1
|
Pasieczna M, Kuran-Ohde J, Grzyb A, Bokiniec R, Wójcik-Sęp A, Czajkowski K, Szymkiewicz-Dangel J. Value of fetal echocardiographic examination in pregnancies complicated by preterm premature rupture of membranes. J Perinat Med 2024; 52:538-545. [PMID: 38639637 DOI: 10.1515/jpm-2023-0448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/10/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVES Cardiopulmonary and infectious complications are more common in preterm newborns after preterm premature rupture of membranes (pPROM). Fetal echocardiography may be helpful in predicting neonatal condition. Our aim was to assess the cardiovascular changes in fetuses from pregnancies complicated by pPROM and possible utility in predicting the intrauterine or neonatal infection, and neonatal heart failure (HF). METHODS It was a prospective study enrolling 46 women with singleton pregnancies complicated by pPROM between 18+0 and 33+6 weeks of gestation and followed until delivery. 46 women with uncomplicated pregnancies served as a control group. Fetal echocardiographic examinations with the assessment of cardiac structure and function (including pulmonary circulation) were performed in all patients. RESULTS Mean gestational age of pPROM patients was 26 weeks. Parameters suggesting impaired cardiac function in fetuses from pPROM were: higher right ventricle Tei index (0.48 vs. 0.42 p<0.001), lower blood flow velocity in Ao z-score (0.14 vs. 0.84 p=0.005), lower cardiovascular profile score (CVPS), higher rate of tricuspid regurgitation (18.2 % vs. 4.4 % p=0.04) and pericardial effusion (32.6 vs. 0 %). Intrauterine infection was diagnosed in 18 patients (39 %). 4 (8.7 %) newborns met the criteria of early onset sepsis (EOS). HF was diagnosed in 9 newborns. In fetal echocardiographic examination HF group had shorter mitral valve inflow time and higher left ventricle Tei index (0.58 vs. 0.49 p=0.007). CONCLUSIONS Worse cardiac function was observed in fetuses from pPROM compared to fetuses from uncomplicated pregnancies.
Collapse
Affiliation(s)
- Monika Pasieczna
- 2nd Department of Obstetrics and Gynecology, 37803 Medical University of Warsaw , Warsaw, Poland
| | - Joanna Kuran-Ohde
- 2nd Department of Obstetrics and Gynecology, 37803 Medical University of Warsaw , Warsaw, Poland
- Department of Perinatal Cardiology and Congenital Defects, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Grzyb
- Department of Perinatal Cardiology and Congenital Defects, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Cardiology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Renata Bokiniec
- Department of Neonatology and Neonatal Intensive Care, 37803 Medical University of Warsaw , Warsaw, Poland
| | - Agata Wójcik-Sęp
- Department of Neonatology and Neonatal Intensive Care, 37803 Medical University of Warsaw , Warsaw, Poland
| | - Krzysztof Czajkowski
- 2nd Department of Obstetrics and Gynecology, 37803 Medical University of Warsaw , Warsaw, Poland
| | - Joanna Szymkiewicz-Dangel
- Department of Perinatal Cardiology and Congenital Defects, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
2
|
Murillo C, Rueda C, Larroya M, Boada D, Grau L, Ponce J, Herranz A, Gómez O, Ferrero S, Andreu-Fernández V, Gratacós E, Crispi F, Palacio M, Cobo T. Intra-amniotic infection and/or inflammation is associated with fetal cardiac concentric hypertrophy and diastolic dysfunction in preterm labor and preterm prelabor rupture of membranes. Am J Obstet Gynecol 2024; 230:665.e1-665.e30. [PMID: 38290925 DOI: 10.1016/j.ajog.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Preterm delivery is associated with cardiovascular remodeling and dysfunction in children and adults. However, it is unknown whether these effects are caused by the neonatal consequences of preterm birth or if these are already present in utero. OBJECTIVE We evaluated fetal cardiac morphology and function in fetuses of mothers admitted for preterm labor or preterm prelabor rupture of membranes and the association of these changes with the presence of intra-amniotic infection and/or inflammation. STUDY DESIGN In this prospective cohort study, fetal echocardiography and amniocentesis were performed at admission in singleton pregnant women with preterm labor and/or preterm prelabor rupture of membranes between 24.0 and 34.0 weeks' gestation with (intra-amniotic infection and/or inflammation group, n=41) and without intra-amniotic infection and/or inflammation (non-intra-amniotic infection and/or inflammation, n=54). Controls (n=48) were outpatient pregnant women without preterm labor or preterm prelabor rupture of membranes. Intra-amniotic infection was defined by a positive amniotic fluid culture or positive 16S ribosomal RNA gene. Intra-amniotic inflammation was defined by using the amniotic fluid interleukin-6 cutoff levels previously reported by our group being >1.43 ng/mL in preterm prelabor rupture of membranes and >13.4 ng/mL in preterm labor. Fetal cardiac morphology and function was evaluated using echocardiography, and troponin-I and N-terminal pro-brain natriuretic peptide concentrations were measured in amniotic fluid from women with preterm labor or preterm prelabor rupture of membranes and compared with 20 amniotic fluid Biobank samples obtained for reasons other than preterm labor or preterm prelabor rupture of membranes or cardiac pathology. The data were adjusted for the estimated fetal weight below the 10th percentile and for preterm prelabor rupture of membranes at admission and also for gestational age at amniocentesis when amniotic fluid biomarkers were compared. RESULTS From 2018 to 2021, 143 fetuses were included; 95 fetuses were from mothers admitted with a diagnosis of preterm labor or preterm prelabor rupture of membranes, and among those, 41 (28.7%) were in the intra-amniotic infection and/or inflammation group and 54 (37.8%) were in the non-intra-amniotic infection and/or inflammation group. A total of 48 (33.6%) fetuses were included in the control group. Fetuses with preterm labor and/or preterm prelabor rupture of membranes had signs of subclinical cardiac concentric hypertrophy (median left wall thickness of 0.93 [interquartile range, 0.72-1.16] in the intra-amniotic infection and/or inflammation group; 0.79 [0.66-0.92] in the non-intra-amniotic infection and/or inflammation group; and 0.69 [0.56-0.83] in controls; P<.001) and diastolic dysfunction (tricuspid A duration 0.23 seconds [0.21-0.25], 0.24 [0.22-0.25], and 0.21 [0.2-0.23]; P=.007). Systolic function was similar among groups. Higher values of amniotic fluid troponin I (1413 pg/mL [927-2334], 1190 [829-1636], and 841 [671-959]; P<.001) and N-terminal pro-brain natriuretic peptide were detected (35.0%, 17%, and 0%; P=.005) in fetuses with preterm labor or preterm prelabor rupture of membranes when compared with the control group. The highest N-terminal pro-brain natriuretic peptide concentrations were found in the intra-amniotic infection and/or inflammation group. CONCLUSION Fetuses with preterm labor or preterm prelabor rupture of membranes showed signs of cardiac remodeling and subclinical dysfunction, which were more pronounced in those exposed to intra-amniotic infection and/or inflammation. These findings support that the cardiovascular effects observed in children and adults born preterm have, at least in part, a prenatal origin.
