51
|
Mosoro E, Wilson AN, Homer CSE, Vogel JP. Assessing the quality of antenatal corticosteroids in low- and middle-income countries: A systematic review. PLoS One 2020; 15:e0243034. [PMID: 33270682 PMCID: PMC7714108 DOI: 10.1371/journal.pone.0243034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/15/2020] [Indexed: 11/26/2022] Open
Abstract
Background The World Health Organization (WHO) recommends the administration of intramuscular antenatal corticosteroids to women at risk of preterm birth to prevent preterm-associated neonatal mortality and morbidity. Poor quality medicines are a major problem for health services in low- and middle-income countries (LMICs), however the quality of antenatal corticosteroids is not well understood. We aimed to conduct a systematic review of available studies describing the quality of recommended injectable antenatal corticosteroids (dexamethasone or betamethasone) in LMICs. Methods Structured search strategy was applied to six databases (MEDLINE, EMBASE, CINAHL, International Pharmaceutical Abstracts, Global Index Medicus, WHO Medicines Quality Database), without year or language restrictions. Any primary study reporting any medicine quality parameter (Active Pharmacological Ingredient, pH and sterility) for injectable dexamethasone or betamethasone was eligible. Two authors independently screened studies for eligibility, extracted data on included studies and applied Medicine Quality Assessment Reporting Guidelines tool to assess study quality. Results were reported narratively, stratified by country of manufacture, organisation type and level of care. Results In total, 15,547 citations were screened with two eligible studies identified that focussed on dexamethasone quality (no studies of betamethasone were identified). One study included 19 samples from 9 LMICs, and the other included “less than 100 samples” from India. The prevalence of failed dexamethasone samples ranged from 3.14% to 32.2% due to inadequate Active Pharmacological Ingredient. A higher prevalence of failed dexamethasone samples were seen at the point of care and the public sector. Conclusions Poor quality maternal and newborn health medicines can endanger women and newborns. Though available evidence on antenatal corticosteroids quality in LMICs is limited, results suggested poor quality dexamethasone may be prevalent in some countries. More primary studies are required to confirm these findings and guide policymakers on procurement of good-quality maternal and newborn health medicines.
Collapse
Affiliation(s)
- Euodia Mosoro
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Australia
- School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Alyce N. Wilson
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Australia
- School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Caroline S. E. Homer
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Australia
- School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Joshua P. Vogel
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Australia
- School of Population and Global Health, University of Melbourne, Melbourne, Australia
- * E-mail:
| |
Collapse
|
52
|
Cobo T, Kacerovsky M, Jacobsson B. Risk factors for spontaneous preterm delivery. Int J Gynaecol Obstet 2020; 150:17-23. [PMID: 32524595 DOI: 10.1002/ijgo.13184] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/23/2020] [Accepted: 04/23/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND There is a substantial variation in rates of preterm delivery between different parts of the world. The understanding of these variations, as well as the biological mechanisms behind spontaneous preterm delivery, is limited. Although the benefit of antenatal interventions has been shown to be limited, using well-known risk factors for spontaneous preterm delivery to select the correct pregnant women for targeted interventions is important from both a medical and caregiving perspective. OBJECTIVE To provide an introduction to a substantial research area dealing with risk factors of spontaneous preterm delivery. METHODS Risk factors in this review were classified as demographical, obstetrical, and gynecological and those related to the current pregnancy according to high-quality evidence of recent literature. RESULTS AND CONCLUSION An introduction to a substantial research area in maternal and fetal medicine was provided that might help clinicians to better understand the risk factors related to preterm delivery and select the correct pregnant women for targeted interventions.
Collapse
Affiliation(s)
- Teresa Cobo
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clinic and Hospital Sant Joan de Deu), Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Marian Kacerovsky
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic.,Department of Obstetrics and Gynecology, Faculty of Medicine Hradec Kralove, Charles University in Prague, Hradec Kralove, Czech Republic
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Department of Genetics and Bioinformatics, Domain of Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
53
|
Sabihi M, Böttcher M, Pelczar P, Huber S. Microbiota-Dependent Effects of IL-22. Cells 2020; 9:E2205. [PMID: 33003458 PMCID: PMC7599675 DOI: 10.3390/cells9102205] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokines are important contributors to immune responses against microbial and environmental threats and are of particular importance at epithelial barriers. These interfaces are continuously exposed to external factors and thus require immune components to both protect the host from pathogen invasion and to regulate overt inflammation. Recently, substantial efforts have been devoted to understanding how cytokines act on certain cells at barrier sites, and why the dysregulation of immune responses may lead to pathogenesis. In particular, the cytokine IL-22 is involved in preserving an intact epithelium, maintaining a balanced microbiota and a functioning defense system against external threats. However, a tight regulation of IL-22 is generally needed, since uncontrolled IL-22 production can lead to the progression of autoimmunity and cancer. Our aim in this review is to summarize novel findings on IL-22 and its interactions with specific microbial stimuli, and subsequently, to understand their contributions to the function of IL-22 and the clinical outcome. We particularly focus on understanding the detrimental effects of dysregulated control of IL-22 in certain disease contexts.
Collapse
Affiliation(s)
| | | | | | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; (M.S.); (M.B.); (P.P.)
| |
Collapse
|
54
|
Romantsik O, Bruschettini M, Ley D. Intraventricular Hemorrhage and White Matter Injury in Preclinical and Clinical Studies. Neoreviews 2020; 20:e636-e652. [PMID: 31676738 DOI: 10.1542/neo.20-11-e636] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Germinal matrix-intraventricular hemorrhage (IVH) occurs in nearly half of infants born at less than 26 weeks' gestation. Up to 50% of survivors with IVH develop cerebral palsy, cognitive deficits, behavioral disorders, posthemorrhagic ventricular dilatation, or a combination of these sequelae. After the initial bleeding and the primary brain injury, inflammation and secondary brain injury might lead to periventricular leukomalacia or diffuse white matter injury. Potential factors that are involved include microglia and astrocyte activation, degradation of blood components with release of "toxic" products, infiltration of the brain by systemic immune cells, death of neuronal and glial cells, and arrest of preoligodendrocyte maturation. In addition, impairment of the blood-brain barrier may play a major role in the pathophysiology. A wide range of animal models has been used to explore causes and mechanisms leading to IVH-induced brain injury. Preclinical studies have identified potential targets for enhancing brain repair. However, little has been elucidated about the effectiveness of potential interventions in clinical studies. A systematic review of available preclinical and clinical studies might help identify research gaps and which types of interventions may be prioritized. Future trials should report clinically robust and long-term outcomes after IVH.
Collapse
Affiliation(s)
- Olga Romantsik
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
55
|
Menon R, Behnia F, Polettini J, Richardson LS. Novel pathways of inflammation in human fetal membranes associated with preterm birth and preterm pre-labor rupture of the membranes. Semin Immunopathol 2020; 42:431-450. [PMID: 32785751 DOI: 10.1007/s00281-020-00808-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Spontaneous preterm birth (PTB) and preterm pre-labor rupture of the membranes (pPROM) are major pregnancy complications. Although PTB and pPROM have common etiologies, they arise from distinct pathophysiologic pathways. Inflammation is a common underlying mechanism in both conditions. Balanced inflammation is required for fetoplacental growth; however, overwhelming inflammation (physiologic at term and pathologic at preterm) can lead to term and preterm parturition. A lack of effective strategies to control inflammation and reduce the risk of PTB and pPROM suggests that there are several modes of the generation of inflammation which may be dependent on the type of uterine tissue. The avascular fetal membrane (amniochorion), which provides structure, support, and protection to the intrauterine cavity, is one of the key contributors of inflammation. Localized membrane inflammation helps tissue remodeling during pregnancy. Two unique mechanisms that generate balanced inflammation are the progressive development of senescence (aging) and cyclic cellular transitions: epithelial to mesenchymal (EMT) and mesenchymal to epithelial (MET). The intrauterine build-up of oxidative stress at term or in response to risk factors (preterm) can accelerate senescence and promote a terminal state of EMT, resulting in the accumulation of inflammation. Inflammation degrades the matrix and destabilizes membrane function. Inflammatory mediators from damaged membranes are propagated via extracellular vesicles (EV) to maternal uterine tissues and transition quiescent maternal uterine tissues into an active state of labor. Membrane inflammation and its propagation are fetal signals that may promote parturition. This review summarizes the mechanisms of fetal membrane cellular senescence, transitions, and the generation of inflammation that contributes to term and preterm parturitions.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA.
| | - Faranak Behnia
- Department of Obstetrics, Gynecology, and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, UT Health, Houston, Texas, USA
| | - Jossimara Polettini
- Universidade Federal da Fronteira Sul, Campus Passo Fundo, Rua Capitão Araujo, 20, Centro, Passo Fundo, Rio Grande do Sul, Brazil
| | - Lauren S Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA
| |
Collapse
|
56
|
Deng W, Yuan J, Cha J, Sun X, Bartos A, Yagita H, Hirota Y, Dey SK. Endothelial Cells in the Decidual Bed Are Potential Therapeutic Targets for Preterm Birth Prevention. Cell Rep 2020; 27:1755-1768.e4. [PMID: 31067461 DOI: 10.1016/j.celrep.2019.04.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/18/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
Preterm birth (PTB) is a syndrome with many origins. Among them, infection or inflammation are major risk factors for PTB; however, local defense mechanisms to mount anti-inflammatory responses against inflammation-induced PTB are poorly understood. Here, we show that endothelial TLR4 in the decidual bed is critical for sensing inflammation during pregnancy because mice with endothelial Tlr4 deletion are resistant to lipopolysaccharide (LPS)-induced PTB. Under inflammatory conditions, IL-6 is readily expressed in decidual endothelial cells with signal transducer and activator of transcription 3 (Stat3) phosphorylation in perivascular stromal cells, which then regulates expression of anti-inflammatory IL-10. Our observation that administration of an IL-10 neutralizing antibody predisposing mice to PTB shows IL-10's anti-inflammatory role to prevent PTB. We show that the integration of endothelial and perivascular stromal signaling can determine pregnancy outcomes. These findings highlight a role for endothelial TLR4 in inflammation-induced PTB and may offer a potential therapeutic target to prevent PTB.
Collapse
Affiliation(s)
- Wenbo Deng
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA; College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jia Yuan
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA; College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jeeyeon Cha
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xiaofei Sun
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA; College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Amanda Bartos
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA; College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, University of Tokyo, Japan
| | - Sudhansu K Dey
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45299, USA; College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA.
| |
Collapse
|
57
|
Batura R, Colbourn T. A stitch in time: narrative review of interventions to reduce preterm births in Malawi. Int Health 2020; 12:213-221. [PMID: 31867622 PMCID: PMC7320425 DOI: 10.1093/inthealth/ihz101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/14/2019] [Accepted: 12/16/2019] [Indexed: 11/13/2022] Open
Abstract
Background The rising rate of preterm births (PTBs) is a global concern, and Malawi has a high rate of PTBs (10.5%). The resulting neonatal and under-5 mortality, morbidity and lifelong disability represent a significant loss of human potential affecting individuals, families and society as a whole. This study aims to review the literature to determine the risk factors for PTB in Malawi and to identify effective interventions to prevent PTBs. Methods A literature search yielded 22 studies that were categorized according to risk factors implicated for PTBs and health interventions to reduce the risks. Results The study has shown that maternal pregnancy factors, infections, nutrition, anaemia and young maternal age are the main causes and risk factors of PTBs in Malawi. The literature revealed no evidence of community-based interventions for reducing the rates of PTBs in Malawi. Conclusions Any successful effort to reduce the rate of PTBs will require a multisector, multilevel strategy targeted at the community, homes and individuals as a package to improve the education, nutrition and reproductive health of girls and women as well as focus on improving the delivery of antenatal services in the community.
