabolites. These compounds are produced in response to biotic and abiotic stresses that the plants are exposed to in the highly biodiverse environment. This review is an attempt to highlight anti-inflammatory compounds isolated from Australian plants.
Collapse
|
13
|
Zhang J, Luo X, Huang C, Pei Z, Xiao H, Luo X, Huang S, Chang Y. Erythropoietin prevents LPS-induced preterm birth and increases offspring survival. Am J Reprod Immunol 2020; 84:e13283. [PMID: 32506750 PMCID: PMC7507205 DOI: 10.1111/aji.13283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/13/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
PROBLEM Preterm delivery is the leading cause of neonatal mortality and contributes to delayed physical and cognitive development in children. At present, there is no efficient therapy to prevent preterm labor. A large body of evidence suggests that infections might play a significant and potentially preventable cause of premature birth. This work assessed the effects of erythropoietin (EPO) in a murine model of inflammation-associated preterm delivery, which mimics central features of preterm infections in humans. METHOD OF STUDY BALB/c mice were injected i.p. with 20 000 IU/kg EPO or normal saline twice on gestational day (GD) 15, with a 3 hours time interval between injections. An hour after the first EPO or normal saline injection, all mice received two injections of 50 μg/kg LPS, also given 3 hours apart. RESULTS EPO significantly prevented preterm labor and increased offspring survival in an LPS induced preterm delivery model. EPO prevented LPS-induced leukocyte infiltration into the placenta. Moreover, EPO inhibited the expression of pro-inflammatory cytokines, interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumour necrosis factor-α (TNF-α) in maternal serum and amniotic fluid. EPO also prevented LPS-induced increase in placental prostaglandin (PG)E2 and uterine inducible nitric oxide synthase (iNOS) production, while decreasing nuclear factor kappa-B (NF-κβ) activity in the myometrium. EPO also increased the gene expression of placental programmed cell death ligand 1 (PD-L1) in LPS-treated mice. CONCLUSIONS Our results suggest that EPO could be a potential novel therapeutic strategy to tackle infection-related preterm labor.
Collapse
Affiliation(s)
- Jie Zhang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Xianqiong Luo
- Department of PediatricsGuangdong Women and Children HospitalGuangzhouChina
| | - Caicai Huang
- Department of ObstetricsGuangdong Women and Children HospitalGuangzhouChina
| | - Zheng Pei
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Huimei Xiao
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Xingang Luo
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Shuangmiao Huang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Yanqun Chang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| |
Collapse
|
14
|
Firmal P, Shah VK, Chattopadhyay S. Insight Into TLR4-Mediated Immunomodulation in Normal Pregnancy and Related Disorders. Front Immunol 2020; 11:807. [PMID: 32508811 PMCID: PMC7248557 DOI: 10.3389/fimmu.2020.00807] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Unlike organ transplants where an immunosuppressive environment is required, a successful pregnancy involves an extremely robust, dynamic, and responsive maternal immune system to maintain the development of the fetus. A specific set of hormones and cytokines are associated with a particular stage of pregnancy. Any disturbance that alters this fine balance could compromise the development and function of the placenta. Although there are numerous underlying causes of pregnancy-related complications, untimely activation of Toll-like receptors (TLR), primarily TLR4, by intrauterine microbes poses the greatest risk. TLR4 is an important Pattern Recognition Receptor (PRR), which activates both innate and adaptive immune cells. TLR4 activation by LPS or DAMPs leads to the production of pro-inflammatory cytokines via the MyD88 dependent or independent pathway. Immune cells modulate the materno–fetal interface by TLR4-mediated cytokine production, which changes at different stages of pregnancy. In most pregnancy disorders, such as PTB, PE, or placental malaria, the TLR4 expression is upregulated in immune cells or in maternal derived cells, leading to the aberrant production of pro-inflammatory cytokines at the materno–fetal interface. Lack of functional TLR4 in mice has reduced the pro-inflammatory responses, leading to an improved pregnancy, which further strengthens the fact that abnormal TLR4 activation creates a hostile environment for the developing fetus. A recent study proposed that endothelial and perivascular stromal cells should interact with each other in order to maintain a homeostatic balance during TLR4-mediated inflammation. It has been reported that depleting immune cells or supplying anti-inflammatory cytokines can prevent PTB, PE, or fetal death. Blocking TLR4 signaling or its downstream molecule by inhibitors or antagonists has proven to improve pregnancy-related complications to some extent in clinical and animal models. To date, there has been a lack of knowledge regarding whether TLR4 accessories such as CD14 and MD-2 are important in pregnancy and whether these accessory molecules could be promising drug targets for combinatorial treatment of various pregnancy disorders. This review mainly focuses on the activation of TLR4 during pregnancy, its immunomodulatory functions, and the upcoming advancement in this field regarding the improvement of pregnancy-related issues by various therapeutic approaches.