Collapse
Affiliation(s)
- Clara Murillo
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain
| | - Claudia Rueda
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain
| | - Marta Larroya
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain
| | - David Boada
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain
| | - Laia Grau
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain
| | - Júlia Ponce
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain
| | - Ana Herranz
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain
| | - Olga Gómez
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBER-ER), Instituto Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Silvia Ferrero
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Biosanitary Research Institute, Valencian International University (VIU), Valencia, Spain. Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain
| | - Eduard Gratacós
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBER-ER), Instituto Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Fàtima Crispi
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBER-ER), Instituto Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Montse Palacio
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBER-ER), Instituto Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Teresa Cobo
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, Institut Clínic de Ginecología, Obstetrícia I Neonatología, Fetal i+D Fetal Medicine Research Center, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer (IIS-FRCB-IDIBAPS), Universitat de Barcelona. Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBER-ER), Instituto Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain.
| |
Collapse
|
3
|
Duignan SM, Lakshminrusimha S, Armstrong K, de Boode WP, El-Khuffash A, Franklin O, Molloy EJ. Neonatal sepsis and cardiovascular dysfunction I: mechanisms and pathophysiology. Pediatr Res 2024; 95:1207-1216. [PMID: 38044334 DOI: 10.1038/s41390-023-02926-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 12/05/2023]
Abstract
The highest incidence of sepsis across all age groups occurs in neonates leading to substantial mortality and morbidity. Cardiovascular dysfunction frequently complicates neonatal sepsis including biventricular systolic and/or diastolic dysfunction, vasoregulatory failure, and pulmonary arterial hypertension. The haemodynamic response in neonatal sepsis can be hyperdynamic or hypodynamic and the underlying pathophysiological mechanisms are heterogeneous. The diagnosis and definition of both neonatal sepsis and cardiovascular dysfunction complicating neonatal sepsis are challenging and not consensus-based. Future developments in neonatal sepsis management will be facilitated by common definitions and datasets especially in neonatal cardiovascular optimisation. IMPACT: Cardiovascular dysfunction is common in neonatal sepsis but there is no consensus-based definition, making calculating the incidence and designing clinical trials challenging. Neonatal cardiovascular dysfunction is related to the inflammatory response, which can directly target myocyte function and systemic haemodynamics.
Collapse
Affiliation(s)
- Sophie M Duignan
- Department of Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | | | - Kathryn Armstrong
- Children's Heart Centre, BC Children's Hospital, Vancouver, BC, Canada
| | - Willem P de Boode
- Department of Neonatology, Radboud University Medical Center, Radboud Institute for Health Sciences, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Afif El-Khuffash
- School of Medicine, Department of Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Orla Franklin
- Department of Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
- Discipline of Paediatrics, Trinity College, The University of Dublin, Trinity Research in Childhood (TRiCC) & Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College, The University of Dublin, Trinity Research in Childhood (TRiCC) & Trinity Translational Medicine Institute (TTMI), Dublin, Ireland.
- Department of Neonatology, Children's Health Ireland at Crumlin, Dublin, Ireland.
- Department of Neonatology, Coombe Women and Infants University Hospital, Dublin, Ireland.
- Paediatric Neurodisability, Children's Health Ireland at Tallaght, Dublin, Ireland.
| |
Collapse
|
4
|
Yildirim M, Oluklu D, Menekse Beser D, Uyan Hendem D, Tanacan A, Sahin D. The Importance of Tissue Doppler Imaging and M-Mode Ultrasonography in Fetuses With Preterm Premature Rupture of Membranes: A Case-Control Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:85-94. [PMID: 37772464 DOI: 10.1002/jum.16340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/29/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023]
Abstract
OBJECTIVE To compare the cardiac functions of fetuses with preterm premature rupture of membranes (PPROM) between their control groups and investigate its relationship with perinatal outcomes. METHODS This prospective study was conducted with 102 pregnant women. Pregnant women with PPROM were divided into two subgroups Group A, between 26 and 30 weeks, and Group B, between 30 and 34 weeks. A control group was formed by randomly including one healthy pregnant woman for each study patient. Sociodemographic, obstetric data, tissue Doppler imaging, and M-mode imaging results were compared. The relationship between echocardiographic parameters and perinatal outcomes was also investigated. RESULTS Tricuspid annular plane systolic excursion (TAPSE), S', and ET' of systolic cardiac parameters were shortened in both groups compared with their controls. Diastolic function indicator E'/A', and global function indicator myocardial performance index' increased in both groups. Isovolumetric contraction time' did not change between groups. A correlation was found between myocardial performance index', and the length of neonatal intensive care unit stay in Group A and TAPSE and duration of respiratory support and length of neonatal intensive care unit stay in Group B. CONCLUSIONS The fetal cardiac function seems to be affected by PPROM, and these changes are associated with neonatal outcomes. Therefore, administering fetal cardiac function evaluation in pregnancies complicated by PPROM may help physicians establish more appropriate clinical management protocols in this special population.
Collapse
Affiliation(s)
- Muradiye Yildirim
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health, Ankara City Hospital, Ankara, Turkey
| | - Deniz Oluklu
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health, Ankara City Hospital, Ankara, Turkey
| | - Dilek Menekse Beser
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health, Ankara City Hospital, Ankara, Turkey
| | - Derya Uyan Hendem
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health, Ankara City Hospital, Ankara, Turkey
| | - Atakan Tanacan
- Department of Obstetrics and Gynecology, Division of Perinatology, University of Health Sciences, Turkish Ministry of Health, Ankara City Hospital, Ankara, Turkey
| | - Dilek Sahin
- Department of Obstetrics and Gynecology, Division of Perinatology, University of Health Sciences, Turkish Ministry of Health, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
5
|
Yildirim M, Oluklu D, Beser DM, Hendem DU, Aktas BA, Yildiz EG, Kara O, Sahin D. The assessment of fetal cardiac functions in pregnancies with autoimmune diseases: a prospective case-control study. J Perinat Med 2023; 51:1074-1081. [PMID: 37531189 DOI: 10.1515/jpm-2023-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/24/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVES This study aimed to assess the effect of the inflammatory process on fetal cardiac functions in pregnant women with autoimmune diseases (AID). METHODS This prospective study included 36 pregnant women with diagnosed AID. Nineteen systemic lupus erythematosus, 12 antiphospholipid syndrome, 5 Sjögren's syndrome, and 72 healthy pregnancies were included. Fetal cardiac functions were evaluated with pulsed-wave, tissue Doppler, and M-mode echocardiography. RESULTS Sociodemographic data were similar in both groups. Significant increases were found in tricuspid E (43.5 ± 0.9, p<0.001) and A (59.2 ± 2.2, p<0.001) and E/A (0.74 ± 0.03, p<0.001), E'/A' (0.64 ± 0.15, p<0.001), E/E' (6.5 ± 0.6, p<0.001), and left ventricular myocardial performance index (0.54 ± 0.03, p=0.005). We demonstrated a significant decrease in tricuspid E' (6.7 ± 0.6, p<0.001) and S' (6.9 ± 1, p<0.001) and in TAPSE (7.7 ± 0.5, p=0.002). We also found a significantly prolonged PR interval (130 ± 8, p<0.001). There was a significant increase in E' (6.8, p=0.033) and a significant decrease in E/E' ratio (6.4, p=0.027) in the group using hydroxychloroquine (HCQ) compared to non-users. CONCLUSIONS We found that pregnancy with autoimmune diseases affects fetal heart functions. Additionally, hydroxychloroquine may positively affect the heart of AID fetuses. This information might be useful to clinicians in the follow-up of cardiovascular diseases.
Collapse
Affiliation(s)
- Muradiye Yildirim
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Deniz Oluklu
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Dilek Menekse Beser
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Derya Uyan Hendem
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Betul Akgun Aktas
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Esra Gulen Yildiz
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Ozgur Kara
- Department of Obstetrics and Gynecology, Division of Perinatology, University of Health Sciences, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| | - Dilek Sahin
- Department of Obstetrics and Gynecology, Division of Perinatology, University of Health Sciences, Turkish Ministry of Health Ankara City Hospital, Ankara, Türkiye
| |
Collapse
|
6
|
Usuda H, Watanabe S, T H, Saito M, Sato S, Ikeda H, Kumagai Y, Choolani MC, Kemp MW. Artificial placenta technology: History, potential and perception. Placenta 2023; 141:10-17. [PMID: 37743742 DOI: 10.1016/j.placenta.2022.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/20/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
As presently conceptualised, the artificial placenta (AP) is an experimental life support platform for extremely preterm infants (i.e. 400-600 g; 21-23+6 weeks of gestation) born at the border of viability. It is based around the oxygenation of the periviable fetus using gas-exchangers connected to the fetal vasculature. In this system, the lung remains fluid-filled and the fetus remains in a quiescent state. The AP has been in development for some sixty years. Over this time, animal experimental models have evolved iteratively from employing external pump-driven systems used to support comparatively mature fetuses (generally goats or sheep) to platforms driven by the fetal heart and used successfully to maintain extremely premature fetuses weighing around 600 g. Simultaneously, sizable advances in neonatal and obstetric care mean that the nature of a potential candidate patient for this therapy, and thus the threshold success level for justifying its adoption, have both changed markedly since this approach was first conceived. Five landmark breakthroughs have occurred over the developmental history of the AP: i) the first human studies reported in the 1950's; ii) foundation animal studies reported in the 1960's; iii) the first extended use of AP technology combined with fetal pulmonary resuscitation reported in the 1990s; iv) the development of AP systems powered by the fetal heart reported in the 2000's; and v) the adaption of this technology to maintain extremely preterm fetuses (i.e. 500-600 g body weight) reported in the 2010's. Using this framework, the present paper will provide a review of the developmental history of this long-running experimental system and up-to-date assessment of the published field today. With the apparent acceleration of AP technology towards clinical application, there has been an increase in the attention paid to the field, along with some inaccurate commentary regarding its potential application and merits. Additionally, this paper will address several misrepresentations regarding the potential application of AP technology that serve to distract from the significant potential of this approach to greatly improve outcomes for extremely preterm infants born at or close to the present border of viability.