Collapse
Affiliation(s)
- Rekha Batura
- UCL Institute for Global Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Tim Colbourn
- UCL Institute for Global Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
58
|
Stranik J, Kacerovsky M, Vescicik P, Faist T, Jacobsson B, Musilova I. A rodent model of intra-amniotic inflammation/infection, induced by the administration of inflammatory agent in a gestational sac, associated with preterm delivery: a systematic review. J Matern Fetal Neonatal Med 2020; 35:1592-1600. [PMID: 32349576 DOI: 10.1080/14767058.2020.1757063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Rodents are the most commonly used animals in the study of spontaneous preterm delivery (PTD). Intra-amniotic inflammation/infection is a frequent and important cause of PTD. Intraperitoneal and intrauterine administrations of inflammatory agents are traditional methods to establish a rodent model of PTD associated with inflammation and infection. The intra-amniotic administration of inflammatory or infectious triggering agents to rodents can be useful to study not only intra-amniotic inflammatory response but also PTD associated with intra-amniotic inflammation/infection.Objective: This systematic review aimed mainly to assess and analyze all described methods of intra-amniotic administration of infectious and/or inflammatory agents to create a rodent model of intra-amniotic inflammation associated with PTD.Methods: A literature search through two electronic databases from their earliest entries to February 2019 was performed. The selection criteria were as follows: (1) rodents as model animals, (2) a model of intra-amniotic inflammation/infection associated with PTD, and (3) intra-amniotic administration of triggering agents. Data extraction included specification of the study (author and year of publication), characteristics of study animals (species, strain, and number of animals), characteristics of intervention (timing and used technique), substance used for induction of intra-amniotic inflammation/infection, and outcome assessment.Results: The search identified a total of 4673 articles, of which 118 were selected for full-text reading, but only 13 studies were included in the review. Intra-amniotic administration was used only in the articles that were published beyond 2004. Two different approaches were identified: (1) open surgery with direct puncture of the amniotic sacs and (2) transabdominal ultrasound-guided puncture of the gestational sacs. Live microorganisms (Ureaplasma parvum), bacterial products (extracellular membrane vesicles), and pathogen-associated (lipopolysaccharide) and damage-associated molecular patterns (high mobility group box-1, S100B, and surfactant protein A) were used to simulate intra-amniotic inflammation/infection. Differences in the effect on intra-amniotic inflammation/infection associated with PTD in the mouse model were identified among triggering agents. Intra-amniotic application of lipopolysaccharide in the rat model caused intra-amniotic inflammation, but it did not lead to PTD.Conclusion: The intra-amniotic administration of the triggering agents can be used to study intra-amniotic inflammatory response and intra-amniotic inflammation/infection in the rodents model.
Collapse
Affiliation(s)
- Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Peter Vescicik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tomas Faist
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Department of Genetics and Bioinformatics, Domain of Health Data and Digitalisation, Institute of Public Health, Oslo, Norway
| | - Ivana Musilova
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
59
|
Adu-Bonsaffoh K, Oppong SA, Dassah ET, Seffah JD. Challenges in preterm birth research: Ghanaian perspective. Placenta 2020; 98:24-28. [PMID: 33039028 DOI: 10.1016/j.placenta.2020.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 01/30/2023]
Abstract
Preterm birth is highly prevalent in Ghana. It is a major public health concern because of the high burden as well as the associated immediate and long-term consequences including increased healthcare cost. Studies conducted in high-income countries may not be sufficiently generalizable in our context. Locally generated evidence-based interventions will be indispensable in improving the clinical management and prevention of preterm birth in the country. However, there are limited published literature on preterm birth and prematurity in the country. This review seeks to discuss the major challenges associated with preterm birth research in Ghana and proposes evidence-based strategies to improve biomedical and epidemiological research on preterm birth and prematurity. The limited high quality preterm birth research is partly attributable to a variety of challenges related to accurate gestational age estimation, research training, capacity and support including funding, efficient ethics committees, local and international collaboration as well as effective health management information systems. Other related challenges include unavailability of reliable internet connectivity, poor compensation for researchers and lack of conductive research environment. There is the need to expedite advocacy on implementation of practical interventions and strategies aimed at increasing high quality research in the area of preterm birth and prematurity in the country. A paradigm shift in preterm birth research with appropriate integration of concerted multidisciplinary research groups should be constituted to put basic science research to clinical practice as well as the prevention of preterm birth in the country.
Collapse
Affiliation(s)
- K Adu-Bonsaffoh
- Department of Obstetrics and Gynecology, School of Medicine and Dentistry, University of Ghana, Accra, Ghana.
| | - S A Oppong
- Department of Obstetrics and Gynecology, School of Medicine and Dentistry, University of Ghana, Accra, Ghana
| | - E T Dassah
- School of Public Health, Kwame Nkrumah University of Science & Technology, Kumasi, Ghana; Department of Obstetrics & Gynecology, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - J D Seffah
- Department of Obstetrics and Gynecology, School of Medicine and Dentistry, University of Ghana, Accra, Ghana
| |
Collapse
|
60
|
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: 35] [Impact Index Per Article: 7.0] [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
|
61
|
Zhang Y, Mustieles V, Yland J, Braun JM, Williams PL, Attaman JA, Ford JB, Calafat AM, Hauser R, Messerlian C. Association of Parental Preconception Exposure to Phthalates and Phthalate Substitutes With Preterm Birth. JAMA Netw Open 2020; 3:e202159. [PMID: 32259265 PMCID: PMC7139277 DOI: 10.1001/jamanetworkopen.2020.2159] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
IMPORTANCE Although phthalate exposure during pregnancy has been associated with preterm birth, the association of preconception exposure in either parent with preterm birth constitutes a knowledge gap. OBJECTIVE To examine the association of paternal and maternal preconception urinary concentrations of biomarkers of phthalates and phthalate substitutes with singleton preterm birth. DESIGN, SETTING, AND PARTICIPANTS This study, conducted at an academic fertility center in Boston, Massachusetts, included a prospective preconception cohort of subfertile couples comprising 419 mothers and 229 fathers and their 420 live-born singleton offspring born between January 1, 2005, and December 31, 2018. Statistical analysis was performed from August 1 to October 31, 2019. EXPOSURES Urinary concentrations of metabolites of phthalates and phthalate substitutes obtained before conception. MAIN OUTCOMES AND MEASURES Gestational age was abstracted from delivery records and validated using the American College of Obstetricians and Gynecologists guidelines for births after medically assisted reproduction. The risk ratio (RR) of preterm birth (live birth before 37 completed weeks' gestation) was estimated in association with urinary concentrations of 11 individual phthalate metabolites, the molar sum of 4 di-(2-ethylhexyl) phthalate (ΣDEHP) metabolites, and 2 metabolites of 1,2-cyclohexane dicarboxylic acid diisononyl ester (DINCH, a nonphthalate plasticizer substitute) using modified Poisson regression models adjusted for covariates. RESULTS The mean (SD) age of the 419 mothers was 34.7 (4.0) years, the mean (SD) age of the 229 fathers was 36.0 (4.5) years, and the mean (SD) gestational age of the 420 singleton children (217 boys) was 39.3 (1.7) weeks, with 34 (8%) born preterm. In adjusted models, maternal preconception ΣDEHP concentrations (RR, 1.50; 95% CI, 1.09-2.06; P = .01) and cyclohexane-1,2-dicarboxylic acid monohydroxy isononyl ester (MHiNCH, a metabolite of DINCH) concentrations (RR, 1.70; 95% CI, 0.89-3.24; P = .11) were associated with an increased risk of preterm birth. After additional adjustment for prenatal ΣDEHP or MHiNCH concentrations, the association of maternal preconception exposure to ΣDEHP and preterm birth remained robust (RR, 1.69; 95% CI, 1.17-2.44; P = .006), while the association of maternal preconception exposure to MHiNCH and preterm birth was attenuated (RR, 1.17; 95% CI, 0.49-2.81; P = .72). The remaining urinary metabolites examined in either parent showed no association with preterm birth. CONCLUSIONS AND RELEVANCE In this prospective cohort of subfertile couples, maternal preconception exposure to ΣDEHP metabolites was associated with an increased risk of preterm birth. The results suggest that female exposure to select phthalate plasticizers during the preconception period may be a potential risk factor for adverse pregnancy outcomes, which may need to be considered in preconception care strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- School of Health Humanities, Peking University Health Science Center, Beijing, China
| | - Vicente Mustieles
- University of Granada, Center for Biomedical Research, Instituto de Investigación Biosanitaria Ibs Granada, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada, Spain
| | - Jennifer Yland
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Joseph M. Braun
- Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island
| | - Paige L. Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jill A. Attaman
- Department of Obstetrics and Gynecology, Massachusetts General Hospital Fertility Center, Boston
| | - Jennifer B. Ford
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Antonia M. Calafat
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Obstetrics and Gynecology, Massachusetts General Hospital Fertility Center, Boston
| | - Carmen Messerlian
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
62
|
Mustieles V, Zhang Y, Yland J, Braun JM, Williams PL, Wylie BJ, Attaman JA, Ford JB, Azevedo A, Calafat AM, Hauser R, Messerlian C. Maternal and paternal preconception exposure to phenols and preterm birth. ENVIRONMENT INTERNATIONAL 2020; 137:105523. [PMID: 32120140 PMCID: PMC7169435 DOI: 10.1016/j.envint.2020.105523] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 05/08/2023]
Abstract
BACKGROUND Phenol exposure during pregnancy has been associated with preterm birth, but the potential effect of preconception exposure in either parent is unknown. There is a growing body of evidence to suggest that the preconception period is a critical window of vulnerability for adverse pregnancy outcomes. OBJECTIVE We examined whether maternal and paternal preconception urinary concentrations of select phenols were associated with the risk of preterm birth among couples attending fertility care. METHODS The analysis included 417 female and 229 male participants of the Environment and Reproductive Health (EARTH) Study who gave birth to 418 singleton infants between 2005 and 2018 and for whom we had phenol biomarkers quantified in at least one urine sample collected before conception. Mothers and fathers provided an average of 4 and 3 urine samples during the preconception period, respectively. We calculated the geometric mean of bisphenol A (BPA), bisphenol S (BPS), benzophenone-3, triclosan, and the molar sum of parabens (ΣParabens) urinary concentrations to estimate each participant's preconception exposure. Risk ratios (RRs) of preterm birth (live birth before 37 completed weeks' gestation) were estimated using modified Poisson regression models adjusted for covariates. RESULTS The mean (SD) gestational age among singletons was 39.3 (1.7) weeks with 8% born preterm. A natural log-unit increase in maternal preconception BPA (RR 1.94; 95% CI: 1.20, 3.14) and BPS (RR 2.42; 95% CI: 1.01, 5.77) concentration was associated with an increased risk of preterm birth. These associations remained after further adjustment for maternal prenatal and paternal preconception biomarker concentrations. Paternal preconception ΣParabens concentrations showed a possible elevated risk of preterm birth (RR 1.36; 95% CI: 0.94, 1.96). No consistent pattern of association was observed for benzophenone-3 or triclosan biomarkers in either parent. DISCUSSION Maternal preconception urinary BPA and BPS concentrations, as well as paternal preconception urinary parabens concentrations were prospectively associated with a higher risk of preterm birth. Subfertile couples' exposure to select phenols during the preconception period may be an unrecognized risk factor for adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Vicente Mustieles
- University of Granada, Center for Biomedical Research (CIBM), Spain. Instituto de Investigación Biosanitaria Ibs GRANADA, Spain. Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 18100, Spain
| | - Yu Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; School of Health Humanities, Peking University Health Science Center, Beijing, Beijing, China
| | - Jennifer Yland
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Paige L Williams
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Blair J Wylie
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jill A Attaman
- Department of Obstetrics and Gynecology, Massachusetts General Hospital Fertility Center, Boston, MA, USA
| | - Jennifer B Ford
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexandra Azevedo
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Antonia M Calafat
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Carmen Messerlian
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
63
|
Busse M, Campe KNJ, Redlich A, Oettel A, Hartig R, Costa SD, Zenclussen AC. Regulatory B Cells Are Decreased and Impaired in Their Function in Peripheral Maternal Blood in Pre-term Birth. Front Immunol 2020; 11:386. [PMID: 32265904 PMCID: PMC7099879 DOI: 10.3389/fimmu.2020.00386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 02/18/2020] [Indexed: 01/01/2023] Open
Abstract
Preterm birth (PTB) is defined as birth before 37 completed weeks of gestation. The causes of PTB are multiple and complex, the underlying pathophysiology being largely unknown. Interferences in the fine-tuned balance of the maternal immune system have been pointed to as one possible cause of PTB. Regulatory B cells (Breg) are part of the adaptive immune response, and recent data suggest that they may contribute to a healthy pregnancy by their regulatory/suppressive function. We investigated the frequency of Breg cells in peripheral blood of women undergoing PTB and control women immediately before giving birth via cesarean section. We detected an enhanced number of B cells, but a reduced number of Breg cells in women delivering preterm. In addition, the percentage of IL-10-producing B cells was decreased in PTB following stimulation with TLR agonists CpG or LPS, alone or combined with CD40L. This was associated with increased levels of pro-inflammatory cytokines in maternal serum. Moreover, isolated maternal B cells before delivering premature babies secreted higher level of the pro-inflammatory cytokine IL-6. No alterations in the frequency of regulatory T cells were found. Our data indicate that alterations in the number and function of Breg cells in peripheral maternal blood contribute to the immunological changes observed in preterm delivery and suggest these cells as important regulators of maternal immune responses.