Collapse
Affiliation(s)
- Priyanka Firmal
- National Centre for Cell Science, S. P. Pune University Campus, Pune, India
| | - Vibhuti Kumar Shah
- National Centre for Cell Science, S. P. Pune University Campus, Pune, India
| | - Samit Chattopadhyay
- National Centre for Cell Science, S. P. Pune University Campus, Pune, India.,Department of Biological Sciences, BITS Pilani, K. K. Birla Goa Campus, Goa, India.,Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
15
|
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
|
16
|
Robertson SA, Hutchinson MR, Rice KC, Chin PY, Moldenhauer LM, Stark MJ, Olson DM, Keelan JA. Targeting Toll-like receptor-4 to tackle preterm birth and fetal inflammatory injury. Clin Transl Immunology 2020; 9:e1121. [PMID: 32313651 PMCID: PMC7156293 DOI: 10.1002/cti2.1121] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/30/2022] Open
Abstract
Every year, 15 million pregnancies end prematurely, resulting in more than 1 million infant deaths and long-term health consequences for many children. The physiological processes of labour and birth involve essential roles for immune cells and pro-inflammatory cytokines in gestational tissues. There is compelling evidence that the mechanisms underlying spontaneous preterm birth are initiated when a premature and excessive inflammatory response is triggered by infection or other causes. Exposure to pro-inflammatory mediators is emerging as a major factor in the 'fetal inflammatory response syndrome' that often accompanies preterm birth, where unscheduled effects in fetal tissues interfere with normal development and predispose to neonatal morbidity. Toll-like receptors (TLRs) are critical upstream gatekeepers of inflammatory activation. TLR4 is prominently involved through its ability to sense and integrate signals from a range of microbial and endogenous triggers to provoke and perpetuate inflammation. Preclinical studies have identified TLR4 as an attractive pharmacological target to promote uterine quiescence and protect the fetus from inflammatory injury. Novel small-molecule inhibitors of TLR4 signalling, specifically the non-opioid receptor antagonists (+)-naloxone and (+)-naltrexone, are proving highly effective in animal models for preventing preterm birth induced by bacterial mimetic LPS, heat-killed Escherichia coli, or the TLR4-dependent pro-inflammatory lipid, platelet-activating factor (PAF). Here, we summarise the rationale for targeting TLR4 as a master regulator of inflammation in fetal and gestational tissues, and the potential utility of TLR4 antagonists as candidates for preventative and therapeutic application in preterm delivery and fetal inflammatory injury.
Collapse
Affiliation(s)
- Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Mark R Hutchinson
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia.,ARC Centre for Nanoscale Biophotonics and Adelaide Medical School University of Adelaide Adelaide SA 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 MD USA
| | - Peck-Yin Chin
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Michael J Stark
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - David M Olson
- Department of Obstetrics and Gynecology Department of Physiology and Pediatrics 220 HMRC University of Alberta Edmonton AB Canada
| | - Jeffrey A Keelan
- Division of Obstetrics & Gynaecology University of Western Australia Perth WA Australia
| |
Collapse
|
17
|
Cui HS, Joo SY, Cho YS, Kim JB, Seo CH. CPEB1 or CPEB4 knockdown suppresses the TAK1 and Smad signalings in THP-1 macrophage-like cells and dermal fibroblasts. Arch Biochem Biophys 2020; 683:108322. [DOI: 10.1016/j.abb.2020.108322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 01/03/2023]
|
18
|
ASK1 promotes uterine inflammation leading to pathological preterm birth. Sci Rep 2020; 10:1887. [PMID: 32024889 PMCID: PMC7002619 DOI: 10.1038/s41598-020-58653-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
It is widely accepted that enhanced uterine inflammation associated with microbial infection is a main causative factor for preterm birth. However, little is known about the molecular basis by which inflammation is associated with preterm birth. Here, we demonstrate that apoptosis signal-regulating kinase 1 (ASK1), a member of the mitogen-activated protein 3-kinase family, facilitates inflammation-induced preterm birth and that inhibition of ASK1 activity is sufficient to suppress preterm birth. ASK1-deficient pregnant mice exhibited reduced incidence of lipopolysaccharide (LPS)-induced preterm birth. ASK1 was required for the induction of LPS-induced inflammatory responses related to preterm birth, including pro-inflammatory cytokine production in the uterus and peritoneal cavities. In addition, selective suppression of uterine ASK1 activity through a chemical genetic approach reduced the incidence of LPS-induced preterm birth. Moreover, translational studies with human choriodecidua demonstrated that ASK1 was required for LPS-induced activation of JNK and p38 and pro-inflammatory cytokine production. Our findings suggest that ASK1 activation is responsible for the induction of inflammation that leads to preterm birth and that the blockade of ASK1 signaling might be a promising therapeutic target for preventing preterm birth.
Collapse
|
19
|
Effects of inflammation on the developing respiratory system: Focus on hypoglossal (XII) neuron morphology, brainstem neurochemistry, and control of breathing. Respir Physiol Neurobiol 2020; 275:103389. [PMID: 31958568 DOI: 10.1016/j.resp.2020.103389] [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] [Received: 06/03/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 12/20/2022]
Abstract
Breathing is fundamental to life and any adverse change in respiratory function can endanger the health of an organism or even be fatal. Perinatal inflammation is known to adversely affect breathing in preterm babies, but lung infection/inflammation impacts all stages of life from birth to death. Little is known about the role of inflammation in respiratory control, neuronal morphology, or neural function during development. Animal models of inflammation can provide understanding and insight into respiratory development and how inflammatory processes alter developmental phenotype in addition to providing insight into new treatment modalities. In this review, we focus on recent work concerning the development of neurons, models of perinatal inflammation with an emphasis on two common LPS-based models, inflammation and its impact on development, and current and potential treatments for inflammation within the respiratory control circuitry of the mammalian brainstem. We have also discussed models of inflammation in adults and have specifically focused on hypoglossal motoneurons (XII) and neurons of the nucleus tractus solitarii (nTS) as these nuclei have been studied more extensively than other brainstem nuclei participating in breathing and airway control. Understanding the impact of inflammation on the developmental aspects of respiratory control and breathing pattern is critical to addressing problems of cardiorespiratory dysregulation in disease and this overview points out many gaps in our current knowledge.