Collapse
Affiliation(s)
- H Usuda
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - S Watanabe
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hanita T
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - M Saito
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - S Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - H Ikeda
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Y Kumagai
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - M C Choolani
- Women and Infants Research Foundation, King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - M W Kemp
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan; School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia; Women and Infants Research Foundation, King Edward Memorial Hospital, Perth, Western Australia, Australia; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
7
|
Zamstein O, Wainstock T, Sheiner E. Intrapartum Maternal Fever and Long-Term Infectious Morbidity of the Offspring. J Clin Med 2023; 12:jcm12093329. [PMID: 37176769 PMCID: PMC10179301 DOI: 10.3390/jcm12093329] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Maternal intrapartum fever can lead to various maternal and neonatal complications and is attributed to various etiologies including infectious and non-infectious processes. In this study, we evaluated whether intrapartum fever affects the offspring's tendency to long-term infectious morbidity. A population-based cohort analysis including deliveries between 1991 and 2021 was conducted. The incidence of hospitalizations of the offspring up to the age of 18 years, due to various infectious conditions, was compared between pregnancies complicated by intrapartum fever and those that were not. A Kaplan-Meier survival curve was used to assess cumulative hospitalization incidence. A Cox proportional hazards model was used to control for confounders. Overall, 538 of the 356,356 included pregnancies were complicated with fever. A higher rate of pediatric hospitalizations due to various infectious conditions was found among the exposed group, which was significant for viral, fungal and ENT infections (p < 0.05 for all). The total number of infectious-related hospitalizations was significantly higher (30.1% vs. 24.1%; OR = 1.36; p = 0.001), as was the cumulative incidence of hospitalizations. This association remained significant after controlling for confounders using a Cox proportional hazards model (adjusted HR = 1.21; 95% CI 1.04-1.41, p = 0.016). To conclude, fever diagnosed close to delivery may influence offspring susceptibility to pediatric infections.
Collapse
Affiliation(s)
- Omri Zamstein
- The Obstetrics and Gynecology Division, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva POB 151, Israel
| | - Tamar Wainstock
- The Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva POB 653, Israel
| | - Eyal Sheiner
- The Obstetrics and Gynecology Division, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva POB 151, Israel
| |
Collapse
|
8
|
Owen JC, Garrick SP, Peterson BM, Berger PJ, Nold MF, Sehgal A, Nold-Petry CA. The role of interleukin-1 in perinatal inflammation and its impact on transitional circulation. Front Pediatr 2023; 11:1130013. [PMID: 36994431 PMCID: PMC10040554 DOI: 10.3389/fped.2023.1130013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/31/2023] Open
Abstract
Preterm birth is defined as delivery at <37 weeks of gestational age (GA) and exposes 15 million infants worldwide to serious early life diseases. Lowering the age of viability to 22 weeks GA entailed provision of intensive care to a greater number of extremely premature infants. Moreover, improved survival, especially at extremes of prematurity, comes with a rising incidence of early life diseases with short- and long-term sequelae. The transition from fetal to neonatal circulation is a substantial and complex physiologic adaptation, which normally happens rapidly and in an orderly sequence. Maternal chorioamnionitis or fetal growth restriction (FGR) are two common causes of preterm birth that are associated with impaired circulatory transition. Among many cytokines contributing to the pathogenesis of chorioamnionitis-related perinatal inflammatory diseases, the potent pro-inflammatory interleukin (IL)-1 has been shown to play a central role. The effects of utero-placental insufficiency-related FGR and in-utero hypoxia may also be mediated, in part, via the inflammatory cascade. In preclinical studies, blocking such inflammation, early and effectively, holds great promise for improving the transition of circulation. In this mini-review, we outline the mechanistic pathways leading to abnormalities in transitional circulation in chorioamnionitis and FGR. In addition, we explore the therapeutic potential of targeting IL-1 and its influence on perinatal transition in the context of chorioamnionitis and FGR.
Collapse
Affiliation(s)
- Josephine C. Owen
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Steven P. Garrick
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Briana M. Peterson
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Philip J. Berger
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Marcel F. Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Claudia A. Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Correspondence: Claudia A. Nold-Petry
| |
Collapse
|
9
|
Motomura K, Romero R, Galaz J, Tao L, Garcia-Flores V, Xu Y, Done B, Arenas-Hernandez M, Miller D, Gutierrez-Contreras P, Farias-Jofre M, Aras S, Grossman LI, Tarca AL, Gomez-Lopez N. Fetal and maternal NLRP3 signaling is required for preterm labor and birth. JCI Insight 2022; 7:158238. [PMID: 35993366 PMCID: PMC9462488 DOI: 10.1172/jci.insight.158238] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Li Tao
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Pedro Gutierrez-Contreras
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Farias-Jofre
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Siddhesh Aras
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Lawrence I. Grossman
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, Michigan, USA, and Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
10
|
Korada S, Jebbia MR, Pavlek LR. Linking the Perinatal Environment to Neonatal Cardiovascular Outcomes. Neoreviews 2022; 23:e400-e408. [PMID: 35641456 DOI: 10.1542/neo.23-6-e400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cases of high-risk pregnancies continue to rise throughout the United States and globally, increasing rates of maternal and neonatal morbidity. Common pregnancy complications and morbidities include preterm birth, hypertensive disorders, fetal growth restriction, diabetes mellitus, and chorioamnionitis. Exposure to these perinatal conditions contributes to cardiac morbidities in the fetus and neonate, including altered cardiac growth, congenital heart disease, and cardiac dysfunction. Significant research has demonstrated lasting effects of these pregnancy complications, with increased rates of cardiac morbidities seen in children and adults after these perinatal exposures. The link between the perinatal environment and long-term outcomes has not been fully elucidated. The aim of this review is to discuss the current understanding of the implications of a high-risk pregnancy on fetal and neonatal cardiac development.
Collapse
Affiliation(s)
- Saichidroopi Korada
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Maria R Jebbia
- Division of Neonatology, Nationwide Children's Hospital, Columbus, OH.,Department of Pediatrics, The Ohio State University, Columbus, OH
| | - Leeann R Pavlek
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH.,Division of Neonatology, Nationwide Children's Hospital, Columbus, OH.,Department of Pediatrics, The Ohio State University, Columbus, OH
| |
Collapse
|
11
|
Oluklu D, Kara O, Turgut E, Goncu Ayhan S, Yildirim M, Sahin D. Evaluation of fetal cardiac morphology and functions in pregnant women with familial Mediterranean fever. Echocardiography 2022; 39:606-611. [PMID: 35279878 DOI: 10.1111/echo.15336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE We aimed to evaluate and compare fetal cardiac morphology and functions of pregnant women with familial Mediterranean fever (FMF) and healthy pregnant women. METHODS The study included 34 pregnant women with FMF and 68 healthy pregnant women matched with maternal age, gravidity, parity, gestational age, and pre-pregnancy body mass index (BMI) in 34th-37th gestational weeks. Fetal echocardiographic evaluation was performed with two-dimensional (2D) imaging, M-mode imaging, pulsed wave (PW) Doppler, and tissue Doppler imaging (TDI). RESULTS Fetal cardiac morphological measures, including cardiothoracic ratio, cardiac axis angle, right and left ventricular area, sphericity index, and ventricular septal thickness was similar in both groups. Compared with the control group, myocardial performance index (MPI), which indicates global myocardial performance, was significantly higher, and ejection time (ET) was significantly shortened in the FMF group. In addition, which shows the diastolic functional parameters such as, tricuspid E wave, E/A, E/E' ratio, and mitral E wave, E/A, E/E' ratio, were significantly higher; tricuspid A and mitral A waves were significantly lower. We found that mitral and tricuspid annular plane systolic excursion (MAPSE and TAPSE) were significantly lower in those with FMF duration over eight years than those with FMF duration less than 8 years. CONCLUSION There is no fetal cardiac morphological change in pregnant women with FMF. However, there may be changes in diastolic function. As the maternal FMF duration increases, systolic functions may also change.