Collapse
Affiliation(s)
- Mandy Busse
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Kim-Norina Jutta Campe
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anke Redlich
- University Hospital for Gynecology, Obstetrics, and Reproductive Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Anika Oettel
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.,University Hospital for Gynecology, Obstetrics, and Reproductive Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Roland Hartig
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Serban-Dan Costa
- University Hospital for Gynecology, Obstetrics, and Reproductive Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Ana Claudia Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
64
|
Park B, Khanam R, Vinayachandran V, Baqui AH, London SJ, Biswal S. Epigenetic biomarkers and preterm birth. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa005. [PMID: 32551139 PMCID: PMC7293830 DOI: 10.1093/eep/dvaa005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 05/06/2023]
Abstract
Preterm birth (PTB) is a major public health challenge, and novel, sensitive approaches to predict PTB are still evolving. Epigenomic markers are being explored as biomarkers of PTB because of their molecular stability compared to gene expression. This approach is also relatively new compared to gene-based diagnostics, which relies on mutations or single nucleotide polymorphisms. The fundamental principle of epigenome diagnostics is that epigenetic reprogramming in the target tissue (e.g. placental tissue) might be captured by more accessible surrogate tissue (e.g. blood) using biochemical epigenome assays on circulating DNA that incorporate methylation, histone modifications, nucleosome positioning, and/or chromatin accessibility. Epigenomic-based biomarkers may hold great potential for early identification of the majority of PTBs that are not associated with genetic variants or mutations. In this review, we discuss recent advances made in the development of epigenome assays focusing on its potential exploration for association and prediction of PTB. We also summarize population-level cohort studies conducted in the USA and globally that provide opportunities for genetic and epigenetic marker development for PTB. In addition, we summarize publicly available epigenome resources and published PTB studies. We particularly focus on ongoing genome-wide DNA methylation and epigenome-wide association studies. Finally, we review the limitations of current research, the importance of establishing a comprehensive biobank, and possible directions for future studies in identifying effective epigenome biomarkers to enhance health outcomes for pregnant women at risk of PTB and their infants.
Collapse
Affiliation(s)
- Bongsoo Park
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, International Center for Maternal and Newborn Health, Baltimore, MD 21205, USA
| | - Vinesh Vinayachandran
- School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Abdullah H Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, International Center for Maternal and Newborn Health, Baltimore, MD 21205, USA
| | - Stephanie J London
- Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
65
|
Migault L, Garlantézec R, Piel C, Marchand-Martin L, Orazio S, Cheminat M, Zaros C, Carles C, Cardis E, Ancel PY, Charles MA, de Seze R, Baldi I, Bouvier G. Maternal cumulative exposure to extremely low frequency electromagnetic fields, prematurity and small for gestational age: a pooled analysis of two birth cohorts. Occup Environ Med 2019; 77:22-31. [DOI: 10.1136/oemed-2019-105785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/12/2019] [Accepted: 11/06/2019] [Indexed: 01/29/2023]
Abstract
BackgroundData on the effects of extremely low frequency electromagnetic fields (ELF-EMF) on pregnancy outcomes are inconclusive.ObjectiveTo study the relation between maternal cumulative exposure to ELF-EMF during pregnancy and the risk of prematurity or small for gestational age (SGA) in a pooled analysis of two French birth cohorts.MethodsElfe and Epipage2 are both population-based birth cohorts initiated in 2011 and included 18 329 and 8400 births, respectively. Health data and household, mother and child characteristics were obtained from medical records and questionnaires at maternity and during follow-up. A job exposure matrix was used to assess cumulative exposure to ELF-EMF during three periods: (1) until 15 weeks of gestation, (2) until 28 weeks of gestation and (3) until 32 weeks of gestation. Analyses were restricted to single live births in mainland France and to mothers with documented jobs (N=19 894). Adjusted logistic regression models were used.ResultsAccording to the period studied, 3.2%–4% of mothers were classified as highly exposed. Results were heterogeneous. Increased risks of prematurity were found among low exposed mothers for the three periods, and no association was observed among the most exposed (OR1=0.92 (95% CI 0.74 to 1.15); OR2=0.98 (95% CI 0.80 to 1.21); OR3=1.14 (95% CI 0.92 to 1.41)). For SGA, no association was observed with the exception of increased risk among the low exposed mothers in period 2 and the most exposed in period 3 (OR=1.25 (95% CI 1.02 to 1.53)).ConclusionSome heterogeneous associations between ELF-EMF exposure and prematurity and SGA were observed. However, due to heterogeneity (ie, their independence regarding the level of exposure), associations cannot be definitely explained by ELF-EMF exposure.
Collapse
|
66
|
Ulrich D, Schneider V, Pichler G, Haas J, Culea V, Joksch M, Mager C, Schmied C, Lakovschek IC, Lang U, Reif P. Neonatal Outcome After Hexoprenaline Compared with Atosiban After Preterm Premature Rupture of Membranes. JOURNAL OF FETAL MEDICINE 2019. [DOI: 10.1007/s40556-019-00225-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AbstractPreterm premature rupture of membranes (PPROM) occurs in up to 3% of all pregnancies. Only few comparative studies have investigated potential risks and benefits between different tocolytic substances in women with PPROM. The aim of this study was to compare the neonatal short term outcome after tocolysis with Atosiban or Hexoprenaline in women with PPROM. This is a retrospective observational cohort study of women with PPROM between 24 and 32 weeks of gestation comparing neonatal and maternal outcome after tocolysis with atosiban or hexoprenaline. Outcome parameters were short term neonatal outcome, maternal tocolytic efficacy, effectiveness and tolerability and neonatal neurodevelopmental long-term outcome. Continuous variables were compared using t-Test or Mann–Whitney U test, as appropriate. For categorical variables Chi-square after Pearson and Fisher exact-test were used to compare the two groups. 93 women were included into this study with 42 women receiving hexoprenaline and 51 women receiving atosiban as primary tocolytic treatment. Mean gestational age was 29 weeks in both groups at the time PPROM. No differences were found for any short term neonatal outcome parameters, tocolytic efficacy, effectiveness and tolerability and neonatal neurodevelopmental long-term outcome. Both hexoprenaline and atosiban do not affect the short and long term neonatal outcome in women with PPROM for the time of lung maturation.
Collapse
|
67
|
Placental Structure in Preterm Birth Among HIV-Positive Versus HIV-Negative Women in Kenya. J Acquir Immune Defic Syndr 2019; 80:94-102. [PMID: 30272633 PMCID: PMC6289800 DOI: 10.1097/qai.0000000000001871] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Preterm birth (PTB) is a major cause of infant morbidity and mortality in developing countries. Recent data suggest that in addition to Human Immunodeficiency Virus (HIV) infection, use of antiretroviral therapy (ART) increases the risk of PTB. As the mechanisms remain unexplored, we conducted this study to determine whether HIV and ART were associated with placental changes that could contribute to PTB. Setting: We collected and evaluated placentas from 38 HIV-positive women on ART and 43 HIV-negative women who had preterm deliveries in Nairobi, Kenya. Methods: Anatomical features of the placentas were examined at gross and microscopic levels. Cases were matched for gestational age and compared by the investigators who were blinded to maternal HIV serostatus. Results: Among preterm placentas, HIV infection was significantly associated with thrombosis (P = 0.001), infarction (P = 0.032), anomalies in cord insertion (P = 0.02), gross evidence of membrane infection (P = 0.043), and reduced placental thickness (P = 0.010). Overall, preterm placentas in both groups were associated with immature villi, syncytial knotting, villitis, and deciduitis. Features of HIV-positive versus HIV-negative placentas included significant fibrinoid deposition with villus degeneration, syncytiotrophoblast delamination, red blood cell adhesion, hypervascularity, and reduction in both surface area and perimeter of the terminal villi. Conclusions: These results imply that HIV infection and/or ART are associated with morphological changes in preterm placentas that contribute to delivery before 37 weeks. Hypervascularity suggests that the observed pathologies may be attributable, in part, to hypoxia. Further research to explore potential mechanisms will help elucidate the pathways that are involved perhaps pointing to interventions for decreasing the risk of prematurity among HIV-positive women.
Collapse
|
68
|
Stefanovic V, Andersson S, Vento M. Oxidative stress - Related spontaneous preterm delivery challenges in causality determination, prevention and novel strategies in reduction of the sequelae. Free Radic Biol Med 2019; 142:52-60. [PMID: 31185254 DOI: 10.1016/j.freeradbiomed.2019.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022]
Abstract
Spontaneous preterm birth (PTB) is one of the major complications of pregnancy and the main cause of neonatal mortality and morbidity. Despite the efforts devoted to the understanding of this obstetrical syndrome and improved medical care, there has been a tendency for the PTB rate to increase in the last decades globally. The costs of the screening for spontaneous PTB, its management, and treatment of the sequelae represent a major burden to the health service economy of high-income countries. In this scenario, it has been widely acknowledged that oxidative stress (OS) plays an important role in the pathogenicity of human disease in wide range of areas of medicine. There is an emerging evidence that an imbalance between pro-and-antioxidants may be associated with spontaneous PTB. However, there are still many controversies on the mechanisms by which OS are involved in the pathogenesis of prematurity. Moreover, the crucial question whether the OS is the cause or consequence of the disease is yet to be answered. The purpose of this article is to briefly summarize the current knowledge and controversies on oxidative stress-related spontaneous PTB and to give a critical approach on future perspectives on this topic as a classical example of translational medicine. Placenta-mediated pregnancy adverse outcome associated with OS leading to iatrogenic PTB (e.g. pre-eclampsia, intrauterine growth restriction, gestational diabetes) will not be discussed.
Collapse
Affiliation(s)
- Vedran Stefanovic
- Department of Obstetrics and Gynecology, Fetomaternal Medical Center, Helsinki University and Helsinki University Hospital, Finland
| | - Sture Andersson
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Maximo Vento
- Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain; Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain.
| |
Collapse
|
69
|
Samuel TM, Sakwinska O, Makinen K, Burdge GC, Godfrey KM, Silva-Zolezzi I. Preterm Birth: A Narrative Review of the Current Evidence on Nutritional and Bioactive Solutions for Risk Reduction. Nutrients 2019; 11:E1811. [PMID: 31390765 PMCID: PMC6723114 DOI: 10.3390/nu11081811] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/18/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022] Open
Abstract
Preterm birth (PTB) (<37 weeks of gestation) is the leading cause of newborn death and a risk factor for short and long-term adverse health outcomes. Most cases are of unknown cause. Although the mechanisms triggering PTB remain unclear, an inappropriate increase in net inflammatory load seems to be key. To date, interventions that reduce the risk of PTB are effective only in specific groups of women, probably due to the heterogeneity of its etiopathogenesis. Use of progesterone is the most effective, but only in singleton pregnancies with history of PTB. Thus, primary prevention is greatly needed and nutritional and bioactive solutions are a promising alternative. Among these, docosahexaenoic acid (DHA) is the most promising to reduce the risk for early PTB. Other potential nutrient interventions include the administration of zinc (possibly limited to populations with low nutritional status or poor zinc status) and vitamin D; additional preliminary evidence exists for vitamin A, calcium, iron, folic acid, combined iron-folate, magnesium, multiple micronutrients, and probiotics. Considering the public health relevance of PTB, promising interventions should be studied in large and well-designed clinical trials. The objective of this review is to describe, summarize, and discuss the existing evidence on nutritional and bioactive solutions for reducing the risk of PTB.
Collapse
Affiliation(s)
| | | | | | - Graham C Burdge
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | |
Collapse
|
70
|
Huang J, Meng J, Choonara I, Xiong T, Wang Y, Wang H, Qu Y, Mu D. Antenatal infection and intraventricular hemorrhage in preterm infants: A meta-analysis. Medicine (Baltimore) 2019; 98:e16665. [PMID: 31374040 PMCID: PMC6709165 DOI: 10.1097/md.0000000000016665] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The aim of this study was to summarize current evidence evaluating the association between antenatal infection and intraventricular hemorrhage (IVH) in preterm infants. MATERIALS AND METHODS We searched for published articles on antenatal infection and IVH in 3 English (PubMed, the Cochrane Library, and EBSCO) and 3 Chinese (VEIPU, CNKI, and WANFANG) databases on May 19, 2019. In addition, the references of these articles were screened. The included studies had to meet all of the following criteria: preterm infants (<37 weeks); comparing antenatal infection with no infection; the outcomes included IVH (all grades), mild IVH, or sereve IVH; the type of study was randomized controlled trial or cohort study. RESULTS A total of 23 cohort studies involving 13,605 preterm infants met our inclusion criteria. Antenatal infection increased the risk of IVH (odds ratios ([OR] 2.18, 95% confidence intervals [CI] 1.58-2.99), mild IVH (OR 1.95, 95% CI 1.09-3.49) and severe IVH (OR 2.65, 95% CI 1.52-4.61). For type of antenatal infection, the ORs and 95% CI were as follows: 2.21 (1.60-3.05) for chorioamnionitis, 2.26 (1.55-3.28) for histologic chorioamnionitis, 1.88 (1.22-2.92) for clinical chorioamnionitis, and 1.88 (1.14-3.10) for ureaplasma. CONCLUSIONS Antenatal infection may increase the risk of developing IVH in the preterm infant. The evidence base is however of low quality and well-designed studies are needed.