Collapse
|
20
|
Brabin B, Tinto H, Roberts SA. Testing an infection model to explain excess risk of preterm birth with long-term iron supplementation in a malaria endemic area. Malar J 2019; 18:374. [PMID: 31771607 PMCID: PMC6880560 DOI: 10.1186/s12936-019-3013-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/16/2019] [Indexed: 12/15/2022] Open
Abstract
Background In view of recent evidence from a randomized trial in Burkina Faso that periconceptional iron supplementation substantially increases risk of spontaneous preterm birth (< 37 weeks) in first pregnancies (adjusted relative risk = 2.22; 95% CI 1.39–3.61), explanation is required to understand potential mechanisms, including progesterone mediated responses, linking long-term iron supplementation, malaria and gestational age. Methods The analysis developed a model based on a dual hit inflammatory mechanism arising from simultaneous malaria and gut infections, supported in part by published trial results. This model is developed to understand mechanisms linking iron supplementation, malaria and gestational age. Background literature substantiates synergistic inflammatory effects of these infections where trial data is unavailable. A path modelling exercise assessed direct and indirect paths influencing preterm birth and gestation length. Results A dual hit hypothesis incorporates two main pathways for pro-inflammatory mechanisms, which in this model, interact to increase hepcidin expression. Trial data showed preterm birth was positively associated with C-reactive protein (P = 0.0038) an inflammatory biomarker. The malaria pathway upregulates C-reactive protein and serum hepcidin, thereby reducing iron absorption. The enteric pathway results from unabsorbed gut iron, which induces microbiome changes and pathogenic gut infections, initiating pro-inflammatory events with lipopolysaccharide expression. Data from the trial suggest that raised hepcidin concentration is a mediating catalyst, being inversely associated with shorter gestational age at delivery (P = 0.002) and positively with preterm incidence (P = 0.007). A segmented regression model identified a change-point consisting of two segments before and after a sharp rise in hepcidin concentration. This showed a post change hepcidin elevation in women with increasing C-reactive protein values in late gestation (post-change slope 0.55. 95% CI 0.39–0.92, P < 0.001). Path modelling confirmed seasonal malaria effects on preterm birth, with mediation through C-reactive protein and (non-linear) hepcidin induction. Conclusions Following long-term iron supplementation, dual inflammatory pathways that mediate hepcidin expression and culminate in progesterone withdrawal may account for the reduction in gestational age observed in first pregnancies in this area of high malaria exposure. If correct, this model strongly suggests that in such areas, effective infection control is required prior to iron supplementation to avoid increasing preterm births. Trial registration NCT01210040. Registered with Clinicaltrials.gov on 27th September 2010
Collapse
Affiliation(s)
- Bernard Brabin
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, L35QA, UK. .,Institute of Infection and Global Health, University of Liverpool, Liverpool, UK. .,Global Child Health Group, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | - Halidou Tinto
- Clinical Research Unit of Nanoro (URCN/IRSS), Nanoro, Burkina Faso
| | - Stephen A Roberts
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary Care, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre (MAHSC), Manchester, UK
| |
Collapse
|
21
|
Reuschel E, Toelge M, Entleutner K, Deml L, Seelbach-Goebel B. Cytokine profiles of umbilical cord blood mononuclear cells upon in vitro stimulation with lipopolysaccharides of different vaginal gram-negative bacteria. PLoS One 2019; 14:e0222465. [PMID: 31536529 PMCID: PMC6752847 DOI: 10.1371/journal.pone.0222465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/30/2019] [Indexed: 01/01/2023] Open
Abstract
Inflammatory immune responses induced by lipopolysaccharides (LPS) of gram-negative bacteria play an important role in the pathogenesis of preterm labor and delivery, and in neonatal disorders. To better characterize LPS-induced inflammatory response, we determined the cytokine profile of umbilical cord blood mononuclear cells (UBMC) stimulated with LPS of seven vaginal gram-negative bacteria commonly found in pregnant women with preterm labor and preterm rupture of membrane. UBMC from ten newborns of healthy volunteer mothers were stimulated with purified LPS of Escherichia coli, Enterobacter aerogenes, Klebsiella pneumoniae, Proteus mirabilis, Acinetobacter calcoaceticus, Citrobacter freundii, and Pseudomonas aeruginosa. UBMC supernatants were tested for the presence of secreted pro-inflammatory cytokines (IL-6, IL-1β, TNF), anti-inflammatory cytokine (IL-10), TH1-type cytokines (IL-12, IFN-γ), and chemokines (IL-8, MIP-1α, MIP-1β, MCP-1) by Luminex technology. The ten cytokines were differentially induced by the LPS variants. LPS of E. coli and E. aerogenes showed the strongest stimulatory activity and P. aeruginosa the lowest. Interestingly, the ability of UBMC to respond to LPS varied greatly among donors, suggesting a strong individual heterogeneity in LPS-triggered inflammatory response.
Collapse
Affiliation(s)
- Edith Reuschel
- Department of Obstetrics and Gynecology, University of Regensburg, Hospital of the Barmherzige Brueder, Clinic St Hedwig, Regensburg, Germany
- * E-mail:
| | - Martina Toelge
- Institute of Medical Microbiology, University Hospital Regensburg, Regensburg, Germany
| | - Kathrin Entleutner
- Department of Obstetrics and Gynecology, University of Regensburg, Hospital of the Barmherzige Brueder, Clinic St Hedwig, Regensburg, Germany
| | - Ludwig Deml
- Institute of Medical Microbiology, University Hospital Regensburg, Regensburg, Germany
| | - Birgit Seelbach-Goebel
- Department of Obstetrics and Gynecology, University of Regensburg, Hospital of the Barmherzige Brueder, Clinic St Hedwig, Regensburg, Germany
| |
Collapse
|
22
|
Lamont RF. Spontaneous preterm labour that leads to preterm birth: An update and personal reflection. Placenta 2019; 79:21-29. [PMID: 30981438 DOI: 10.1016/j.placenta.2019.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The objective was to provide an update of progress made over time (including personal reflection) of our attempts to reduce the mortality and morbidity associated with spontaneous preterm labour that leads to preterm birth. METHODS An experienced and evidence based approach was taken to provide an overview of progress made over a generation (∼40 years) in our understanding of spontaneous preterm labour. RESULTS It is evident that we have made significant progress in our understanding of the aetiology, the measurement of the burden, the basic science, systems biology and mechanical pathways of the preterm parturition syndrome. We have better ways of predicting, preventing and managing spontaneous preterm labour than existed a generation ago. CONCLUSIONS The profile of spontaneous preterm labour that leads to preterm birth, thanks to organisations such as the March of Dimes, WHO and PREBIC is much more evident than before. However, while we have come a long way, we must not be complacent, and clinicians and basic scientists must continue to work in harmony, while recruiting and encouraging young investigators to join the effort to improve survival and handicap in what is one of the Great Obstetric Syndromes.