Collapse
Affiliation(s)
- Deniz Oluklu
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Ozgur Kara
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Ezgi Turgut
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Sule Goncu Ayhan
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Muradiye Yildirim
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Dilek Sahin
- Department of Obstetrics and Gynecology, Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
12
|
Yallapragada SG, Savani RC, Goss KN. Cardiovascular impact and sequelae of bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:3453-3463. [PMID: 33756045 DOI: 10.1002/ppul.25370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
The development, growth, and function of the cardiac, pulmonary, and vascular systems are closely intertwined during both fetal and postnatal life. In utero, placental, environmental, and genetic insults may contribute to abnormal pulmonary alveolarization and vascularization that increase susceptibility to the development of bronchopulmonary dysplasia (BPD) in preterm infants. However, the shared milieu of stressors may also contribute to abnormal cardiac or vascular development in the fetus and neonate, leading to the potential for cardiovascular dysfunction. Further, cardiac or pulmonary maladaptation can potentiate dysfunction in the other organ, amplify the risk for BPD in the neonate, and increase the trajectory for overall neonatal morbidity. Beyond infancy, there is an increased risk for systemic and pulmonary vascular disease including hypertension, as well as potential cardiac dysfunction, particularly within the right ventricle. This review will focus on the cardiovascular antecedents of BPD in the fetus, cardiovascular consequences of preterm birth in the neonate including associations with BPD, and cardiovascular impact of prematurity and BPD throughout the lifespan.
Collapse
Affiliation(s)
- Sushmita G Yallapragada
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kara N Goss
- Division of Pulmonary and Critical Care, Departments of Medicine and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
13
|
Yoshikawa K, Kiyoshima C, Hirakawa T, Urushiyama D, Fukagawa S, Izuchi D, Sanui A, Kurakazu M, Miyata K, Nomiyama M, Setoue T, Nagamitsu S, Nabeshima K, Hata K, Yasunaga S, Miyamoto S. Diagnostic predictability of miR-4535 and miR-1915-5p expression in amniotic fluid for foetal morbidity of infection. Placenta 2021; 114:68-75. [PMID: 34479063 DOI: 10.1016/j.placenta.2021.08.059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/26/2021] [Accepted: 08/23/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Clinical prediction of foetal inflammatory response syndrome (FIRS) is highly necessary. We have previously reported that miR-4535 and miR-1915-5p are potential biomarkers for severe chorioamnionitis based on the results of microRNA array analysis. Therefore, we evaluated the relationship between foetal morbidity of infection and miR-4535, miR-1915-5p, interleukin (IL)-6, or 16S rDNA copy number levels in amniotic fluid from pregnant women with chorioamnionitis. METHODS Amniotic fluid from 57 pregnant women with preterm premature membrane rupture or threatened premature labour were collected. Infants with WBC counts <5000/μL or >20,000/μL, CRP >0.5 mg/mL, or IgM >20 mg/mL at birth received a diagnosis of suspicious foetal infection, and those requiring antibiotic administration for >5 days were considered infected newborns. miR-4535, miR-1915-5p, and IL-6 levels and 16S rDNA copy number were evaluated. Mann-Whitney U test and Dunn's test were used for comparison. The area under the curve (AUC) and Youden index were calculated to examine the diagnostic accuracy of foetal morbidity of infection. RESULTS miR-4535, miR-1915-5p, 16S rDNA, and IL-6 were significantly higher in patients with severe chorioamnionitis than in patients with chorionitis or sub-chorionitis (P < 0.05). miR-4535 and miR-1915-5p levels were significantly associated with WBC counts <5000/μL or >20,000/μL, CRP >0.5 mg/mL, or IgM >20 mg/mL (P < 0.05). AUC values of miR-4535 and miR-1915-5p indicated moderate or low accuracy for foetal morbidity of infection, while those of IL-6 and 16S rDNA seemed unreliable. DISCUSSION MiR-4535 and miR-1915-5p levels in amniotic fluid may be considered clinically predictive for foetal morbidity of infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ayako Sanui
- Department of Obstetrics & Gynecology, Japan
| | | | | | - Makoto Nomiyama
- Department of Obstetrics and Gynecology, National Hospital Organization Saga Hospital, Saga, Japan
| | - Takashi Setoue
- Center for Maternal, Foetal & Neonatal Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | | | | | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | | |
Collapse
|
14
|
Matthiesen NB, Østergaard JR, Hjortdal VE, Henriksen TB. Congenital Heart Defects and the Risk of Spontaneous Preterm Birth. J Pediatr 2021; 229:168-174.e5. [PMID: 32980375 DOI: 10.1016/j.jpeds.2020.09.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVES To estimate the association between major types of congenital heart defects (CHD) and spontaneous preterm birth, and to assess the potential underlying mechanisms. STUDY DESIGN This nationwide, registry-based study included a cohort of all singleton pregnancies in Denmark from 1997 to 2013. The association between CHD and spontaneous preterm birth was estimated by multivariable Cox regression, adjusted for potential confounders. The following potential mechanisms were examined: maternal genetics (sibling analyses), polyhydramnios, preterm prelabor rupture of membranes, preeclampsia, and indicators of fetal and placental growth. RESULTS The study included 1 040 474 births. Compared with the general population, CHD was associated with an increased risk of spontaneous preterm birth, adjusted hazard ratio 2.1 (95% CI, 1.9-2.4). Several subtypes were associated with increased risks, including pulmonary stenosis combined with a septal defect, 5.2 (95% CI, 3.7-7.5); pulmonary stenosis or atresia, 3.1 (95% CI, 2.4-4.1); tetralogy of Fallot 2.5 (95% CI, 1.6-3.8); coarctation or interrupted aortic arch 2.2 (95% CI, 1.5-3.2); and hypoplastic left heart syndrome, 2.0 (95% CI, 1.0-4.1). Overall, preterm prelabor rupture of membranes mediated more than one-half of the association. Maternal genetics, polyhydramnios, or indicators of fetal or placental growth did not explain the reported associations. CONCLUSIONS CHD, especially right ventricular outflow tract obstructions, were associated with an increased risk of spontaneous preterm birth. The risk was carried by the CHD and not by maternal genetics. Moreover, preterm prelabor rupture of membranes was identified as a potential underlying mechanism.
Collapse
Affiliation(s)
- Niels B Matthiesen
- Perinatal Epidemiology Research Unit, Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Pediatrics and Adolescent Medicine, Randers Regional Hospital, Randers, Denmark.