Collapse
Affiliation(s)
- Jinglan Huang
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Junjie Meng
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Imti Choonara
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, UK
| | - Tao Xiong
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yibin Wang
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
71
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
72
|
Sun X, Guo JH, Zhang D, Chen JJ, Lin WY, Huang Y, Chen H, Huang WQ, Liu Y, Tsang LL, Yu MK, Chung YW, Jiang X, Huang H, Chan HC, Ruan YC. Activation of the epithelial sodium channel (ENaC) leads to cytokine profile shift to pro-inflammatory in labor. EMBO Mol Med 2019; 10:emmm.201808868. [PMID: 30154237 PMCID: PMC6402451 DOI: 10.15252/emmm.201808868] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The shift of cytokine profile from anti‐ to pro‐inflammatory is the most recognizable sign of labor, although the underlying mechanism remains elusive. Here, we report that the epithelial sodium channel (ENaC) is upregulated and activated in the uterus at labor in mice. Mechanical activation of ENaC results in phosphorylation of CREB and upregulation of pro‐inflammatory cytokines as well as COX‐2/PGE2 in uterine epithelial cells. ENaC expression is also upregulated in mice with RU486‐induced preterm labor as well as in women with preterm labor. Interference with ENaC attenuates mechanically stimulated uterine contractions and significantly delays the RU486‐induced preterm labor in mice. Analysis of a human transcriptome database for maternal–fetus tissue/blood collected at onset of human term and preterm births reveals significant and positive correlation of ENaC with labor‐associated pro‐inflammatory factors in labored birth groups (both term and preterm), but not in non‐labored birth groups. Taken together, the present finding reveals a pro‐inflammatory role of ENaC in labor at term and preterm, suggesting it as a potential target for the prevention and treatment of preterm labor.
Collapse
Affiliation(s)
- Xiao Sun
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jing Hui Guo
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China.,Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Dan Zhang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun-Jiang Chen
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China.,Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Wei Yin Lin
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun Huang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Chen
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wen Qing Huang
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yifeng Liu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lai Ling Tsang
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Mei Kuen Yu
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yiu Wa Chung
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaohua Jiang
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hefeng Huang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,International Peace Maternal and Child Health Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
73
|
Hoh JK, Lappas M, Liu C, Qiao C, Pallavi K, Takeda J, Kim YJ. Preterm birth rate and dilemma of preterm labor treatment in Asia. Placenta 2019; 79:68-71. [DOI: 10.1016/j.placenta.2019.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/20/2018] [Accepted: 01/04/2019] [Indexed: 01/05/2023]
|
74
|
Weckend MJ, Spineli LM, Grylka-Baeschlin S, Gross MM. Association between increased antenatal vaginal pH and preterm birth rate: a systematic review. J Perinat Med 2019; 47:142-151. [PMID: 29995636 DOI: 10.1515/jpm-2018-0097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/11/2018] [Indexed: 12/28/2022]
Abstract
Background Worldwide, 14.9 million infants (11%) are born preterm each year. Up to 40% of preterm births (PTBs) are associated with genital tract infections. The vaginal pH can reflect changes in the vaginal milieu and, if elevated, indicates an abnormal flora or infection. Objective The aim of the study was to investigate whether an increased antenatal vaginal pH >4.5 in pre-labour pregnant women is associated with an increased PTB rate <37 completed weeks gestation. Search strategy Key databases included SCOPUS, EMBASE, MEDLINE, PsycInfo and the Cochrane Central Register of Controlled Trials, complemented by hand search, up to January 2017. Selection criteria Primary research reporting vaginal pH assessment in pre-labour pregnant women and PTB rate. Data collection and analysis Data extraction and appraisal were carried out in a pre-defined standardised manner, applying the Newcastle-Ottawa scale (NOS) and Cochrane risk of bias tool. Analysis included calculation of risk difference (RD) and narrative synthesis. It was decided to abstain from pooling of the studies due to missing information in important moderators. Main results Of 986 identified records, 30 were included in the systematic review. The risk of bias was considered mostly high (40%) or moderate (37%). Fifteen studies permitted a calculation of RD. Of these, 14 (93%) indicated a positive association between increased antenatal vaginal pH and PTB (RD range: 0.02-0.75). Conclusion An increased antenatal vaginal pH >4.5 may be associated with a higher risk for PTB. It is recommended to conduct a randomised controlled trial (RCT) to investigate the effectiveness of antenatal pH screening to prevent PTB. Tweetable abstract Pregnant women with an increased vaginal pH >4.5 may be at higher risk to experience preterm birth.
Collapse
Affiliation(s)
- Marina J Weckend
- Midwifery Research and Education Unit, Department of Obstetric, Gynaecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | - Loukia M Spineli
- Institute for Biostatistics, Hannover Medical School, Hannover, Germany
| | - Susanne Grylka-Baeschlin
- Midwifery Research and Education Unit, Department of Obstetric, Gynaecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | - Mechthild M Gross
- Midwifery Research and Education Unit, Department of Obstetric, Gynaecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
75
|
Soltani M, Tabatabaee HR, Saeidinejat S, Eslahi M, Yaghoobi H, Mazloumi E, Rajabi A, Ghasemi A, Keyghobadi N, Enayatrad M, Noori A, Hashemi SA, Zolfizadeh F, Mahdavi S, Valadbeigi T, Etemad K, Taghipour A, Salehnasab C, Hajipour M. Assessing the risk factors before pregnancy of preterm births in Iran: a population-based case-control study. BMC Pregnancy Childbirth 2019; 19:57. [PMID: 30727983 PMCID: PMC6364407 DOI: 10.1186/s12884-019-2183-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/08/2019] [Indexed: 12/04/2022] Open
Abstract
Background Preterm birth is a major cause of prenatal and postnatal mortality particularly in developing countries. This study investigated the maternal risk factors associated with the risk of preterm birth. Methods A population-based case-control study was conducted in several provinces of Iran on 2463 mothers referred to health care centers. Appropriate descriptive and analytical statistical methods were used to evaluate the association between maternal risk factors and the risk of preterm birth. All tests were two-sided, and P values < 0.05 were considered to be statistically significant. Results The mean gestational age was 31.5 ± 4.03 vs. 38.8 ± 1.06 weeks in the case and control groups, respectively. Multivariate regression analysis showed a statistically significant association between preterm birth and mother’s age and ethnicity. Women of Balooch ethnicity and age ≥ 35 years were significantly more likely to develop preterm birth (OR: 1.64; 95% CI: 1.01–-2.44 and OR: 9.72; 95% CI: 3.07–30.78, respectively). However, no statistically significant association was observed between preterm birth and mother’s place of residence, level of education, past history of cesarean section, and BMI. Conclusion Despite technological advances in the health care system, preterm birth still remains a major concern for health officials. Providing appropriate perinatal health care services as well as raising the awareness of pregnant women, especially for high-risk groups, can reduce the proportion of preventable preterm births.
Collapse
Affiliation(s)
- Maryam Soltani
- Razi Clinical Research Development Unit(RCRDU), Birjand University of Medical Sciences(BUMS), Birjand, Iran
| | - Hamid Reza Tabatabaee
- Research Center for Health Sciences, Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahin Saeidinejat
- Department of Health Education and Health Promotion, School of Health Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Eslahi
- Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Halime Yaghoobi
- Social Determinants in Health Promotion Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ehsan Mazloumi
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolhalim Rajabi
- Department of Epidemiology, Faculty of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Ghasemi
- Birjand University of Medical Sciences, Birjand, Iran
| | - Naeimeh Keyghobadi
- Department of Biostatistics Epidemiology, Health Faculty, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mostafa Enayatrad
- Epidemiology Department, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abed Noori
- Medical Education, Health Management and Social Development Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyyed Aliasghar Hashemi
- Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Zolfizadeh
- Health Care Management, Mother and Child Welfare Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sepideh Mahdavi
- Clinical Research Development Unit, Imam Hossein Hospital, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Tannaz Valadbeigi
- Clinical Research Development Unit, Imam Hossein Hospital, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Koorosh Etemad
- Department of Epidemiology, Environmental and Occupational Hazards Control Research Center, Faculty of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Taghipour
- Health Sciences Research Centre, Cancer Research Center, Department of Biostatistics and Epidemiology, School of Health, Faculty of Mashhad University of Medical Sciences, Mashhad, Iran
| | - Cirruse Salehnasab
- Social Determinants of Health Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahmoud Hajipour
- Student Research Committee, Epidemiology Department, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
76
|
Kennedy SH, Victora CG, Craik R, Ash S, Barros FC, Barsosio HC, Berkley JA, Carvalho M, Fernandes M, Cheikh Ismail L, Lambert A, Lindgren CM, McGready R, Munim S, Nellåker C, Noble JA, Norris SA, Nosten F, Ohuma EO, Papageorghiou AT, Stein A, Stones W, Tshivuila-Matala COO, Staines Urias E, Vatish M, Wulff K, Zainab G, Zondervan KT, Uauy R, Bhutta ZA, Villar J. Deep clinical and biological phenotyping of the preterm birth and small for gestational age syndromes: The INTERBIO-21 st Newborn Case-Control Study protocol. Gates Open Res 2019; 2:49. [PMID: 31172050 PMCID: PMC6545521 DOI: 10.12688/gatesopenres.12869.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background: INTERBIO-21
st is Phase II of the INTERGROWTH-21
st Project, the population-based, research initiative involving nearly 70,000 mothers and babies worldwide coordinated by Oxford University and performed by a multidisciplinary network of more than 400 healthcare professionals and scientists from 35 institutions in 21 countries worldwide. Phase I, conducted 2008-2015, consisted of nine complementary studies designed to describe optimal human growth and neurodevelopment, based conceptually on the WHO prescriptive approach. The studies generated a set of international standards for monitoring growth and neurodevelopment, which complement the existing WHO Child Growth Standards. Phase II aims to improve the functional classification of the highly heterogenous preterm birth and fetal growth restriction syndromes through a better understanding of how environmental exposures, clinical conditions and nutrition influence patterns of human growth from conception to childhood, as well as specific neurodevelopmental domains and associated behaviors at 2 years of age. Methods: In the INTERBIO-21
st Newborn Case-Control Study, a major component of Phase II, our objective is to investigate the mechanisms potentially responsible for preterm birth and small for gestational age and their interactions, using deep phenotyping of clinical, growth and epidemiological data and associated nutritional, biochemical, omic and histological profiles. Here we describe the study sites, population characteristics, study design, methodology and standardization procedures for the collection of longitudinal clinical data and biological samples (maternal blood, umbilical cord blood, placental tissue, maternal feces and infant buccal swabs) for the study that was conducted between 2012 and 2018 in Brazil, Kenya, Pakistan, South Africa, Thailand and the UK. Discussion: Our study provides a unique resource for the planned analyses given the range of potentially disadvantageous exposures (including poor nutrition, pregnancy complications and infections) in geographically diverse populations worldwide. The study should enhance current medical knowledge and provide new insights into environmental influences on human growth and neurodevelopment.