Collapse
Affiliation(s)
- Ronald F Lamont
- Department of Gynaecology and Obstetrics, University of Southern Denmark, Odense University Hospital, Odense, Denmark, and Division of Surgery, University College London, Northwick Park Institute for Medical Research Campus, London, UK.
| |
Collapse
|
23
|
Pustotina O. Effects of antibiotic therapy in women with the amniotic fluid "sludge" at 15-24 weeks of gestation on pregnancy outcomes. J Matern Fetal Neonatal Med 2019; 33:3016-3027. [PMID: 30621474 DOI: 10.1080/14767058.2019.1567706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective: The aim of this prospective study was to assess the efficacy of antibiotic therapy for the prevention of adverse pregnancy outcomes in women with the amniotic fluid "sludge" at 15-24 weeks of gestation.Methods: 245 women underwent transvaginal ultrasound cervical length measurement at 15-24 weeks of pregnancy and 29 out of them with amniotic fluid "sludge" were included in the study. Eight women with the "sludge" had cervical length >25 mm (Group I), seven-an asymptomatic short cervix (Group IIa) and 14 women with a short cervix had symptoms like low abdominal pain, back pain, and menstrual-like cramps (Group IIb). All participants received intravenous, oral and/or vaginal antibiotic therapy. Participants in Group IIa were additionally given vaginal progesterone (VP), and in Group IIb-VP and indomethacin. Placentas from women with preterm birth (PTB) underwent histological examination.Results: The amniotic fluid "sludge" detected at an ultrasound scan between 15-24 weeks of gestation was associated with long-term maternal infections, histological chorioamnionitis, and was viewed as a marker of intra-amniotic infection. Absence of intravenous antibiotic therapy during midtrimester of pregnancy in these women was associated with neonatal infection with intrauterine onset in 61.1%, postpartum endometritis in 23.1%, and rate of PTB 46.2%. Intravenous antibiotic therapy eliminated sonographic presence of the sludge and resulted in prevented of neonatal and postpartum infections, prevented the risk of PTB in women with the cervical length >25 mm, in those with an asymptomatic short cervix receiving VP, and in 70% of symptomatic women with a short cervix, who received them in combination VP/indomethacin. For those women whose approach was not fully beneficial, it allowed to delay delivery in 11-17 weeks.Conclusions: Although we found that intravenous antibiotic therapy at 15-24 weeks of gestation in women with amniotic fluid "sludge" can protect from infection-related complications and demonstrated high beneficial effects of adding antibiotics to anti-inflammatory drug (indomethacin) and/or VP in women with a short cervix, further larger studies are needed.
Collapse
Affiliation(s)
- Olga Pustotina
- Obstetrics, Gynecology and Perinatology, Peoples' Friendship University of Russia, Moscow, Russia
| |
Collapse
|
24
|
Gurney LRI, Taggart J, Tong WC, Jones AT, Robson SC, Taggart MJ. Inhibition of Inflammatory Changes in Human Myometrial Cells by Cell Penetrating Peptide and Small Molecule Inhibitors of NFκB. Front Immunol 2018; 9:2966. [PMID: 30619324 PMCID: PMC6307458 DOI: 10.3389/fimmu.2018.02966] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/03/2018] [Indexed: 01/17/2023] Open
Abstract
Complications arising from Preterm Birth are the leading causes of neonatal death globally. Current therapeutic strategies to prevent Preterm Birth are yet to demonstrate success in terms of reducing this neonatal disease burden. Upregulation of intracellular inflammatory pathways in uterine cells, including those involving nuclear factor kappa-B (NFκB), have been causally linked to both human term and preterm labor, but the barrier presented by the cell membrane presents an obstacle to interventions aimed at dampening these inflammatory responses. Cell penetrating peptides (CPPs) are novel vectors that can traverse cell membranes without the need for recognition by cell surface receptors and offer the ability to deliver therapeutic cargo internal to cell membranes. Using a human uterine cell culture inflammatory model, this study aimed to test the effectiveness of CPP-cargo delivery to inhibit inflammatory responses, comparing this effect with a small molecule inhibitor (Sc514) that has a similar intracellular target of action within the NFκB pathway (the IKK complex). The CPP Penetratin, conjugated to rhodamine, was able to enter uterine cells within a 60 min timeframe as assessed by live confocal microscopy, this phenomena was not observed with the use of a rhodamine-conjugated inert control peptide (GC(GS)4). Penetratin CPP conjugated to an IKK-inhibitory peptide (Pen-NBD) demonstrated ability to inhibit both the IL1β-induced expression of the inflammatory protein COX2 and dampen the expression of a bespoke array of inflammatory genes. Truncation of the CPP vector rendered the CPP-cargo conjugate much less effective, demonstrating the importance of careful vector selection. The small molecule inhibitor Sc514 also demonstrated ability to inhibit COX2 protein responses and a broad down-regulatory effect on uterine cell inflammatory gene expression. These results support the further exploration of either CPP-based or small molecular treatment strategies to dampen gestational cell inflammatory responses in the context of preterm birth. The work underlines both the importance of careful selection of CPP vector-cargo combinations and basic testing over a broad time and concentration range to ensure effective responses. Further work should demonstrate the effectiveness of CPP-linked cargos to dampen alternative pathways of inflammation linked to Preterm Birth such as MAP Kinase or AP1.