| | - John R Østergaard
- Center for Rare Diseases, Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Vibeke E Hjortdal
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Aarhus, Denmark; Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen, Denmark
| | - Tine B Henriksen
- Perinatal Epidemiology Research Unit, Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Kelleher MA, Lee JY, Roberts VHJ, Novak CM, Baschat AA, Morgan TK, Novy MJ, Räsänen JP, Frias AE, Burd I. Maternal azithromycin therapy for Ureaplasma parvum intraamniotic infection improves fetal hemodynamics in a nonhuman primate model. Am J Obstet Gynecol 2020; 223:578.e1-578.e11. [PMID: 32343954 PMCID: PMC7591241 DOI: 10.1016/j.ajog.2020.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ureaplasma parvum infection is a prevalent cause of intrauterine infection associated with preterm birth, preterm premature rupture of membranes, fetal inflammatory response syndrome, and adverse postnatal sequelae. Elucidation of diagnostic and treatment strategies for infection-associated preterm labor may improve perinatal and long-term outcomes for these cases. OBJECTIVE This study assessed the effect of intraamniotic Ureaplasma infection on fetal hemodynamic and cardiac function and the effect of maternal antibiotic treatment on these outcomes. STUDY DESIGN Chronically catheterized pregnant rhesus monkeys were assigned to control (n=6), intraamniotic inoculation with Ureaplasma parvum (107 colony-forming units/mL, n=15), and intraamniotic infection plus azithromycin treatment (12.5 mg/kg twice a day intravenously, n=8) groups. At approximately 135 days' gestation (term=165 days), pulsed and color Doppler ultrasonography was used to obtain measurements of fetal hemodynamics (pulsatility index of umbilical artery, ductus venosus, descending aorta, ductus arteriosus, aortic isthmus, right pulmonary artery, middle cerebral artery and cerebroplacental ratio, and left and right ventricular cardiac outputs) and cardiac function (ratio of peak early vs late transmitral flow velocity [marker of ventricular function], Tei index [myocardial performance index]). These indices were stratified by amniotic fluid proinflammatory mediator levels and cardiac histology. RESULTS Umbilical and fetal pulmonary artery vascular impedances were significantly increased in animals from the intraamniotic inoculation with Ureaplasma parvum group (P<.05). Azithromycin treatment restored values to control levels. Amniotic fluid prostaglandin F2 alpha levels were significantly higher in animals with abnormal umbilical artery pulsatility index (>1.1) than in those with normal blood flow (P<.05; Spearman ρ=0.6, P<.05). In the intraamniotic inoculation with Ureaplasma parvum group, left ventricular cardiac output was significantly decreased (P<.001), and more animals had abnormal right-to-left ventricular cardiac output ratios (defined as >1.6, P<.05). Amniotic fluid interleukin-6 concentrations were elevated in cases of abnormal right-to-left ventricular cardiac output ratios compared with those in normal cases (P<.05). CONCLUSION Fetal hemodynamic alterations were associated with intraamniotic Ureaplasma infection and ameliorated after maternal antibiotic treatment. Doppler ultrasonographic measurements merit continuing investigation as a diagnostic method to identify fetal cardiovascular and hemodynamic compromise associated with intrauterine infection or inflammation and in the evaluation of therapeutic interventions or clinical management of preterm labor.
Collapse
Affiliation(s)
- Meredith A Kelleher
- Division of Reproductive and Development Sciences, Oregon National Primate Research Center, Beaverton, OR.
| | - Ji Yeon Lee
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Victoria H J Roberts
- Division of Reproductive and Development Sciences, Oregon National Primate Research Center, Beaverton, OR
| | - Christopher M Novak
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ahmet A Baschat
- Johns Hopkins Center for Fetal Therapy, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Terry K Morgan
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR
| | - Miles J Novy
- Division of Reproductive and Development Sciences, Oregon National Primate Research Center, Beaverton, OR
| | - Juha P Räsänen
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR; University of Helsinki, Helsinki, Finland
| | - Antonio E Frias
- Division of Reproductive and Development Sciences, Oregon National Primate Research Center, Beaverton, OR; Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
16
|
Jung E, Romero R, Yeo L, Diaz-Primera R, Marin-Concha J, Para R, Lopez AM, Pacora P, Gomez-Lopez N, Yoon BH, Kim CJ, Berry SM, Hsu CD. The fetal inflammatory response syndrome: the origins of a concept, pathophysiology, diagnosis, and obstetrical implications. Semin Fetal Neonatal Med 2020; 25:101146. [PMID: 33164775 PMCID: PMC10580248 DOI: 10.1016/j.siny.2020.101146] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetus can deploy a local or systemic inflammatory response when exposed to microorganisms or, alternatively, to non-infection-related stimuli (e.g., danger signals or alarmins). The term "Fetal Inflammatory Response Syndrome" (FIRS) was coined to describe a condition characterized by evidence of a systemic inflammatory response, frequently a result of the activation of the innate limb of the immune response. FIRS can be diagnosed by an increased concentration of umbilical cord plasma or serum acute phase reactants such as C-reactive protein or cytokines (e.g., interleukin-6). Pathologic evidence of a systemic fetal inflammatory response indicates the presence of funisitis or chorionic vasculitis. FIRS was first described in patients at risk for intraamniotic infection who presented preterm labor with intact membranes or preterm prelabor rupture of the membranes. However, FIRS can also be observed in patients with sterile intra-amniotic inflammation, alloimmunization (e.g., Rh disease), and active autoimmune disorders. Neonates born with FIRS have a higher rate of complications, such as early-onset neonatal sepsis, intraventricular hemorrhage, periventricular leukomalacia, and death, than those born without FIRS. Survivors are at risk for long-term sequelae that may include bronchopulmonary dysplasia, neurodevelopmental disorders, such as cerebral palsy, retinopathy of prematurity, and sensorineuronal hearing loss. Experimental FIRS can be induced by intra-amniotic administration of bacteria, microbial products (such as endotoxin), or inflammatory cytokines (such as interleukin-1), and animal models have provided important insights about the mechanisms responsible for multiple organ involvement and dysfunction. A systemic fetal inflammatory response is thought to be adaptive, but, on occasion, may become dysregulated whereby a fetal cytokine storm ensues and can lead to multiple organ dysfunction and even fetal death if delivery does not occur ("rescued by birth"). Thus, the onset of preterm labor in this context can be considered to have survival value. The evidence so far suggests that FIRS may compound the effects of immaturity and neonatal inflammation, thus increasing the risk of neonatal complications and long-term morbidity. Modulation of a dysregulated fetal inflammatory response by the administration of antimicrobial agents, anti-inflammatory agents, or cell-based therapy holds promise to reduce infant morbidity and mortality.
Collapse
Affiliation(s)
- Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA; Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA.
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Julio Marin-Concha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ashley M Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chong Jai Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
17
|
Antibiotic administration can eradicate intra-amniotic infection or intra-amniotic inflammation in a subset of patients with preterm labor and intact membranes. Am J Obstet Gynecol 2019; 221:142.e1-142.e22. [PMID: 30928566 DOI: 10.1016/j.ajog.2019.03.018] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/13/2019] [Accepted: 03/21/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND Intra-amniotic infection is present in 10% of patients with an episode of preterm labor, and is a risk factor for impending preterm delivery and neonatal morbidity/mortality. Intra-amniotic inflammation is often associated with intra-amniotic infection, but is sometimes present in the absence of detectable microorganisms. Antibiotic treatment of intra-amniotic infection has traditionally been considered to be ineffective. Intra-amniotic inflammation without microorganisms has a prognosis similar to that of intra-amniotic infection. OBJECTIVE To determine whether antibiotics can eradicate intra-amniotic infection or intra-amniotic inflammation in a subset of patients with preterm labor and intact membranes. MATERIALS AND METHODS The study population consisted of women who met the following criteria: 1) singleton gestation between 20 and 34 weeks; 2) preterm labor and intact membranes; 3) transabdominal amniocentesis performed for the evaluation of the microbiologic/inflammatory status of the amniotic cavity; 4) intra-amniotic infection and/or intra-amniotic inflammation; and 5) received antibiotic treatment that consisted of ceftriaxone, clarithromycin, and metronidazole. Follow-up amniocentesis was performed in a subset of patients. Amniotic fluid was cultured for aerobic and anaerobic bacteria and genital mycoplasmas, and polymerase chain reaction was performed for Ureaplasma spp. Intra-amniotic infection was defined as a positive amniotic fluid culture or positive polymerase chain reaction, and intra-amniotic inflammation was suspected when there was an elevated amniotic fluid white blood cell count or a positive result of a rapid test for matrix metalloproteinase-8. For this study, the final diagnosis of intra-amniotic inflammation was made by measuring the interleukin-6 concentration in stored amniotic fluid (>2.6 ng/mL). These results were not available to managing clinicians. Treatment success was defined as eradication of intra-amniotic infection and/or intra-amniotic inflammation or delivery ≥37 weeks. RESULTS Of 62 patients with intra-amniotic infection and/or intra-amniotic inflammation, 50 received the antibiotic regimen. Of those patients, 29 were undelivered for ≥7 days and 19 underwent a follow-up amniocentesis. Microorganisms were identified by culture or polymerase chain reaction of amniotic fluid obtained at admission in 21% of patients (4/19) who had a follow-up amniocentesis, and were eradicated in 3 of the 4 patients. Resolution of intra-amniotic infection/inflammation was confirmed in 79% of patients (15/19), and 1 other patient delivered at term, although resolution of intra-amniotic inflammation could not be confirmed after a follow-up amniocentesis. Thus, resolution of intra-amniotic inflammation/infection or term delivery (treatment success) occurred in 84% of patients (16/19) who had a follow-up amniocentesis. Treatment success occurred in 32% of patients (16/50) with intra-amniotic infection/inflammation who received antibiotics. The median amniocentesis-to-delivery interval was significantly longer among women who received the combination of antibiotics than among those who did not (11.4 days vs 3.1 days: P = .04). CONCLUSION Eradication of intra-amniotic infection/inflammation after treatment with antibiotics was confirmed in 79% of patients with preterm labor, intact membranes, and intra-amniotic infection/inflammation who had a follow-up amniocentesis. Treatment success occurred in 84% of patients who underwent a follow-up amniocentesis and in 32% of women who received the antibiotic regimen.