Collapse
Affiliation(s)
- Stephen H Kennedy
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Cesar G Victora
- Programa de Pós-Graduação em Epidemiologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rachel Craik
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Stephen Ash
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Ludwig Institute, University of Oxford, Oxford, UK
| | - Fernando C Barros
- Programa de Pós-Graduação em Saúde e Comportamento, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Hellen C Barsosio
- KEMRI-Coast Centre for Geographical Medicine and Research, University of Oxford, Kilifi, Kenya
| | - James A Berkley
- KEMRI-Coast Centre for Geographical Medicine and Research, University of Oxford, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Maria Carvalho
- Faculty of Health Sciences, Aga Khan University, Nairobi, Kenya
| | - Michelle Fernandes
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Faculty of Medicine, Department of Paediatrics, University of Southampton, Southampton, UK
| | - Leila Cheikh Ismail
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ann Lambert
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Shama Munim
- Department of Obstetrics and Gynaecology, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Christoffer Nellåker
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Julia A Noble
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Shane A Norris
- SAMRC Developmental Pathways For Health Research Unit, Department of Paediatrics & Child Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Eric O Ohuma
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Centre for Statistics in Medicine, Botnar Research Centre, University of Oxford, Oxford, UK.,Center for Global Child Health, Hospital for Sick Children, Toronto, Canada
| | - Aris T Papageorghiou
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Alan Stein
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - William Stones
- Faculty of Health Sciences, Aga Khan University, Nairobi, Kenya.,Departments of Public Health and Obstetrics & Gynaecology, Malawi College of Medicine, Blantyre, Malawi
| | - Chrystelle O O Tshivuila-Matala
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,SAMRC Developmental Pathways For Health Research Unit, Department of Paediatrics & Child Health, University of the Witwatersrand, Johannesburg, South Africa.,Health, Nutrition & Population Global Practice, World Bank Group, Washington, DC, USA
| | - Eleonora Staines Urias
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Katharina Wulff
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Ghulam Zainab
- Department of Obstetrics and Gynaecology, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Krina T Zondervan
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ricardo Uauy
- Division of Paediatrics, Pontifical Universidad Catolica de Chile, Santiago, Chile.,Department of Nutrition and Public Health Interventions Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Zulfiqar A Bhutta
- Center for Global Child Health, Hospital for Sick Children, Toronto, Canada.,Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - José Villar
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| |
Collapse
|
77
|
Kennedy SH, Victora CG, Craik R, Ash S, Barros FC, Barsosio HC, Berkley JA, Carvalho M, Fernandes M, Cheikh Ismail L, Lambert A, Lindgren CM, McGready R, Munim S, Nellåker C, Noble JA, Norris SA, Nosten F, Ohuma EO, Papageorghiou AT, Stein A, Stones W, Tshivuila-Matala COO, Staines Urias E, Vatish M, Wulff K, Zainab G, Zondervan KT, Uauy R, Bhutta ZA, Villar J. Deep clinical and biological phenotyping of the preterm birth and small for gestational age syndromes: The INTERBIO-21 st Newborn Case-Control Study protocol. Gates Open Res 2019. [PMID: 31172050 DOI: 10.12688/gatesopenres.12869.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: INTERBIO-21 st is Phase II of the INTERGROWTH-21 st Project, the population-based, research initiative involving nearly 70,000 mothers and babies worldwide coordinated by Oxford University and performed by a multidisciplinary network of more than 400 healthcare professionals and scientists from 35 institutions in 21 countries worldwide. Phase I, conducted 2008-2015, consisted of nine complementary studies designed to describe optimal human growth and neurodevelopment, based conceptually on the WHO prescriptive approach. The studies generated a set of international standards for monitoring growth and neurodevelopment, which complement the existing WHO Child Growth Standards. Phase II aims to improve the functional classification of the highly heterogenous preterm birth and fetal growth restriction syndromes through a better understanding of how environmental exposures, clinical conditions and nutrition influence patterns of human growth from conception to childhood, as well as specific neurodevelopmental domains and associated behaviors at 2 years of age. Methods: In the INTERBIO-21 st Newborn Case-Control Study, a major component of Phase II, our objective is to investigate the mechanisms potentially responsible for preterm birth and small for gestational age and their interactions, using deep phenotyping of clinical, growth and epidemiological data and associated nutritional, biochemical, omic and histological profiles. Here we describe the study sites, population characteristics, study design, methodology and standardization procedures for the collection of longitudinal clinical data and biological samples (maternal blood, umbilical cord blood, placental tissue, maternal feces and infant buccal swabs) for the study that was conducted between 2012 and 2018 in Brazil, Kenya, Pakistan, South Africa, Thailand and the UK. Discussion: Our study provides a unique resource for the planned analyses given the range of potentially disadvantageous exposures (including poor nutrition, pregnancy complications and infections) in geographically diverse populations worldwide. The study should enhance current medical knowledge and provide new insights into environmental influences on human growth and neurodevelopment.
Collapse
Affiliation(s)
- Stephen H Kennedy
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Cesar G Victora
- Programa de Pós-Graduação em Epidemiologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rachel Craik
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Stephen Ash
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Ludwig Institute, University of Oxford, Oxford, UK
| | - Fernando C Barros
- Programa de Pós-Graduação em Saúde e Comportamento, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Hellen C Barsosio
- KEMRI-Coast Centre for Geographical Medicine and Research, University of Oxford, Kilifi, Kenya
| | - James A Berkley
- KEMRI-Coast Centre for Geographical Medicine and Research, University of Oxford, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Maria Carvalho
- Faculty of Health Sciences, Aga Khan University, Nairobi, Kenya
| | - Michelle Fernandes
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Faculty of Medicine, Department of Paediatrics, University of Southampton, Southampton, UK
| | - Leila Cheikh Ismail
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ann Lambert
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Shama Munim
- Department of Obstetrics and Gynaecology, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Christoffer Nellåker
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Julia A Noble
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Shane A Norris
- SAMRC Developmental Pathways For Health Research Unit, Department of Paediatrics & Child Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Eric O Ohuma
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Centre for Statistics in Medicine, Botnar Research Centre, University of Oxford, Oxford, UK.,Center for Global Child Health, Hospital for Sick Children, Toronto, Canada
| | - Aris T Papageorghiou
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Alan Stein
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - William Stones
- Faculty of Health Sciences, Aga Khan University, Nairobi, Kenya.,Departments of Public Health and Obstetrics & Gynaecology, Malawi College of Medicine, Blantyre, Malawi
| | - Chrystelle O O Tshivuila-Matala
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,SAMRC Developmental Pathways For Health Research Unit, Department of Paediatrics & Child Health, University of the Witwatersrand, Johannesburg, South Africa.,Health, Nutrition & Population Global Practice, World Bank Group, Washington, DC, USA
| | - Eleonora Staines Urias
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Katharina Wulff
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Ghulam Zainab
- Department of Obstetrics and Gynaecology, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Krina T Zondervan
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ricardo Uauy
- Division of Paediatrics, Pontifical Universidad Catolica de Chile, Santiago, Chile.,Department of Nutrition and Public Health Interventions Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Zulfiqar A Bhutta
- Center for Global Child Health, Hospital for Sick Children, Toronto, Canada.,Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - José Villar
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Oxford Maternal & Perinatal Health Institute, Green Templeton College, Oxford, UK
| |
Collapse
|
78
|
Good clinical practice advice: Prediction of preterm labor and preterm premature rupture of membranes. Int J Gynaecol Obstet 2019; 144:340-346. [PMID: 30710365 DOI: 10.1002/ijgo.12744] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
79
|
The Myometrium: From Excitation to Contractions and Labour. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:233-263. [PMID: 31183830 DOI: 10.1007/978-981-13-5895-1_10] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
We start by describing the functions of the uterus, its structure, both gross and fine, innervation and blood supply. It is interesting to note the diversity of the female's reproductive tract between species and to remember it when working with different animal models. Myocytes are the overwhelming cell type of the uterus (>95%) and our focus. Their function is to contract, and they have an intrinsic pacemaker and rhythmicity, which is modified by hormones, stretch, paracrine factors and the extracellular environment. We discuss evidence or not for pacemaker cells in the uterus. We also describe the sarcoplasmic reticulum (SR) in some detail, as it is relevant to calcium signalling and excitability. Ion channels, including store-operated ones, their contributions to excitability and action potentials, are covered. The main pathway to excitation is from depolarisation opening voltage-gated Ca2+ channels. Much of what happens downstream of excitability is common to other smooth muscles, with force depending upon the balance of myosin light kinase and phosphatase. Mechanisms of maintaining Ca2+ balance within the myocytes are discussed. Metabolism, and how it is intertwined with activity, blood flow and pH, is covered. Growth of the myometrium and changes in contractile proteins with pregnancy and parturition are also detailed. We finish with a description of uterine activity and why it is important, covering progression to labour as well as preterm and dysfunctional labours. We conclude by highlighting progress made and where further efforts are required.
Collapse
|
80
|
Parrish LK, Muglia LJ, DeFranco EA. Bleeding during pregnancy is associated with familial preterm birth. J Matern Fetal Neonatal Med 2019; 32:73-79. [PMID: 28835134 DOI: 10.1080/14767058.2017.1371693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/22/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE the purpose of this study is to identify risk factors for familial, likely genetically-determined, preterm birth. MATERIALS AND METHODS We performed a case-control study, enrolling 211 patients (103 cases and 108 controls). Cases delivered between 20 and 35 weeks gestation, with a prior preterm birth or first-degree relative born prematurely. Controls delivered between 37-42 weeks. Groups were compared using a comprehensive questionnaire validated by medical record. Multivariate logistic regression assessed risk factor associations. RESULTS Of cases, 30% reported bleeding during pregnancy compared with 5% of controls, adjusted odds ratio (adjOR) 9.0, 95%CI 3.31-24.47. Of cases that delivered at 20-28 weeks, 44.8% reported bleeding during pregnancy compared with 24.6% at 29-35 weeks, p = .04. Other associations were prior first-trimester miscarriage adjOR 2.55 (CI 1.21-5.35) or second-trimester miscarriage, adjOR 6.3 (CI 1.76-22.56). CONCLUSIONS Bleeding during pregnancy and prior miscarriage were significantly associated with familial preterm birth. The magnitude of effect for bleeding in pregnancy was higher with earlier preterm births. These associations warrant further investigation.
Collapse
Affiliation(s)
- Lindsay Kennedy Parrish
- a Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology , University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Louis J Muglia
- a Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology , University of Cincinnati College of Medicine , Cincinnati , OH , USA
- b Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA
| | - Emily A DeFranco
- a Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology , University of Cincinnati College of Medicine , Cincinnati , OH , USA
- b Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA
| |
Collapse
|
81
|
Di Renzo GC, Cabero Roura L, Facchinetti F, Helmer H, Hubinont C, Jacobsson B, Jørgensen JS, Lamont RF, Mikhailov A, Papantoniou N, Radzinsky V, Shennan A, Ville Y, Wielgos M, Visser GHA. Preterm Labor and Birth Management: Recommendations from the European Association of Perinatal Medicine. J Matern Fetal Neonatal Med 2018; 30:2011-2030. [PMID: 28482713 DOI: 10.1080/14767058.2017.1323860] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- G C Di Renzo
- a Department of Obstetrics and Gynecology , University of Perugia , Perugia , Italy
| | - L Cabero Roura
- b Department of Obstetrics and Gynecology , Hospital Vall D'Hebron , Barcelona , Spain
| | - F Facchinetti
- c Mother-Infant Department, School of Midwifery , University of Modena and Reggio Emilia , Italy
| | - H Helmer
- d Department of Obstetrics and Gynaecology , General Hospital, University of Vienna , Vienna , Austria
| | - C Hubinont
- e Department of Obstetrics , Saint Luc University Hospital, Université de Louvain , Brussels , Belgium
| | - B Jacobsson
- f Department of Obstetrics and Gynecology , Institute of Clinical Sciences, University of Gothenburg , Gothenburg , Sweden
| | - J S Jørgensen
- g Department of Obstetrics and Gynaecology , Odense University Hospital , Odense , Denmark
| | - R F Lamont
- h Department of Gynaecology and Obstetrics , University of Southern Denmark, Odense University Hospital , Odense , Denmark.,i Division of Surgery , University College London, Northwick Park Institute of Medical Research Campus , London , UK
| | - A Mikhailov
- j Department of Obstetrics and Gynecology , 1st Maternity Hospital, State University of St. Petersburg , Russia
| | - N Papantoniou
- k Department of Obstetrics and Gynaecology , Athens University School of Medicine , Athens , Greece
| | - V Radzinsky
- l Department of Medicine , Peoples' Friendship University of Russia , Moscow , Russia
| | - A Shennan
- m St. Thomas Hospital, Kings College London , UK
| | - Y Ville
- n Service d'Obstétrique et de Médecine Foetale , Hôpital Necker Enfants Malades , Paris , France
| | - M Wielgos
- p Department of Obstetrics and Gynecology , Medical University of Warsaw , Warsaw , Poland
| | - G H A Visser
- o Department of Obstetrics , University Medical Center , Utrecht , The Netherlands
| |
Collapse
|
82
|
Baker KK, Story WT, Walser-Kuntz E, Zimmerman MB. Impact of social capital, harassment of women and girls, and water and sanitation access on premature birth and low infant birth weight in India. PLoS One 2018; 13:e0205345. [PMID: 30296283 PMCID: PMC6175511 DOI: 10.1371/journal.pone.0205345] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/24/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Globally, preterm birth (PTB) and low infant birth weight (LBW) are leading causes of maternal and child morbidity and mortality. Inadequate water and sanitation access (WASH) are risk factors for PTB and LBW in low-income countries. Physical stress from carrying water and psychosocial stress from addressing sanitation needs in the open may be mechanisms underlying these associations. If so, then living in a community with strong social capital should be able to buffer the adverse effects of WASH on birth outcomes. The objective of this study is to assess the relationships between WASH access and social conditions (including harassment and social capital) on PTB and LBW outcomes among Indian women, and to test whether social conditions modified the association between WASH and birth outcomes. METHODS AND FINDINGS This cohort study examined the effect of pre-birth WASH and social conditions on self-reported PTB status and LBW status for 7,926 women who gave birth between 2004/2005 and 2011/2012 Waves of the India Human Development Survey. PTB and LBW occurred in 14.9% and 15.5% of women, respectively. After adjusting for maternal biological and socioeconomic conditions, PTB was associated with sharing a building/compound latrine (Odds Ratio (OR) = 1.55; 95% Confidence Interval (CI) = 1.01, 2.38) versus private latrine access, but suggested an effect in the opposite direction for sharing a community/public latrine (OR = 0.67; CI = 0.45, 1.01). Open defecation, type of drinking water source, minutes per day spent fetching water, and one-way time to a drinking water source were not associated with PTB. LBW was associated with spending more than two hours per day fetching water compared to less than two hours (OR = 1.33; CI = 1.05, 1.70) and suggested an association with open defecation (OR = 1.22; CI = 1.00, 1.48), but was not associated with other types of sanitation, type of drinking water source, or time to a drinking water source. Harassment of women and girls in the community was associated with both PTB (OR = 1.33; CI = 1.09, 1.62) and LBW (OR = 1.26; CI = 1.03, 1.54). The data also showed a possible association of local crime with LBW (OR = 1.30; CI = 1.00, 1.68). Statistically significant (p<0.05) evidence of effect modification was only found for collective efficacy on the association between type of sanitation access and PTB. In addition, stratified analyses identified differences in effect size for walking time to the primary drinking water source and PTB by crime, sanitation access and PTB by harassment, and total hours per day fetching water and LBW by collective efficacy. Limitations of this observational study include risk of bias, inability to confirm causality, reliance on self-reported outcomes, and limited sub-group sample sizes for testing effect modification. CONCLUSIONS The relationship between adverse birth outcomes and sanitation access, domestic water fetching, crime, and gender-based harassment suggests physical and psychosocial stress are possible mechanisms by which WASH access affects PTB and LBW among Indian women. Interventions that reduce domestic responsibilities related to water and sanitation and change social norms related to gender-based harassment may reduce rates of PTB and LBW in India.