Collapse
Affiliation(s)
- Leo R. I. Gurney
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Julie Taggart
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Wing-Chiu Tong
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Arwyn T. Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | - Stephen C. Robson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Michael J. Taggart
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
25
|
Amabebe E, Anumba DOC. The Vaginal Microenvironment: The Physiologic Role of Lactobacilli. Front Med (Lausanne) 2018; 5:181. [PMID: 29951482 PMCID: PMC6008313 DOI: 10.3389/fmed.2018.00181] [Citation(s) in RCA: 353] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/29/2018] [Indexed: 11/21/2022] Open
Abstract
In addition to being a passage for sperm, menstruum, and the baby, the human vagina and its microbiota can influence conception, pregnancy, the mode and timing of delivery, and the risk of acquiring sexually transmitted infections. The physiological status of the vaginal milieu is important for the wellbeing of the host as well as for successful reproduction. High estrogen states, as seen during puberty and pregnancy, promote the preservation of a homeostatic (eubiotic) vaginal microenvironment by stimulating the maturation and proliferation of vaginal epithelial cells and the accumulation of glycogen. A glycogen-rich vaginal milieu is a haven for the proliferation of Lactobacilli facilitated by the production of lactic acid and decreased pH. Lactobacilli and their antimicrobial and anti-inflammatory products along with components of the epithelial mucosal barrier provide an effective first line defense against invading pathogens including bacterial vaginosis, aerobic vaginitis-associated bacteria, viruses, fungi and protozoa. An optimal host-microbial interaction is required for the maintenance of eubiosis and vaginal health. This review explores the composition, function and adaptive mechanisms of the vaginal microbiome in health and those disease states in which there is a breach in the host-microbial relationship. The potential impact of vaginal dysbiosis on reproduction is also outlined.
Collapse
Affiliation(s)
- Emmanuel Amabebe
- Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Dilly O C Anumba
- Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
26
|
Intrauterine inflammatory activation, functional progesterone withdrawal, and the timing of term and preterm birth. J Reprod Immunol 2018; 125:89-99. [DOI: 10.1016/j.jri.2017.12.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 12/17/2017] [Accepted: 12/21/2017] [Indexed: 01/19/2023]
|
27
|
Furfaro LL, Chang BJ, Payne MS. Applications for Bacteriophage Therapy during Pregnancy and the Perinatal Period. Front Microbiol 2018; 8:2660. [PMID: 29375525 PMCID: PMC5768649 DOI: 10.3389/fmicb.2017.02660] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022] Open
Abstract
Pregnant women and their unborn children are a population that is particularly vulnerable to bacterial infection. Physiological changes that occur during pregnancy affect the way women respond to such infections and the options that clinicians have for treatment. Antibiotics are still considered the best option for active infections and a suitable prophylaxis for prevention of potential infections, such as vaginal/rectal Streptococcus agalactiae colonization prior to birth. The effect of such antibiotic use on the developing fetus, however, is still largely unknown. Recent research has suggested that the fetal gut microbiota plays a critical role in fetal immunologic programming. Hence, even minor alterations in this microbiota may have potentially significant downstream effects. An ideal antibacterial therapeutic for administration during pregnancy would be one that is highly specific for its target, leaving the surrounding microbiota intact. This review first provides a basic overview of the challenges a clinician faces when administering therapeutics to a pregnant patient and then goes on to explore common bacterial infections in pregnancy, use of antibiotics for treatment/prevention of such infections and the consequences of such treatment for the mother and infant. With this background established, the review then explores the potential for use of bacteriophage (phage) therapy as an alternative to antibiotics during the antenatal period. Many previous reviews have highlighted the revitalization of and potential for phage therapy for treatment of a range of bacterial infections, particularly in the context of the increasing threat of widespread antibiotic resistance. However, information on the potential for the use of phage therapeutics in pregnancy is lacking. This review aims to provide a thorough overview of studies of this nature and discuss the feasibility of bacteriophage use during pregnancy to treat and/or prevent bacterial infections.
Collapse
Affiliation(s)
- Lucy L. Furfaro
- Division of Obstetrics and Gynecology, School of Medicine, The University of Western Australia, Crawley, WA, Australia
| | - Barbara J. Chang
- Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Matthew S. Payne
- Division of Obstetrics and Gynecology, School of Medicine, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
28
|
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
|
29
|
Preterm birth: Inflammation, fetal injury and treatment strategies. J Reprod Immunol 2016; 119:62-66. [PMID: 28122664 DOI: 10.1016/j.jri.2016.11.008] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 11/23/2016] [Accepted: 11/29/2016] [Indexed: 12/18/2022]
Abstract
Preterm birth (PTB) is the leading cause of childhood mortality in children under 5 and accounts for approximately 11% of births worldwide. Premature babies are at risk of a number of health complications, notably cerebral palsy, but also respiratory and gastrointestinal disorders. Preterm deliveries can be medically indicated/elective procedures or they can occur spontaneously. Spontaneous PTB is commonly associated with intrauterine infection/inflammation. The presence of inflammatory mediators in utero has been associated with fetal injury, particularly affecting the fetal lungs and brain. This review will outline (i) the role of inflammation in term and PTB, (ii) the effect infection/inflammation has on fetal development and (iii) recent strategies to target PTB. Further research is urgently required to develop effective methods for the prevention and treatment of PTB and above all, to reduce fetal injury.