Collapse
|
18
|
Eloundou SN, Lee J, Wu D, Lei J, Feller MC, Ozen M, Zhu Y, Hwang M, Jia B, Xie H, Clemens JL, McLane MW, AlSaggaf S, Nair N, Wills-Karp M, Wang X, Graham EM, Baschat A, Burd I. Placental malperfusion in response to intrauterine inflammation and its connection to fetal sequelae. PLoS One 2019; 14:e0214951. [PMID: 30943260 PMCID: PMC6447225 DOI: 10.1371/journal.pone.0214951] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/22/2019] [Indexed: 11/29/2022] Open
Abstract
Exposure to intrauterine inflammation (IUI) is associated with short- and long-term adverse perinatal outcomes. However, little data exist on utilizing placenta to prognosticate fetal injury in this scenario. Our study aimed to utilize imaging modalities to evaluate mechanisms contributing to placental injury following IUI exposure and correlated it to concomitant fetal brain injury. CD1 pregnant dams underwent laparotomies and received intrauterine injections of either lipopolysaccharide (LPS; a model of IUI) or phosphate-buffered saline (PBS). In utero ultrasound Doppler velocimetry of uterine and umbilical arteries and magnetic resonance imaging (MRI) of placental volumes with confirmatory immunohistochemical (vimentin) and histochemistry (fibrin) analyses were performed. ELISA for thrombosis markers, fibrinogen and fibrin was performed to analyze thrombi in placenta. Fetal brain immunohistochemistry was performed to detect microglial activation (ionized calcium-binding adaptor molecule 1, Iba1). On ultrasound, LPS group demonstrated elevated resistance indices, pulsatility indices and a greater occurrence of absent end-diastolic flow in the umbilical and uterine arteries. In the fetus, there was an increased cardiac Tei indices in the LPS group. MRI revealed decreased volume of placenta in the LPS group associated with placental thinning and placental endothelial damage on immunohistochemistry. Decreased fibrinogen content and more thrombi staining in placenta exposed to maternal LPS indicated the hypercoagulability. Furthermore, the expression of Iba1was significantly associated with placental thickness (r = -0.7890, Pearson correlation coefficient). Our data indicate that IUI can trigger events leading to maternal placental malperfusion and fetal vessel resistance, as well as predispose the developing fetus to cardiac dysfunction and brain damage. Furthermore, our data suggest that prenatal ultrasound can be a real-time clinical tool for assessing fetal risk for adverse neurologic outcomes following the potential IUI exposure.
Collapse
Affiliation(s)
- Solange N. Eloundou
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - JiYeon Lee
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Dan Wu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Mia C. Feller
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Maide Ozen
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Yan Zhu
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Misun Hwang
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Bei Jia
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Han Xie
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Julia L. Clemens
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Michael W. McLane
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Samar AlSaggaf
- Department of Pathology, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Nita Nair
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins University, School of Public Health, Baltimore, MD, United States of America
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Center on Early Life Origins of Disease, Johns Hopkins University, School of Public Health, Baltimore, MD, United States of America
| | - Ernest M. Graham
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - Ahmet Baschat
- Fetal Therapy, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, United States of America
| | - 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, MD, United States of America
- * E-mail:
| |
Collapse
|
19
|
Musilova I, Spacek R, Stranik J, Jacobsson B, Kacerovsky M. Fetal Portal System Flowmetry and Intra-Amniotic Inflammation in Preterm Prelabor Rupture of Membranes. Fetal Diagn Ther 2019; 46:323-332. [PMID: 30889602 DOI: 10.1159/000496203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 12/13/2018] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To determine the pulsatility index (PI) in the fetal splenic vein, the main portal vein, the left portal vein, and the ductus venosus with respect to the presence or absence of intra-amniotic inflammation (IAI) in preterm prelabor rupture of membranes (PPROM). METHOD Women with singleton pregnancies and PPROM, ranging in gestational age from 22+0 to 36+6 weeks, were included. Amniotic fluid samples were obtained by transabdominal amniocentesis and the amniotic fluid level of interleukin-6 (IL-6) was assessed by a point-of-care test. Doppler examination of the selected veins was performed, and the PI was assessed. IAI was defined as amniotic fluid levels of IL-6 ≥745 pg/mL. RESULTS In total, 42 women were included. Fetuses with IAI compared with those without IAI exhibited a higher PI in the splenic vein (p = 0.005) and the main portal vein (p = 0.05). No differences were observed in the left portal vein PI (p = 0.36) and the ductus venosus PI (p = 0.98). CONCLUSION IAI was associated with increased fetal splenic vein PI and main portal vein PI in PPROM. The absence of changes in the left portal vein PI and ductus venosus PI supports the local cause of the finding.
Collapse
Affiliation(s)
- Ivana Musilova
- Department of Obstetrics and Gynecology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital in Hradec Kralove, Hradec Kralove, Czechia,
| | - Richard Spacek
- Department of Gynecology and Obstetrics, University Hospital Ostrava, Ostrava, Czechia
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital in Hradec Kralove, Hradec Kralove, Czechia
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Department of Genetics and Bioinformatics, Area of Health Data and Digitalisation, Norwegian Institute of Public Health, Oslo, Norway
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital in Hradec Kralove, Hradec Kralove, Czechia.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czechia
| |
Collapse
|
20
|
Abstract
With the introduction of the electronic 4-dimensional and spatial-temporal image Correlation (e-STIC), it is now possible to obtain large volume datasets of the fetal heart that are virtually free of artifact. This allows the examiner to use a number of imaging modalities when recording the volumes that include two-dimensional real time, power and color Doppler, and B-flow images. Once the volumes are obtained, manipulation of the volume dataset allows the examiner to recreate views of the fetal heart that enable examination of cardiac anatomy. The value of this technology is that a volume of the fetal heart can be obtained, irrespective of the position of the fetus in utero, and manipulated to render images for interpretation and diagnosis. This article presents a summary of the various imaging techniques and provides clinical examples of its application used for prenatal diagnosis of congenital heart defects and abnormal cardiac function.