Collapse
Affiliation(s)
- Kelly K. Baker
- Department of Occupational and Environmental Health, University of Iowa College of Public Health, IA, United States of America
| | - William T. Story
- Department of Community and Behavioral Health, University of Iowa College of Public Health, IA, United States of America
| | - Evan Walser-Kuntz
- Department of Biostatistics, University of Iowa College of Public Health, IA, United States of America
| | - M. Bridget Zimmerman
- Department of Biostatistics, University of Iowa College of Public Health, IA, United States of America
| |
Collapse
|
83
|
Cheon IS, Son YM, Jiang L, Goplen NP, Kaplan MH, Limper AH, Kita H, Paczesny S, Prakash YS, Tepper R, Ahlfeld SK, Sun J. Neonatal hyperoxia promotes asthma-like features through IL-33-dependent ILC2 responses. J Allergy Clin Immunol 2018; 142:1100-1112. [PMID: 29253513 PMCID: PMC6003836 DOI: 10.1016/j.jaci.2017.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 11/06/2017] [Accepted: 11/24/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Premature infants often require oxygen supplementation and, therefore, are exposed to oxidative stress. Following oxygen exposure, preterm infants frequently develop chronic lung disease and have a significantly increased risk of asthma. OBJECTIVE We sought to identify the underlying mechanisms by which neonatal hyperoxia promotes asthma development. METHODS Mice were exposed to neonatal hyperoxia followed by a period of room air recovery. A group of mice was also intranasally exposed to house dust mite antigen. Assessments were performed at various time points for evaluation of airway hyperresponsiveness, eosinophilia, mucus production, inflammatory gene expression, and TH and group 2 innate lymphoid cell (ILC2) responses. Sera from term- and preterm-born infants were also collected and levels of IL-33 and type 2 cytokines were measured. RESULTS Neonatal hyperoxia induced asthma-like features including airway hyperresponsiveness, mucus hyperplasia, airway eosinophilia, and type 2 pulmonary inflammation. In addition, neonatal hyperoxia promoted allergic TH responses to house dust mite exposure. Elevated IL-33 levels and ILC2 responses were observed in the lungs most likely due to oxidative stress caused by neonatal hyperoxia. IL-33 receptor signaling and ILC2s were vital for the induction of asthma-like features following neonatal hyperoxia. Serum IL-33 levels correlated significantly with serum levels of IL-5 and IL-13 but not IL-4 in preterm infants. CONCLUSIONS These data demonstrate that an axis involving IL-33 and ILC2s is important for the development of asthma-like features following neonatal hyperoxia and suggest therapeutic potential for targeting IL-33, ILC2s, and oxidative stress to prevent and/or treat asthma development related to prematurity.
Collapse
Affiliation(s)
- In Su Cheon
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Young Min Son
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Li Jiang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Nicholas P Goplen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Mark H Kaplan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind
| | - Andrew H Limper
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Hirohito Kita
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind
| | - Y S Prakash
- Department of Anesthesiology, Mayo Clinic College of Medicine and Science, Rochester, Minn; Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minn
| | - Robert Tepper
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind
| | - Shawn K Ahlfeld
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Jie Sun
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minn; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minn.
| |
Collapse
|
84
|
Gilman-Sachs A, Dambaeva S, Salazar Garcia MD, Hussein Y, Kwak-Kim J, Beaman K. Inflammation induced preterm labor and birth. J Reprod Immunol 2018; 129:53-58. [PMID: 30025845 DOI: 10.1016/j.jri.2018.06.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 06/25/2018] [Indexed: 01/23/2023]
Abstract
Preterm birth which occurs before 37 weeks gestation is one of the most common obstetrical complication in humans. After many studies, it appears that "not one answer fits all" regarding the risk factors, causes and the treatments for this syndrome. However, it is becoming more evident that one of the major risk factors is inflammation and/or infection in the fetoplacental unit. In animal models (usually consisting of mice injected with lipopolysaccharide at 14 days of gestation), IL-22 and IL-6 have been identified as factors related to preterm birth. There are some clinical tests available to determine the risk for preterm labor and delivery, which can be identified before, during early, or at mid-gestation. However, treatment of preterm birth with antibiotics so far has not been "curable" and studies using anti-inflammatory treatments are not readily available. More studies regarding causes and treatments for preterm labor and delivery in humans are necessary to prevent neonatal deaths and/or developmental abnormalities associated with this common syndrome.
Collapse
Affiliation(s)
- Alice Gilman-Sachs
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States.
| | - Svetlana Dambaeva
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States
| | - Maria D Salazar Garcia
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medicine School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, United States
| | - Youssef Hussein
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medicine School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, United States
| | - Joanne Kwak-Kim
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States; Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medicine School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, United States
| | - Kenneth Beaman
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States
| |
Collapse
|
85
|
Glavind MT, Møllgaard MV, Iversen ML, Arendt LH, Forman A. Obstetrical outcome in women with endometriosis including spontaneous hemoperitoneum and bowel perforation: a systematic review. Best Pract Res Clin Obstet Gynaecol 2018; 51:41-52. [DOI: 10.1016/j.bpobgyn.2018.01.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/23/2018] [Indexed: 12/29/2022]
|
86
|
Lim R, Barker G, Lappas M. IRF5 is increased in labouring myometrium and regulates pro-labour mediators. Reproduction 2018; 156:207-218. [PMID: 30006439 DOI: 10.1530/rep-18-0140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/12/2018] [Indexed: 01/16/2023]
Abstract
Preterm birth continues to be the leading cause of neonatal mortality and morbidities that can extend into adult life. Few treatment options stem from our incomplete understanding of the mechanisms of human labour and delivery. Activation of the inflammatory response in gestational tissues by inflammation and/or infection leads to the production of pro-inflammatory and pro-labour mediators, thus preterm birth. Interferon regulatory factor 5 (IRF5) has recently emerged as an important pro-inflammatory transcription factor involved in acute and chronic inflammation. The aims of this study were to determine the expression of IRF5 in human myometrium from labouring and non-labouring women, and whether IRF5 is involved in the genesis of pro-inflammatory and pro-labour mediators induced by pro-inflammatory cytokines or toll-like receptor (TLR) ligands. IRF5 mRNA and protein expression was significantly higher in human myometrium after spontaneous term labour, compared to non-labouring tissues. IRF5 mRNA expression was also significantly higher in primary myometrial cells treated with the pro-inflammatory cytokines IL1B or TNF. In primary myometrial cells, IRF5 knockdown by siRNA (siIRF5) was associated with significantly decreased expression and or secretion of pro-inflammatory cytokines (IL1A, IL6), chemokines (CXCL8, CCL2), adhesion molecules (ICAM1, VCAM1) and contraction-associated proteins PTGS2, PGF2α and PTGFR when in the presence of IL1B, TNF, fsl-1 (TLR2/6 ligand) or flagellin (TLR5 ligand). siIRF5-transfected cells also displayed decreased NF-κB RELA transcriptional activity in the presence of these preterm birth mediators. Our study suggests a novel role for IRF5 in the regulation of the inflammatory response in human myometrium.
Collapse
Affiliation(s)
- Ratana Lim
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Gillian Barker
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
87
|
Javorski N, Lima C, Silva L, Crovella S, de Azêvedo Silva J. Vitamin D receptor (VDR) polymorphisms are associated to spontaneous preterm birth and maternal aspects. Gene 2018; 642:58-63. [DOI: 10.1016/j.gene.2017.10.087] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 01/22/2023]
|
88
|
Ben Youssef G, Tourret M, Salou M, Ghazarian L, Houdouin V, Mondot S, Mburu Y, Lambert M, Azarnoush S, Diana JS, Virlouvet AL, Peuchmaur M, Schmitz T, Dalle JH, Lantz O, Biran V, Caillat-Zucman S. Ontogeny of human mucosal-associated invariant T cells and related T cell subsets. J Exp Med 2018; 215:459-479. [PMID: 29339446 PMCID: PMC5789419 DOI: 10.1084/jem.20171739] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/27/2017] [Accepted: 12/06/2017] [Indexed: 12/31/2022] Open
Abstract
There are very few human MAIT cells in cord blood. Ben Youssef et al. show that they slowly expand during childhood and point to a critical role of the TCRαβ repertoire in determining their unique ability to recognize MR1-restricted microbial antigens. Mucosal-associated invariant T (MAIT) cells are semi-invariant Vα7.2+ CD161highCD4− T cells that recognize microbial riboflavin precursor derivatives such as 5-OP-RU presented by MR1. Human MAIT cells are abundant in adult blood, but there are very few in cord blood. We longitudinally studied Vα7.2+ CD161high T cell and related subset levels in infancy and after cord blood transplantation. We show that Vα7.2+ and Vα7.2− CD161high T cells are generated early during gestation and likely share a common prenatal developmental program. Among cord blood Vα7.2+ CD161high T cells, the minority recognizing MR1:5-OP-RU display a TRAV/TRBV repertoire very similar to adult MAIT cells. Within a few weeks of life, only the MR1:5-OP-RU reactive Vα7.2+ CD161high T cells acquire a memory phenotype. Only these cells expand to form the adult MAIT pool, diluting out other Vα7.2+ CD161high and Vα7.2− CD161high populations, in a process requiring at least 6 years to reach adult levels. Thus, the high clonal size of adult MAIT cells is antigen-driven and likely due to the fine specificity of the TCRαβ chains recognizing MR1-restricted microbial antigens.
Collapse
Affiliation(s)
- Ghada Ben Youssef
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France
| | - Marie Tourret
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France
| | - Marion Salou
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Liana Ghazarian
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France
| | - Véronique Houdouin
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France.,Service de Gastroentérologie et Pneumologie Pédiatrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Stanislas Mondot
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Yvonne Mburu
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Marion Lambert
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France
| | - Saba Azarnoush
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France
| | - Jean-Sébastien Diana
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France
| | - Anne-Laure Virlouvet
- Service de Pédiatrie et Réanimation Néonatale, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Michel Peuchmaur
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France.,Service de Pathologie Pédiatrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thomas Schmitz
- Service d'Obstétrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Hugues Dalle
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France.,Service d'Hématologie Pédiatrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Olivier Lantz
- Institut Curie, PSL Research University, INSERM U932, Paris, France.,Centre d'Investigations Cliniques CIC-BT1428 IGR/Curie, Paris, France.,Equipe labellisée de la Ligue de Lutte contre le Cancer, Institut Curie, Paris, France.,Département de Biopathologie, Institut Curie, Paris, France
| | - Valérie Biran
- Service de Pédiatrie et Réanimation Néonatale, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sophie Caillat-Zucman
- Institut national de recherche médicale (INSERM) UMR1149, Center for Research on Inflammation, Paris Diderot University, Paris, France .,Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
89
|
Nevin CL, Formosa E, Maki Y, Matushewski B, Regnault TRH, Richardson BS. Maternal nutrient restriction in guinea pigs as an animal model for studying growth-restricted offspring with postnatal catch-up growth. Am J Physiol Regul Integr Comp Physiol 2018; 314:R647-R654. [PMID: 29351419 DOI: 10.1152/ajpregu.00317.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We determined the impact of moderate maternal nutrient restriction (MNR) in guinea pigs with fetal growth restriction (FGR) on offspring body and organ weights, hypothesizing that FGR-MNR animals will show catch-up growth but with organ-specific differences. Guinea pig sows were fed ad libitum (Control) or 70% of the control diet from 4 weeks preconception, switching to 90% at midpregnancy (MNR). Control newborns >95 g [appropriate for gestational age (AGA); n = 37] and MNR newborns <85 g (FGR; n = 37) were monitored until neonatal (~25 days) or adult (~110 days) necropsy. Birth weights and body/organ weights at necropsy were used to calculate absolute and fractional growth rates (FRs). FGR-MNR birth weights were decreased ~32% compared with the AGA-Controls. FGR-MNR neonatal whole body FRs were increased ~36% compared with Controls indicating catch-up growth, with values negatively correlated to birth weights indicating the degree of FGR leads to greater catch-up growth. However, the increase in organ FRs in the FGR-MNR neonates compared with Controls was variable, being similar for the brain and kidneys indicating comparable catch-up growth to that of the whole body and twofold increased for the liver but negligible for the heart indicating markedly increased and absent catch-up growth, respectively. While FGR-MNR body and organ weights were unchanged from the AGA-Controls by adulthood, whole body growth rates were increased. These findings confirm early catch-up growth in FGR-MNR guinea pigs but with organ-specific differences and enhanced growth rates by adulthood, which are likely to have implications for structural alterations and disease risk in later life.