Collapse
|
30
|
Ireland DJ, Nathan EA, Li S, Charles AK, Stinson LF, Kemp MW, Newnham JP, Keelan JA. Preclinical evaluation of drugs to block inflammation-driven preterm birth. Innate Immun 2016; 23:20-33. [PMID: 27821647 DOI: 10.1177/1753425916672313] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Intrauterine inflammation, the major cause of early preterm birth, can have microbial and sterile aetiologies. We assessed in a Transwell model the anti-inflammatory efficacies of five drugs on human extraplacental membranes delivered after preterm spontaneous labour (30-34 wk). Drugs [TPCA1 (IKKβ inhibitor), 5 z-7-oxozeaenol (OxZ, TAK1 inhibitor), inhibitor of NF-κB essential modulator binding domain (iNBD), SB239063 (p38 MAPK inhibitor) and N-acetyl cysteine (free radical scavenger free radicals)] were added after 12 h equilibration to the amniotic compartment. Concentrations of IL-6, TNF-α, MCP-1, IL-1β and PGE2 in the media, and IL6, TNFA and PTGS2 mRNA expression levels in membranes, were determined after 12 h. Data were analysed using mixed models analyses. Thirteen of the 28 membranes had histological chorioamnionitis (HCA+); five were positive for bacterial culture and six for fetal inflammatory reaction. Baseline PGE2 and cytokine production was similar between HCA- and HCA+ membranes. Anti-inflammatory effects were also similar between HCA- and HCA+ membranes. TPCA1 and OxZ were the most effective drugs; each inhibited amniotic secretion of 4/5 pro-inflammatory mediators and mRNA levels of 2/3, regardless of stimulus. We conclude that treatment with TPCA1 or OxZ, in combination with antibiotics, may minimise the adverse effects of intrauterine inflammation in pregnancy.
Collapse
Affiliation(s)
- Demelza J Ireland
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia
| | - Elizabeth A Nathan
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia.,2 Women and Infants Research Foundation of Western Australia, King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - Shaofu Li
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia.,2 Women and Infants Research Foundation of Western Australia, King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - Adrian K Charles
- 3 Department of Pathology, Sidra Medical and Research Center, Doha, Qatar
| | - Lisa F Stinson
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia
| | - Matthew W Kemp
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia.,2 Women and Infants Research Foundation of Western Australia, King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - John P Newnham
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia.,2 Women and Infants Research Foundation of Western Australia, King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - Jeffrey A Keelan
- 1 School of Women's and Infants' Health, King Edward Memorial Hospital, The University of Western Australia, Perth, Western Australia, Australia.,2 Women and Infants Research Foundation of Western Australia, King Edward Memorial Hospital, Perth, Western Australia, Australia
| |
Collapse
|
31
|
Chin PY, Dorian CL, Hutchinson MR, Olson DM, Rice KC, Moldenhauer LM, Robertson SA. Novel Toll-like receptor-4 antagonist (+)-naloxone protects mice from inflammation-induced preterm birth. Sci Rep 2016; 6:36112. [PMID: 27819333 PMCID: PMC5098167 DOI: 10.1038/srep36112] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 10/11/2016] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptor 4 (TLR4) activation by bacterial infection, or by sterile inflammatory insult is a primary trigger of spontaneous preterm birth. Here we utilize mouse models to investigate the efficacy of a novel small molecule TLR4 antagonist, (+)-naloxone, the non-opioid isomer of the opioid receptor antagonist (−)-naloxone, in infection-associated preterm birth. Treatment with (+)-naloxone prevented preterm delivery and alleviated fetal demise in utero elicited by i.p. LPS administration in late gestation. A similar effect with protection from preterm birth and perinatal death, and partial correction of reduced birth weight and postnatal mortality, was conferred by (+)-naloxone administration after intrauterine administration of heat-killed E. coli. Local induction by E. coli of inflammatory cytokine genes Il1b, Il6, Tnf and Il10 in fetal membranes was suppressed by (+)-naloxone, and cytokine expression in the placenta, and uterine myometrium and decidua, was also attenuated. These data demonstrate that inhibition of TLR4 signaling with the novel TLR4 antagonist (+)-naloxone can suppress the inflammatory cascade of preterm parturition, to prevent preterm birth and perinatal death. Further studies are warranted to investigate the utility of small molecule inhibition of TLR-driven inflammation as a component of strategies for fetal protection and delaying preterm birth in the clinical setting.