Collapse
Affiliation(s)
- Greggory R DeVore
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Fetal Diagnostic Centers, Pasadena, Tarzana, and Lancaster, CA, USA
| | - Gary Satou
- Division of Pediatric Cardiology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mark Sklansky
- Division of Pediatric Cardiology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
21
|
Mitchell T, MacDonald JW, Srinouanpranchanh S, Bammler TK, Merillat S, Boldenow E, Coleman M, Agnew K, Baldessari A, Stencel-Baerenwald JE, Tisoncik-Go J, Green RR, Gale MJ, Rajagopal L, Adams Waldorf KM. Evidence of cardiac involvement in the fetal inflammatory response syndrome: disruption of gene networks programming cardiac development in nonhuman primates. Am J Obstet Gynecol 2018; 218:438.e1-438.e16. [PMID: 29475580 PMCID: PMC6070341 DOI: 10.1016/j.ajog.2018.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/22/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Most early preterm births are associated with intraamniotic infection and inflammation, which can lead to systemic inflammation in the fetus. The fetal inflammatory response syndrome describes elevations in the fetal interleukin-6 level, which is a marker for inflammation and fetal organ injury. An understanding of the effects of inflammation on fetal cardiac development may lead to insight into the fetal origins of adult cardiovascular disease. OBJECTIVE The purpose of this study was to determine whether the fetal inflammatory response syndrome is associated with disruptions in gene networks that program fetal cardiac development. STUDY DESIGN We obtained fetal cardiac tissue after necropsy from a well-described pregnant nonhuman primate model (pigtail macaque, Macaca nemestrina) of intrauterine infection (n=5) and controls (n=5). Cases with the fetal inflammatory response syndrome (fetal plasma interleukin-6 >11 pg/mL) were induced by either choriodecidual inoculation of a hypervirulent group B streptococcus strain (n=4) or intraamniotic inoculation of Escherichia coli (n=1). RNA and protein were extracted from fetal hearts and profiled by microarray and Luminex (Millipore, Billerica, MA) for cytokine analysis, respectively. Results were validated by quantitative reverse transcriptase polymerase chain reaction. Statistical and bioinformatics analyses included single gene analysis, gene set analysis, Ingenuity Pathway Analysis (Qiagen, Valencia, CA), and Wilcoxon rank sum. RESULTS Severe fetal inflammation developed in the context of intraamniotic infection and a disseminated bacterial infection in the fetus. Interleukin-6 and -8 in fetal cardiac tissues were elevated significantly in fetal inflammatory response syndrome cases vs controls (P<.05). A total of 609 probe sets were expressed differentially (>1.5-fold change, P<.05) in the fetal heart (analysis of variance). Altered expression of select genes was validated by quantitative reverse transcriptase polymerase chain reaction that included several with known functions in cardiac injury, morphogenesis, angiogenesis, and tissue remodeling (eg, angiotensin I converting enzyme 2, STEAP family member 4, natriuretic peptide A, and secreted frizzled-related protein 4; all P<.05). Multiple gene sets and pathways that are involved in cardiac morphogenesis and vasculogenesis were downregulated significantly by gene set and Ingenuity Pathway Analysis (hallmark transforming growth factor beta signaling, cellular morphogenesis during differentiation, morphology of cardiovascular system; all P<.05). CONCLUSION Disruption of gene networks for cardiac morphogenesis and vasculogenesis occurred in the preterm fetal heart of nonhuman primates with preterm labor, intraamniotic infection, and severe fetal inflammation. Inflammatory injury to the fetal heart in utero may contribute to the development of heart disease later in life. Development of preterm labor therapeutics must also target fetal inflammation to lessen organ injury and potential long-term effects on cardiac function.
Collapse
Affiliation(s)
- Timothy Mitchell
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | | | - Theodor K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Sean Merillat
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Erica Boldenow
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | | | - Kathy Agnew
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA
| | - Audrey Baldessari
- Washington National Primate Research Center, University of Washington, Seattle, WA
| | - Jennifer E Stencel-Baerenwald
- Department of Immunology, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Richard R Green
- Department of Immunology, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Michael J Gale
- Department of Immunology, University of Washington, Seattle, WA; Department of Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Lakshmi Rajagopal
- Department of Pediatrics, University of Washington, Seattle, WA; Department of Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA; Department of Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Sahlgrenska Academy, Gothenburg, Sweden.
| |
Collapse
|
22
|
Rafferty AR, McGrory L, Cheung M, Rogerson S, Ziannino D, Pyman J, Davis PG, Burgner D. Inflammation, lipids and aortic intima-media thickness in newborns following chorioamnionitis. Acta Paediatr 2016; 105:e300-6. [PMID: 27002899 DOI: 10.1111/apa.13410] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 03/10/2016] [Accepted: 03/17/2016] [Indexed: 12/23/2022]
Abstract
AIM This study investigated whether chorioamnionitis was associated with increased inflammation, dyslipidaemia and adverse cardiovascular phenotypes in the immediate postnatal period. METHODS This prospective case-control study included preterm infants (30(+0) -35(+6) weeks gestational age, GA) whose mothers did not have pregnancy-related conditions that may influence outcomes. Chorioamnionitis was diagnosed by placental histology, and infants were divided retrospectively into cases (chorioamnionitis-exposed) and controls (unexposed). Serum high-sensitivity C-reactive protein (hsCRP), lipid profile, far-wall abdominal aortic intima-media thickness (aIMT) and blood pressure (BP) were measured in the first week of life. RESULTS There were 20 (16 male, mean GA 32.4 weeks) cases and 31 (12 male, mean GA 32.6 weeks) controls. Histological chorioamnionitis was associated with a significant increase in hsCRP and a non-significant trend towards an adverse lipid profile. There was no evidence of differences in aIMT or BP. CONCLUSION Preterm infants exposed to chorioamnionitis have greater postnatal inflammation. There were no early postnatal differences in aIMT or BP. The inflammatory stimulus of chorioamnionitis late in gestation may be of insufficient intensity and duration to result in immediate postnatal alterations to arterial structure. Cardiovascular follow-up of infants exposed to chorioamnionitis may identify differential risk trajectories and subsequent inflammatory responses.
Collapse
Affiliation(s)
- Anthony R. Rafferty
- Newborn Research Centre; Royal Women's Hospital; Parkville Australia
- Murdoch Childrens Research Institute; Parkville Australia
| | - Lorraine McGrory
- Newborn Research Centre; Royal Women's Hospital; Parkville Australia
- Murdoch Childrens Research Institute; Parkville Australia
- University of Dundee; Dundee United Kingdom
| | - Michael Cheung
- Murdoch Childrens Research Institute; Parkville Australia
- Department of Paediatrics; The University of Melbourne; Parkville Australia
| | - Sheryle Rogerson
- Newborn Research Centre; Royal Women's Hospital; Parkville Australia
| | - Diana Ziannino
- Murdoch Childrens Research Institute; Parkville Australia
| | - Jan Pyman
- Anatomical Pathology; The Royal Women's Hospital; Parkville Australia
| | - Peter G. Davis
- Newborn Research Centre; Royal Women's Hospital; Parkville Australia
- Murdoch Childrens Research Institute; Parkville Australia
- Department of Paediatrics; The University of Melbourne; Parkville Australia
| | - David Burgner
- Murdoch Childrens Research Institute; Parkville Australia
- Department of Paediatrics; The University of Melbourne; Parkville Australia
- Department of Paediatrics; Monash University; Clayton Australia
| |
Collapse
|
23
|
Mastrolia SA, Erez O, Loverro G, Di Naro E, Weintraub AY, Tirosh D, Baron J, Hershkovitz R. Ultrasonographic approach to diagnosis of fetal inflammatory response syndrome: a tool for at-risk fetuses? Am J Obstet Gynecol 2016; 215:9-20. [PMID: 26821337 DOI: 10.1016/j.ajog.2016.01.164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/14/2016] [Accepted: 01/19/2016] [Indexed: 12/11/2022]
Abstract
Preterm parturition is a syndrome that may result from many underlying mechanisms. Infection and inflammation are the prominent ones. Intrauterine infection and inflammation have an effect akin to sepsis, and that is similar to systemic inflammatory response in adults. Indeed, there is evidence to support the association of a fetal inflammatory response syndrome (FIRS) to systemic infection and inflammation. The utilization of invasive procedures for the prenatal diagnosis of FIRS is associated with a risk for complications resulting from the invasive method. The progress in the imaging quality of obstetrical ultrasound and the development of novel methods for functional anatomical assessment of the fetal organs may help to identify, noninvasively, fetuses at risk for FIRS in patients presenting with preterm labor. We review the studies describing advanced sonographic modalities and the imaging findings in the heart, thymus, kidney, adrenal glands, and spleen of these fetuses.