Collapse
Affiliation(s)
- Catherine L Nevin
- Departments of Obstetrics and Gynecology, Physiology and Pharmacology, and Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London , Canada
| | - Evan Formosa
- Departments of Obstetrics and Gynecology, Physiology and Pharmacology, and Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London , Canada
| | - Yohei Maki
- Departments of Obstetrics and Gynecology, Physiology and Pharmacology, and Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London , Canada
| | - Brad Matushewski
- Departments of Obstetrics and Gynecology, Physiology and Pharmacology, and Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London , Canada
| | - Timothy R H Regnault
- Departments of Obstetrics and Gynecology, Physiology and Pharmacology, and Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London , Canada
| | - Bryan S Richardson
- Departments of Obstetrics and Gynecology, Physiology and Pharmacology, and Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London , Canada
| |
Collapse
|
90
|
Nallasamy S, Akins M, Tetreault B, Luby-Phelps K, Mahendroo M. Distinct reorganization of collagen architecture in lipopolysaccharide-mediated premature cervical remodeling. Biol Reprod 2018; 98:63-74. [PMID: 29161343 PMCID: PMC5803761 DOI: 10.1093/biolre/iox155] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/10/2017] [Accepted: 11/16/2017] [Indexed: 11/13/2022] Open
Abstract
Previous work has identified divergent mechanisms by which cervical remodeling is achieved in preterm birth (PTB) induced by hormone withdrawal (mifepristone) or lipopolysaccharide (LPS). Our current study aims to document how collagen architecture is modified to achieve premature cervical remodeling in mice treated with LPS as a model of infection-induced inflammation. Cervices were collected on gestation day (d) 15 from mice with premature cervical ripening induced by LPS and compared to d15 and d18 controls as well as a hormone withdrawal PTB model. Second harmonic generation (SHG) and electron microscopy were utilized for visualization of collagen morphology and ultrastructure. LPS-mediated premature cervical ripening is characterized by unique structural changes in collagen fiber morphology. LPS treatment increased the interfibrillar spacing of collagen fibrils. A preferential disruption of collagen fiber architecture in the subepithelial region compared to midstroma region was evidenced by increased pores lacking collagen signal in SHG images in the LPS-treated mice. Coinciding with this alteration, the infiltration of neutrophils was concentrated in the subepithelial stromal region as compared to midstromal region implicating the potential role of immune cells to extracellular matrix reorganization in inflammation-induced preterm cervical ripening. The current study demonstrates a preferential disorganization of collagen interfibrillar spacing and collagen fiber structure in LPS-mediated ripening.
Collapse
Affiliation(s)
- Shanmugasundaram Nallasamy
- Department of Obstetrics and Gynecology and Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Meredith Akins
- Department of Obstetrics and Gynecology and Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Breanna Tetreault
- Department of Obstetrics and Gynecology and Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kate Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mala Mahendroo
- Department of Obstetrics and Gynecology and Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
91
|
Garton T, Hua Y, Xiang J, Xi G, Keep RF. Challenges for intraventricular hemorrhage research and emerging therapeutic targets. Expert Opin Ther Targets 2017; 21:1111-1122. [PMID: 29067856 PMCID: PMC6097191 DOI: 10.1080/14728222.2017.1397628] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Intraventricular hemorrhage (IVH) affects both premature infants and adults. In both demographics, it has high mortality and morbidity. There is no FDA approved therapy that improves neurological outcome in either population highlighting the need for additional focus on therapeutic targets and treatments emerging from preclinical studies. Areas covered: IVH induces both initial injury linked to the physical effects of the blood (mass effect) and secondary injury linked to the brain response to the hemorrhage. Preclinical studies have identified multiple secondary injury mechanisms following IVH, and particularly the role of blood components (e.g. hemoglobin, iron, thrombin). This review, with an emphasis on pre-clinical IVH research, highlights therapeutic targets and treatments that may be of use in prevention, acute care, or repair of damage. Expert opinion: An IVH is a potentially devastating event. Progress has been made in elucidating injury mechanisms, but this has still to translate to the clinic. Some pathways involved in injury also have beneficial effects (coagulation cascade/inflammation). A greater understanding of the downstream pathways involved in those pathways may allow therapeutic development. Iron chelation (deferoxamine) is in clinical trial for intracerebral hemorrhage and preclinical data suggest it may be a potential treatment for IVH.
Collapse
Affiliation(s)
- Thomas Garton
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Ya Hua
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Jianming Xiang
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Guohua Xi
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Richard F Keep
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
92
|
Anderson C, Smitherman AB, Engel SM, Nichols HB. Modifiable and non-modifiable risk factors for preterm delivery among adolescent and young adult cancer survivors. Cancer Causes Control 2017; 29:289-295. [PMID: 29196836 DOI: 10.1007/s10552-017-0992-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/28/2017] [Indexed: 11/27/2022]
Abstract
PURPOSE A cancer diagnosis in adolescence and young adulthood (AYA, ages 15-39) may affect future pregnancy outcomes. Previous studies have reported an increased risk of preterm delivery (< 37 weeks of gestation) after maternal cancer treatment. In this analysis, we evaluated whether non-cancer characteristics modify the association between an AYA cancer history and preterm birth. METHODS North Carolina Central Cancer Registry records (2000-2013) were linked to state birth certificate files (2000-2014) to identify births to AYA cancer survivors (n = 1,980). A comparison cohort of births to women without a cancer diagnosis was selected from birth certificate files (n = 11,860). Log-binomial regression was used to estimate risk ratios (RR) and 95% confidence intervals (CI) for preterm delivery. Effect modification by early prenatal care (1st trimester; yes/no), race/ethnicity (white/black/other), previous live births (0/1+), maternal age (< 25/25-29/30-34/35+), smoking during pregnancy (any/none), and education (high school or less/some college/Bachelor's degree or higher) was evaluated using likelihood ratio tests (LRT). RESULTS Overall, preterm births were more common among AYA survivors than the comparison group (RR = 1.24, CI 1.07-1.43). The association was stronger among those who did not receive early prenatal care (RR = 1.73, CI 1.26-2.37) than among those who did (RR = 1.15, CI 0.98-1.35; LRT p = 0.03). Maternal age < 25 was also associated with a greater increase in preterm birth (< 25: RR = 1.80, CI 1.27-2.54; LRT p = 0.07). Associations did not vary strongly by other factors evaluated. CONCLUSIONS An AYA cancer diagnosis may be associated with an increased risk of preterm birth, particularly among women who are younger and receive late or no prenatal care.
Collapse
Affiliation(s)
- Chelsea Anderson
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC, 27599, USA.
| | - Andrew B Smitherman
- Division of Pediatric Hematology & Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Stephanie M Engel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Hazel B Nichols
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| |
Collapse
|
93
|
Cervical HSV-2 infection causes cervical remodeling and increases risk for ascending infection and preterm birth. PLoS One 2017; 12:e0188645. [PMID: 29190738 PMCID: PMC5708831 DOI: 10.1371/journal.pone.0188645] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/10/2017] [Indexed: 11/20/2022] Open
Abstract
Preterm birth (PTB), or birth before 37 weeks gestation, is the leading cause of neonatal mortality worldwide. Cervical viral infections have been established as risk factors for PTB in women, although the mechanism leading to increased risk is unknown. Using a mouse model of pregnancy, we determined that intra-vaginal HSV2 infection caused increased rates of preterm birth following an intra-vaginal bacterial infection. HSV2 infection resulted in histological changes in the cervix mimicking cervical ripening, including significant collagen remodeling and increased hyaluronic acid synthesis. Viral infection also caused aberrant expression of estrogen and progesterone receptor in the cervical epithelium. Further analysis using human ectocervical cells demonstrated a role for Src kinase in virus-mediated changes in estrogen receptor and hyaluronic acid expression. In conclusion, HSV2 affects proteins involved in tissue hormone responsiveness, causes significant changes reminiscent of premature cervical ripening, and increases risk of preterm birth. Studies such as this improve our chances of identifying clinical interventions in the future.
Collapse
|
94
|
Braveman P, Heck K, Egerter S, Dominguez TP, Rinki C, Marchi KS, Curtis M. Worry about racial discrimination: A missing piece of the puzzle of Black-White disparities in preterm birth? PLoS One 2017; 12:e0186151. [PMID: 29020025 PMCID: PMC5636124 DOI: 10.1371/journal.pone.0186151] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 09/26/2017] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES The causes of the large and persistent Black-White disparity in preterm birth (PTB) are unknown. It is biologically plausible that chronic stress across a woman's life course could be a contributor. Prior research suggests that chronic worry about experiencing racial discrimination could affect PTB through neuroendocrine, vascular, or immune mechanisms involved in both responses to stress and the initiation of labor. This study aimed to examine the role of chronic worry about racial discrimination in Black-White disparities in PTB. METHODS The data source was cross-sectional California statewide-representative surveys of 2,201 Black and 8,122 White, non-Latino, U.S.-born postpartum women with singleton live births during 2011-2014. Chronic worry about racial discrimination (chronic worry) was defined as responses of "very often" or "somewhat often" (vs. "not very often" or "never") to the question: "Overall during your life until now, how often have you worried that you might be treated or viewed unfairly because of your race or ethnic group?" Prevalence ratios (PRs) with 95% Confidence Intervals (CI) were calculated from sequential logistic regression models, before and after adjustment for multiple social/demographic, behavioral, and medical factors, to estimate the magnitude of: (a) PTB risks associated with chronic worry among Black women and among White women; and (b) Black-White disparities in PTB, before and after adjustment for chronic worry. RESULTS Among Black and White women respectively, 36.9 (95% CI 32.9-40.9) % and 5.5 (95% CI 4.5-6.5) % reported chronic worry about racial discrimination; rates were highest among Black women of higher income and education levels. Chronic worry was significantly associated with PTB among Black women before (PR 1.73, 95% CI 1.12-2.67) and after (PR 2.00, 95% CI 1.33-3.01) adjustment for covariates. The unadjusted Black-White disparity in PTB (PR 1.59, 95%CI 1.21-2.09) appeared attenuated and became non-significant after adjustment for chronic worry (PR 1.30, 95% CI 0.93-1.81); it appeared further attenuated after adding the covariates (PR 1.17, 95% CI 0.85-1.63). CONCLUSIONS Chronic worry about racial discrimination may play an important role in Black-White disparities in PTB and may help explain the puzzling and repeatedly observed greater PTB disparities among more socioeconomically-advantaged women. Although the single measure of experiences of racial discrimination used in this study precluded examination of the role of other experiences of racial discrimination, such as overt incidents, it is likely that our findings reflect an association between one or more experiences of racial discrimination and PTB. Further research should examine a range of experiences of racial discrimination, including not only chronic worry but other psychological and emotional states and both subtle and overt incidents as well. These dramatic results from a large statewide-representative study add to a growing-but not widely known-literature linking racism-related stress with physical health in general, and shed light on the links between racism-related stress and PTB specifically. Without being causally definitive, this study's findings should stimulate further research and heighten awareness of the potential role of unmeasured social variables, such as diverse experiences of racial discrimination, in racial disparities in health.
Collapse
Affiliation(s)
- Paula Braveman
- Department of Family and Community Medicine and Center on Social Disparities in Health, University of California, San Francisco, CA, United States of America
| | - Katherine Heck
- Department of Family and Community Medicine and Center on Social Disparities in Health, University of California, San Francisco, CA, United States of America
| | - Susan Egerter
- Department of Family and Community Medicine and Center on Social Disparities in Health, University of California, San Francisco, CA, United States of America
| | - Tyan Parker Dominguez
- School of Social Work, University of Southern California, Los Angeles, CA, United States of America
| | - Christine Rinki
- California Department of Public Health, Maternal Child, and Adolescent Health Division, Sacramento, CA, United States of America
| | - Kristen S. Marchi
- Department of Family and Community Medicine and Center on Social Disparities in Health, University of California, San Francisco, CA, United States of America
| | - Michael Curtis
- California Department of Public Health, Maternal Child, and Adolescent Health Division, Sacramento, CA, United States of America
| |
Collapse
|
95
|
Abstract
Senescence contributes to the local and systemic aging of tissues and has been associated with age-related diseases. Recently, roles for this process during pregnancy have come to light, the dysregulation of which has been associated with adverse pregnancy outcomes such as preterm birth. Here, we summarize recent advances that support a role for senescence in birth timing and propose new aspects of study in this emerging field.