Collapse
Affiliation(s)
- Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Camilla L Dorian
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Mark R Hutchinson
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA, 5005, Australia
| | - David M Olson
- Departments of Obstetrics &Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, Alberta T6G2S2, Canada
| | - Kenner C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
32
|
Chao MW, Chen CP, Yang YH, Chuang YC, Chu TY, Tseng CY. N-acetylcysteine attenuates lipopolysaccharide-induced impairment in lamination of Ctip2-and Tbr1- expressing cortical neurons in the developing rat fetal brain. Sci Rep 2016; 6:32373. [PMID: 27577752 PMCID: PMC5006028 DOI: 10.1038/srep32373] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/03/2016] [Indexed: 02/02/2023] Open
Abstract
Oxidative stress and inflammatory insults are the major instigating events of bacterial intrauterine infection that lead to fetal brain injury. The purpose of this study is to investigate the remedial effects of N-acetyl-cysteine (NAC) for inflammation-caused deficits in brain development. We found that lipopolysaccharide (LPS) induced reactive oxygen species (ROS) production by RAW264.7 cells. Macrophage-conditioned medium caused noticeable cortical cell damage, specifically in cortical neurons. LPS at 25 μg/kg caused more than 75% fetal loss in rats. An increase in fetal cortical thickness was noted in the LPS-treated group. In the enlarged fetal cortex, laminar positioning of the early born cortical cells expressing Tbr1 and Ctip2 was disrupted, with a scattered distribution. The effect was similar, but minor, in later born Satb2-expressing cortical cells. NAC protected against LPS-induced neuron toxicity in vitro and counteracted pregnancy loss and alterations in thickness and lamination of the neocortex in vivo. Fetal loss and abnormal fetal brain development were due to LPS-induced ROS production. NAC is an effective protective agent against LPS-induced damage. This finding highlights the key therapeutic impact of NAC in LPS-caused abnormal neuronal laminar distribution during brain development.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Chie-Pein Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Hsiu Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Tzu-Yun Chu
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- International Master Program of Biomedical Material and Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- Center for Nano-Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| |
Collapse
|
33
|
Menon R, Papaconstantinou J. p38 Mitogen activated protein kinase (MAPK): a new therapeutic target for reducing the risk of adverse pregnancy outcomes. Expert Opin Ther Targets 2016; 20:1397-1412. [PMID: 27459026 DOI: 10.1080/14728222.2016.1216980] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Spontaneous preterm birth (PTB) and preterm premature rupture of the membranes (pPROM) remain as a major clinical and therapeutic problem for intervention and management. Current strategies, based on our knowledge of pathways of preterm labor, have only been effective, in part, due to major gaps in our existing knowledge of risks and risk specific pathways. Areas covered: Recent literature has identified physiologic aging of fetal tissues as a potential mechanistic feature of normal parturition. This process is affected by telomere dependent and p38 mitogen activated protein kinase (MAPK) induced senescence activation. Pregnancy associated risk factors can cause pathologic activation of this pathway that can cause oxidative stress induced p38 MAPK activation leading to senescence and premature aging of fetal tissues. Premature aging is associated with sterile inflammation capable of triggering preterm labor or preterm premature rupture of membranes. Preterm activation of p38MAPK can be considered as a key contributor to adverse pregnancies. Expert opinion: This review considers p38MAPK activation as a potential target for therapeutic interventions to prevent adverse pregnancy outcomes mediated by stress factors. In this review, we propose multiple strategies to prevent p38MAPK activation.
Collapse
Affiliation(s)
- Ramkumar Menon
- a Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology , The University of Texas Medical Branch at Galveston , Galveston , TX , USA
| | - John Papaconstantinou
- b Department of Biochemistry and Molecular Biology , The University of Texas Medical Branch at Galveston , Galveston , TX , USA
| |
Collapse
|
34
|
Keelan JA, Payne MS, Kemp MW, Ireland DJ, Newnham JP. A New, Potent, and Placenta-Permeable Macrolide Antibiotic, Solithromycin, for the Prevention and Treatment of Bacterial Infections in Pregnancy. Front Immunol 2016; 7:111. [PMID: 27066004 PMCID: PMC4817400 DOI: 10.3389/fimmu.2016.00111] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 03/14/2016] [Indexed: 01/24/2023] Open
Abstract
Intrauterine infection–inflammation is a major cause of early preterm birth and subsequent neonatal mortality and acute or long-term morbidity. Antibiotics can be administered in pregnancy to prevent preterm birth either prophylactically to women at high risk for preterm delivery, or to women with diagnosed intrauterine infection, prelabor rupture of membranes, or in suspected preterm labor. The therapeutic goals of each of these scenarios are different, with different pharmacological considerations, although effective antimicrobial therapy is an essential requirement. An ideal antibiotic for these clinical indications would be (a) one that is easily administered and orally bioactive, (b) has a favorable adverse effect profile (devoid of reproductive toxicity or teratogenicity), (c) is effective against the wide range of microorganisms known to be commonly associated with intra-amniotic infection, (d) provides effective antimicrobial protection within both the fetal and amniotic compartments after maternal delivery, (e) has anti-inflammatory properties, and (f) is effective against antibiotic-resistant microorganisms. Here, we review the evidence from clinical, animal, and ex vivo/in vitro studies that demonstrate that a new macrolide-derived antibiotic – solithromycin – has all of these properties and, hence, may be an ideal antibiotic for the treatment and prevention of intrauterine infection-related pregnancy complications. While this evidence is extremely encouraging, it is still preliminary. A number of key studies need to be completed before solithromycin’s true potential for use in pregnancy can be ascertained.
Collapse
Affiliation(s)
- Jeffrey A Keelan
- King Edward Memorial Hospital, School of Women's and Infants' Health, University of Western Australia , Perth, WA , Australia
| | - Matthew S Payne
- King Edward Memorial Hospital, School of Women's and Infants' Health, University of Western Australia , Perth, WA , Australia
| | - Matthew W Kemp
- King Edward Memorial Hospital, School of Women's and Infants' Health, University of Western Australia , Perth, WA , Australia
| | - Demelza J Ireland
- King Edward Memorial Hospital, School of Women's and Infants' Health, University of Western Australia , Perth, WA , Australia
| | - John P Newnham
- King Edward Memorial Hospital, School of Women's and Infants' Health, University of Western Australia , Perth, WA , Australia
| |
Collapse
|
35
|
Toda A, Sawada K, Fujikawa T, Wakabayashi A, Nakamura K, Sawada I, Yoshimura A, Nakatsuka E, Kinose Y, Hashimoto K, Mabuchi S, Tokuhira A, Nakayama M, Itai A, Kurachi H, Kimura T. Targeting Inhibitor of κB Kinase β Prevents Inflammation-Induced Preterm Delivery by Inhibiting IL-6 Production from Amniotic Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:616-29. [DOI: 10.1016/j.ajpath.2015.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/09/2015] [Accepted: 11/10/2015] [Indexed: 10/22/2022]
|
36
|
Kim SM, Romero R, Lee J, Chaemsaithong P, Lee MW, Chaiyasit N, Lee HJ, Yoon BH. About one-half of early spontaneous preterm deliveries can be identified by a rapid matrix metalloproteinase-8 (MMP-8) bedside test at the time of mid-trimester genetic amniocentesis. J Matern Fetal Neonatal Med 2015; 29:2414-22. [PMID: 26643648 DOI: 10.3109/14767058.2015.1094049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Mid-trimester amniocentesis continues to be used for the prenatal diagnosis of chromosomal anomalies and other genetic disorders. Analysis of amniotic fluid obtained at the time of mid-trimester genetic amniocentesis identifies those patients who are at risk for early spontaneous preterm delivery. This is based on a solid body of evidence that found subclinical intra-amniotic inflammation/infection to be causally linked to early spontaneous preterm birth. Although several biomarkers have been proposed to identify intra-amniotic inflammation, the accumulated data suggest that the determination of amniotic fluid matrix metalloproteinase-8 (MMP-8), or neutrophil collagenase, is a powerful predictor of spontaneous preterm delivery. MMP-8 is released by inflammatory cells in response to microbial products or "danger signals". A rapid point-of-care test has been developed to determine MMP-8 at the bedside within 20 min, and without the requirement of laboratory equipment. The objective of this study was to determine whether an elevation of MMP-8 in the amniotic fluid, measured by a rapid point-of-care test, can identify those patients at risk for spontaneous preterm delivery after a mid-trimester genetic amniocentesis. STUDY DESIGN A case-control study was designed to obtain amniotic fluid from asymptomatic singleton pregnant women who underwent mid-trimester genetic amniocentesis. An MMP-8 bedside test was performed to analyze the amniotic fluid of 64 patients with early spontaneous preterm delivery (<30 weeks) and 128 matched controls with normal pregnancy outcomes. RESULTS (1) The MMP-8 bedside test (Yoon's MMP-8 Check™) was positive in 42.2% (27/64) of patients with spontaneous preterm delivery but in none (0/128) of the control cases (p < 0.001); (2) the MMP-8 bedside test had a sensitivity of 42.2%, and a specificity of 100% in the prediction of spontaneous preterm delivery (<30 weeks) following a mid-trimester genetic amniocentesis; and (3) among the patients with spontaneous preterm delivery, those with a positive MMP-8 bedside test had a significantly higher rate of spontaneous delivery within 2 weeks and 4 weeks of an amniocentesis [40.7% (11/27) versus 5.4% (2/37); 63.0% (17/27) versus 24.3% (9/37)] and a shorter interval-to-delivery period than those with a negative test [interval-to-delivery: median (range), 16 d (0-95 d) versus 42 d (2-91 d); p < 0.05 for each]. CONCLUSION We conclude that 42% of patients with an early spontaneous preterm delivery (< 30 weeks) could be identified by a rapid MMP-8 bedside test at the time of their mid-trimester genetic amniocentesis. The MMP-8 bedside test is a powerful predictor of early spontaneous preterm birth in asymptomatic pregnant women.
Collapse
Affiliation(s)
- Sun Min Kim
- a Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea .,b Department of Obstetrics and Gynecology , Seoul Metropolitan Government-Seoul National University Boramae Medical Center , Seoul , Republic of Korea
| | - Roberto Romero
- c 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 .,d Department of Obstetrics and Gynecology , University of Michigan , Ann Arbor , MI , USA .,e Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA .,f Center for Molecular Medicine and Genetics, Wayne State University , Detroit , MI , USA , and
| | - JoonHo Lee
- a Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Piya Chaemsaithong
- c 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 .,g Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Min-Woo Lee
- a Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Noppadol Chaiyasit
- c 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 .,g Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Hyo-Jin Lee
- a Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Bo Hyun Yoon
- a Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| |
Collapse
|
37
|
Lamont RF. Advances in the Prevention of Infection-Related Preterm Birth. Front Immunol 2015; 6:566. [PMID: 26635788 PMCID: PMC4644786 DOI: 10.3389/fimmu.2015.00566] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 10/23/2015] [Indexed: 11/17/2022] Open
Abstract
Infection-related preterm birth (PTB) is more common at early gestational ages and is associated with major neonatal mortality and morbidity. Abnormal genital tract microflora in early pregnancy predicts late miscarriage and early PTB. Accordingly, it is logical to consider antibiotics as an intervention. Unfortunately, the conclusions of systematic reviews and meta-analyses (SR&MAs) carried out in an attempt to explain the confusion over the heterogeneity of individual studies are flawed by the fact that undue reliance was placed on studies which: (a) had a suboptimal choice of antibiotic (mainly metronidazole) or used antibiotics not recommended for the treatment of bacterial vaginosis (BV) or BV-related organisms; (b) used antibiotics too late in pregnancy to influence outcome (23–27 weeks); and (c) included women whose risk of PTB was not due to abnormal genital tract colonization and hence unlikely to respond to antibiotics. These risks included: (a) previous PTB of indeterminate etiology; (b) low weight/body mass index; or (c) detection of fetal fibronectin, ureaplasmas, Group B streptococcus or Trichomonas vaginalis). While individual studies have found benefit of antibiotic intervention for the prevention of PTB, in meta-analyses these effects have been negated by large methodologically flawed studies with negative results. As a result, many clinicians think that any antibiotic given at any time in pregnancy to any woman at risk of PTB will cause more harm than good. Recently, a more focused SR&MA has demonstrated that antibiotics active against BV-related organisms, used in women whose risk of PTB is due to abnormal microflora, and used early in pregnancy before irreversible inflammatory damage has occurred, can reduce the rate of PTB. This review presents those data, the background and attempts to explain the confusion using new information from culture-independent molecular-based techniques. It also gives guidance on the structure of putative future antibiotic intervention studies.
Collapse
Affiliation(s)
- Ronald F Lamont
- Research Unit of Gynecology and Obstetrics, Department of Gynecology and Obstetrics, Institute of Clinical Research, Odense University Hospital, University of Southern Denmark , Odense , Denmark ; Division of Surgery, University College London , London , UK
| |
Collapse
|