Collapse
Affiliation(s)
- Jeeyeon M Cha
- a Division of Diabetes, Endocrinology and Metabolism , Vanderbilt University Medical Center , Nashville , TN , USA.,b Department of Medicine , Vanderbilt University Medical Center , Nashville , TN , USA
| | - David M Aronoff
- b Department of Medicine , Vanderbilt University Medical Center , Nashville , TN , USA.,c Division of Infectious Diseases , Vanderbilt University Medical Center , Nashville , TN , USA.,d Department of Pathology, Microbiology and Immunology , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
96
|
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
|
97
|
Hong X, Hao K, Ji H, Peng S, Sherwood B, Di Narzo A, Tsai HJ, Liu X, Burd I, Wang G, Ji Y, Caruso D, Mao G, Bartell TR, Zhang Z, Pearson C, Heffner L, Cerda S, Beaty TH, Fallin MD, Lee-Parritz A, Zuckerman B, Weeks DE, Wang X. Genome-wide approach identifies a novel gene-maternal pre-pregnancy BMI interaction on preterm birth. Nat Commun 2017; 8:15608. [PMID: 28598419 PMCID: PMC5472707 DOI: 10.1038/ncomms15608] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
Preterm birth (PTB) contributes significantly to infant mortality and morbidity with lifelong impact. Few robust genetic factors of PTB have been identified. Such 'missing heritability' may be partly due to gene × environment interactions (G × E), which is largely unexplored. Here we conduct genome-wide G × E analyses of PTB in 1,733 African-American women (698 mothers of PTB; 1,035 of term birth) from the Boston Birth Cohort. We show that maternal COL24A1 variants have a significant genome-wide interaction with maternal pre-pregnancy overweight/obesity on PTB risk, with rs11161721 (PG × E=1.8 × 10-8; empirical PG × E=1.2 × 10-8) as the top hit. This interaction is replicated in African-American mothers (PG × E=0.01) from an independent cohort and in meta-analysis (PG × E=3.6 × 10-9), but is not replicated in Caucasians. In adipose tissue, rs11161721 is significantly associated with altered COL24A1 expression. Our findings may provide new insight into the aetiology of PTB and improve our ability to predict and prevent PTB.
Collapse
Affiliation(s)
- Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland 21205, USA
| | - Shouneng Peng
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Ben Sherwood
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland 21205, USA
| | - Antonio Di Narzo
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Hui-Ju Tsai
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Xin Liu
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Yuelong Ji
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Deanna Caruso
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Guangyun Mao
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Tami R. Bartell
- Mary Ann & J. Milburn Smith Child Health Research Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois 60611, USA
| | - Zhongyang Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Colleen Pearson
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts 02118, USA
| | - Linda Heffner
- Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Sandra Cerda
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Terri H. Beaty
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - M. Daniele Fallin
- Department of Mental Health, Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Aviva Lee-Parritz
- Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Barry Zuckerman
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts 02118, USA
| | - Daniel E. Weeks
- Department of Human Genetics and Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
- Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
98
|
He JR, Lai YM, Liu HH, Liu GJ, Li WD, Fan XJ, Wei XL, Xia XY, Kuang YS, Liu XD, Chen NN, Lu JH, Chen QZ, Mai WB, Xia HM, Qiu X. Maternal IGF1 and IGF1R polymorphisms and the risk of spontaneous preterm birth. J Clin Lab Anal 2017; 31. [PMID: 28213921 DOI: 10.1002/jcla.22125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/30/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The insulin-like growth factor (IGF) pathway was involved in the occurrence of spontaneous preterm birth (SPTB), but little is known regarding the relationship between genetic variations in IGF pathway and the risk of SPTB. We aimed to investigate the associations of IGF1 rs972936 and IGF1 receptor (IGF1R) rs2229765 polymorphisms with SPTB risk in a Chinese population. METHOD A total of 114 cases of SPTB and 250 controls of term delivery were included from Guangzhou Women and Children's Medical Center, China. The odds ratios (ORs) and the corresponding 95% confidence intervals (CIs) were calculated using multivariate logistic regression. RESULTS We found that the GA and GA/AA genotypes of IGF1 rs972936 were associated with an increased risk of SPTB, and the adjusted ORs (95% CI) were 1.74 (1.01-3.02) and 1.75 (1.04-2.93) respectively. Women carrying GA and GA/AA genotypes of IGF1R rs2229765 had a reduced risk compared to those with the GG genotype (0.60 [0.37-0.98] and 0.64 [0.40-1.00] respectively). There were significant interactions between IGF1 rs972936 and GDM status (P for interaction=.02), as well as between IGF1R rs2229765 and pre-pregnancy BMI (P for interaction <.001) on the risk of SPTB. CONCLUSION Our findings suggest that polymorphisms of IGF1 rs972936 and IGF1R rs2229765 were associated with the risk of SPTB in Chinese pregnant women and these effects depend on the maternal metabolic status.
Collapse
Affiliation(s)
- Jian-Rong He
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yu-Mian Lai
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hui-Hui Liu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Guang-Jian Liu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei-Dong Li
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xue-Jiao Fan
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xue-Ling Wei
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Yan Xia
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ya-Shu Kuang
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Dan Liu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Nian-Nian Chen
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jin-Hua Lu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiao-Zhu Chen
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei-Bi Mai
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hui-Min Xia
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiu Qiu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
99
|
Canovas S, Ivanova E, Romar R, García-Martínez S, Soriano-Úbeda C, García-Vázquez FA, Saadeh H, Andrews S, Kelsey G, Coy P. DNA methylation and gene expression changes derived from assisted reproductive technologies can be decreased by reproductive fluids. eLife 2017; 6. [PMID: 28134613 PMCID: PMC5340525 DOI: 10.7554/elife.23670] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 01/28/2017] [Indexed: 12/14/2022] Open
Abstract
The number of children born since the origin of Assisted Reproductive Technologies (ART) exceeds 5 million. The majority seem healthy, but a higher frequency of defects has been reported among ART-conceived infants, suggesting an epigenetic cost. We report the first whole-genome DNA methylation datasets from single pig blastocysts showing differences between in vivo and in vitro produced embryos. Blastocysts were produced in vitro either without (C-IVF) or in the presence of natural reproductive fluids (Natur-IVF). Natur-IVF embryos were of higher quality than C-IVF in terms of cell number and hatching ability. RNA-Seq and DNA methylation analyses showed that Natur-IVF embryos have expression and methylation patterns closer to in vivo blastocysts. Genes involved in reprogramming, imprinting and development were affected by culture, with fewer aberrations in Natur-IVF embryos. Methylation analysis detected methylated changes in C-IVF, but not in Natur-IVF, at genes whose methylation could be critical, such as IGF2R and NNAT. DOI:http://dx.doi.org/10.7554/eLife.23670.001 Infertility has become more common in many countries, particularly those where many people delay having children until later in life. To help individuals experiencing infertility conceive a child, scientists have developed treatments called assisted reproductive technologies (or ARTs for short). So far, more than 5 million children have been born with the help of these treatments. Most of the children seem healthy; however, birth defects are more common in ART-conceived babies than those conceived without treatment. The cause of these birth defects is not known, though scientists suspect it may have something to do with techniques used in ART. One possible culprit is the liquid that is used in the laboratory to help the parents’ sperm and egg come together for fertilization. This same liquid is also used to bathe the developing embryo for the first few days after fertilization before it is implanted into its mother’s womb. Some scientists wonder whether adding the fluids normally found in the reproductive tract of their mother to this liquid could reduce defects in children conceived via ART. Now, Canovas et al. have shown that fertilizing and growing pig embryos in liquids supplemented with fluid from the wombs of female pigs results in embryos that are closer to naturally conceived pig embryos than in non-supplemented liquids. In the experiments, naturally conceived embryos were compared to ART embryos exposed to the usual liquids and with ART embryos grown in liquids with fluid collected from the pig’s reproductive tract added. Cutting edge technologies were used to sequence the entire genomes of all of the embryos and compare which genes were active in each case. Canovas et al. also looked at chemical markers on the DNA – called epigenetic changes – that turn on or off the expression of genes without changing the DNA code itself. The analysis showed that ART-conceived embryos grown in the usual liquid had different patterns of gene expression and epigenetic changes compared to naturally conceived embryos. Gene expression and epigenetic changes in the ART embryos grown with the pig reproductive fluid was more similar to the naturally conceived embryos. These findings suggest that abnormal gene expression in the ART-liquid exposed embryos may lead to birth defects, and that using natural reproductive fluids may be safer. To confirm this, scientists will have to implant embryos conceived in these three different conditions into mother pigs and assess the health and gene expression patterns of the resulting piglets. If successful, these new insights might one day lead to improvements in ART techniques used to treat infertility in people. DOI:http://dx.doi.org/10.7554/eLife.23670.002
Collapse
Affiliation(s)
- Sebastian Canovas
- Physiology of Reproduction Group, Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia-Campus Mare Nostrum, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Elena Ivanova
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Raquel Romar
- Physiology of Reproduction Group, Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia-Campus Mare Nostrum, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Soledad García-Martínez
- Physiology of Reproduction Group, Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia-Campus Mare Nostrum, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Cristina Soriano-Úbeda
- Physiology of Reproduction Group, Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia-Campus Mare Nostrum, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Francisco A García-Vázquez
- Physiology of Reproduction Group, Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia-Campus Mare Nostrum, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Heba Saadeh
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom.,Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom.,Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Pilar Coy
- Physiology of Reproduction Group, Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia-Campus Mare Nostrum, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| |
Collapse
|
100
|
Marcellin L, Schmitz T, Messaoudene M, Chader D, Parizot C, Jacques S, Delaire J, Gogusev J, Schmitt A, Lesaffre C, Breuiller-Fouché M, Caignard A, Vaiman D, Goffinet F, Cabrol D, Gorochov G, Méhats C. Immune Modifications in Fetal Membranes Overlying the Cervix Precede Parturition in Humans. THE JOURNAL OF IMMUNOLOGY 2016; 198:1345-1356. [PMID: 28031337 DOI: 10.4049/jimmunol.1601482] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/22/2016] [Indexed: 01/03/2023]
Abstract
In humans, parturition is currently viewed as an intrauterine outbreak of inflammation, accompanied by a massive release of proinflammatory cytokines at the maternal-fetal interface that comprises the maternal decidua, placenta, and fetal membranes. At term, fetal membranes overlying the cervix, the future site of rupture, show altered morphology and are termed the zone of altered morphology (ZAM). These alterations occur in normal fetal membranes during late pregnancy, in preparation for labor. In this study, transcriptome, flow cytometry, electron microscopy, and immunohistochemistry analyses collectively highlight a local shift in gene expression and lymphocyte activation in the ZAM. Just before labor, we show that highly polymorphic HLA-A, -B, and -C determinants of fetal origin are selectively exposed in the ZAM to the maternal immune system. A graft rejection-like program occurs in the ZAM, which involves 1) the activation of cytotoxic decidual NK cells, and 2) the decline of decidual immunotolerant M2-like macrophages. Comparison with a prior cohort of fetal membranes shows that acute inflammation only takes place after these first steps of immune modifications. Our results therefore strongly argue in favor of local immune remodeling at the onset of parturition.
Collapse
Affiliation(s)
- Louis Marcellin
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Service de Gynécologie Obstétrique I, Maternité Port Royal, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France.,Service de Gynécologie Obstétrique II et Médecine de la Reproduction, Maternité Port Royal, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Thomas Schmitz
- Service de Gynécologie Obstétrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France.,Faculté de Médecine, Université Paris Diderot, 75013 Paris, France.,INSERM, U1141, 75013 Paris, France.,Département Hospitalo-Universitaire Protect, Hôpital Robert Debré, 75019 Paris, France
| | - Meriem Messaoudene
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France
| | - Driss Chader
- INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses, 75013 Paris, France
| | - Christophe Parizot
- INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses, 75013 Paris, France.,Département d'Immunologie, Groupement Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France; and
| | - Sébastien Jacques
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France
| | - Jérémy Delaire
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France
| | - Jean Gogusev
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France
| | - Alain Schmitt
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France
| | - Corinne Lesaffre
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France
| | - Michelle Breuiller-Fouché
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France
| | - Anne Caignard
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France
| | - Daniel Vaiman
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France
| | - François Goffinet
- Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Service de Gynécologie Obstétrique I, Maternité Port Royal, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France
| | - Dominique Cabrol
- Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Service de Gynécologie Obstétrique I, Maternité Port Royal, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France
| | - Guy Gorochov
- INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses, 75013 Paris, France.,Département d'Immunologie, Groupement Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France; and.,Centre d'Immunologie et des Maladies Infectieuses, Université Pierre et Marie Curie Université Paris 06, CR7, Sorbonne Universités, 75013 Paris, France
| | - Céline Méhats
- INSERM, U1016, Institut Cochin, 75014 Paris, France; .,CNRS, UMR8104, 75014 Paris, France.,Faculté de Médecine, Université Paris Descartes, 75015 Paris, France.,Département Hospitalo-Universitaire Risques et Grossesse, Maternité Port Royal, 75014 Paris, France
| |
Collapse
|