Collapse
Affiliation(s)
- Salvatore Andrea Mastrolia
- Department of Obstetrics and Gynecology, Azienda Ospedaliero-Universitaria Policlinico di Bari, School of Medicine, University of Bari "Aldo Moro," Bari, Italy; US Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel.
| | - Offer Erez
- Maternity Department D and Obstetrical Day Care Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, School of Medicine, Beer Sheva, Israel
| | - Giuseppe Loverro
- Department of Obstetrics and Gynecology, Azienda Ospedaliero-Universitaria Policlinico di Bari, School of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Edoardo Di Naro
- US Unit, Department of Obstetrics and Gynecology, Azienda Ospedaliero-Universitaria Policlinico di Bari, School of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Adi Yehuda Weintraub
- US Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Dan Tirosh
- US Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Joel Baron
- US Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Reli Hershkovitz
- US Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
24
|
Sanhal CY, Kara O, Yucel A. Can fetal left ventricular modified myocardial performance index predict adverse perinatal outcomes in intrahepatic cholestasis of pregnancy? J Matern Fetal Neonatal Med 2016; 30:911-916. [PMID: 27186866 DOI: 10.1080/14767058.2016.1190824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate fetal left ventricular function using the left ventricular modified myocardial performance index (mod-MPI) and E wave/A wave peak velocity (E/A) ratio, and to explore the success of mod-MPI in the prediction of adverse perinatal outcomes in intrahepatic cholestasis of pregnancy (ICP). METHODS Forty-one ICP cases were compared with 41 gestational age-matched healthy controls. Opening and closing clicks of the mitral and aortic valves were used to define the three time periods [ejection time (ET), isovolumetric contraction time (ICT) and isovolumetric relaxation time (IRT)], which were employed in the calculation of mod-MPI [mod-MPI = (ICT + IRT)/ET]. The E/A ratio was calculated as well. RESULTS Fetal left ventricular mod-MPI values were significantly higher in the ICP group compared to controls (0.56 ± 0.09 versus 0.37 ± 0.04, p < 0.001), whereas the E/A ratio was lower (0.62 ± 0.11 versus 0.69 ± 0.10, p = 0.011). The optimal cutoff level for mod-MPI in prediction of adverse perinatal outcomes was >0.48 [sensitivity: 81.8%, specificity: 67.6%, area under the curve (AUC): 0.750, 95% CI: 0.613-0.887, p = 0.008]. CONCLUSIONS Fetuses of ICP cases have significant left ventricular dysfunction. Mod-MPI can be used in the prediction of adverse perinatal outcomes in ICP.
Collapse
Affiliation(s)
- Cem Yasar Sanhal
- a Department of Perinatology , Zekai Tahir Burak Women's Health Care, Training and Research Hospital , Ankara , Turkey
| | - Ozgur Kara
- a Department of Perinatology , Zekai Tahir Burak Women's Health Care, Training and Research Hospital , Ankara , Turkey
| | - Aykan Yucel
- a Department of Perinatology , Zekai Tahir Burak Women's Health Care, Training and Research Hospital , Ankara , Turkey
| |
Collapse
|
25
|
Zidovudine treatment in HIV-infected pregnant women is associated with fetal cardiac remodelling. AIDS 2016; 30:1393-401. [PMID: 26919731 DOI: 10.1097/qad.0000000000001066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To evaluate the cardiac structure and function of the fetuses of pregnant women with HIV infection on combined antiretroviral treatment (cART) and the HIV-related and nonrelated determinants of abnormal findings. DESIGN A prospective cohort study including 42-noninfected fetuses from HIV pregnant women on cART and 84 fetuses from non-HIV-infected women. METHODS Fetal echocardiography was performed at 26-32 weeks of pregnancy to assess cardiac structure and function. The impact of maternal and perinatal factors on fetal cardiac remodelling was evaluated by multivariate regression analysis. RESULTS Fetuses from HIV pregnant women on cART presented larger hearts and pericardial effusion together with thicker myocardial septal walls (mean 3.56 mm (SD 0.88) vs non-HIV mean 2.75 mm (SD 0.77); P = 0.002) and smaller left ventricular cavities (10.81 mm (SD 2.28) vs 12.3 mm (SD 2.54); P = 0.033). Fetuses from HIV women also presented signs of systolic (mitral systolic annular peak velocity 5.85 cm/s (SD 0.77) vs non-HIV 6.25 cm/s (SD 0.97); P = 0.007) and diastolic (isovolumic relaxation time 52 ms (SD 8.91) vs non-HIV 45 ms (SD 7.98); P < 0.001) dysfunction. In the multivariate analysis, maternal treatment with zidovudine was the only factor significantly associated with fetal cardiac changes (P = 0.014). CONCLUSION Fetuses from HIV-infected mothers on cART have cardiac remodelling and dysfunction, which might explain the cardiovascular changes described in childhood. Fetal cardiac remodelling was essentially associated with maternal treatment with zidovudine which challenges its use during pregnancy.
Collapse
|
26
|
Accordino F, Consonni S, Fedeli T, Kullman G, Moltrasio F, Ghidini A, Locatelli A. Risk factors for cerebral palsy in PPROM and preterm delivery with intact membranes (.). J Matern Fetal Neonatal Med 2016; 29:3854-9. [PMID: 26919411 DOI: 10.3109/14767058.2016.1149562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Gestational age (GA) at delivery and spontaneous prematurity are independent risk factors for cerebral palsy (CP). The aim of this study is to investigate perinatal risk factors for CP in spontaneous preterm delivery. METHODS A retrospective cohort study of all single pregnancies complicated by spontaneous preterm labor (PTL) or PPROM with delivery at <34 weeks from January 2006 to December 2012 was performed. We compared demographic, obstetric, neonatal, and placental histology variables in cases of spontaneous preterm birth in reference to the development of CP. Statistical analysis included chi-square, one-way ANOVA and logistic regression analysis. p < 0.05 was considered significant. RESULTS Two hundred sixty-one women were included for this study. Of 249 survivors, 5 babies died during the first year of life, 52 did not fulfill the inclusion criteria for neurologic follow-up, and 24 were lost to follow up. Thus 168 infants in the study cohort underwent neurologic follow-up. We observed 26 cases of CP. Factors related to CP were lower GA at PROM (p = 0.007) and longer latency from PPROM to delivery (p = 0.002) in the PPROM group, lower GA at delivery (p < 0.001) and presence of funisitis (p <0.001) in the PTL group. CONCLUSIONS GA at membrane rupture in PPROM and GA at delivery in PTL are significantly associated with CP. A process leading to neurological damage may be initiated at the moment of membranes rupture in cases of PPROM and at the time of PTL in the group with intact membranes.
Collapse
Affiliation(s)
- Federica Accordino
- a Department of Obstetrics and Gynecology , San Gerardo Hospital MBBM Foundation, University of Milano-Bicocca , Monza , Italy
| | - Sara Consonni
- a Department of Obstetrics and Gynecology , San Gerardo Hospital MBBM Foundation, University of Milano-Bicocca , Monza , Italy
| | - Tiziana Fedeli
- b Department of Neonatology , San Gerardo Hospital, University of Milano-Bicocca , Monza , Italy
| | - Gaia Kullman
- c Department of Child Neurology and Psychiatry , San Gerardo Hospital, University of Milano-Bicocca , Monza , Italy
| | - Francesca Moltrasio
- d Department of Pathology , San Gerardo Hospital, University of Milano-Bicocca , Monza , Italy , and
| | | | - Anna Locatelli
- a Department of Obstetrics and Gynecology , San Gerardo Hospital MBBM Foundation, University of Milano-Bicocca , Monza , Italy
| |
Collapse
|
27
|
Wu TW, Azhibekov T, Seri I. Transitional Hemodynamics in Preterm Neonates: Clinical Relevance. Pediatr Neonatol 2016; 57:7-18. [PMID: 26482579 DOI: 10.1016/j.pedneo.2015.07.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Each newborn enters this world facing tremendous respiratory, hemodynamic and neuroendocrine challenges while going through drastic physiological changes during the process of adaption from fetal to postnatal life. Even though the vast majority of term infants transition smoothly without apparent consequences, this task becomes increasingly arduous for the extremely preterm infant. METHODS & RESULTS This article reviews the physiology and pathophysiology of cardiovascular adaptation of the very preterm neonate. In particular it describes the physiology of fetal circulation, summarizes the hemodynamic changes occurring during preterm births and discusses the impact of the most frequently seen clinical scenarios that place additional burden on the premature infant during immediate transition. Finally an emphasis is placed on discussing common clinical dilemmas and practical aspects of developmental hemodynamics such as neonatal hypotension and patent ductus arteriosus; clinical presentations the neonatologist encounters on a daily basis. CONCLUSION The review provides a physiology-based view on the hemodynamics of the immediate postnatal transitional period.
Collapse
Affiliation(s)
- Tai-Wei Wu
- Division of Neonatology, Department of Pediatrics, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung College of Medicine, Taoyuan, Taiwan
| | - Timur Azhibekov
- Division of Neonatology, Department of Pediatrics, Children's Hospital Los Angeles, CA, USA; Center for Fetal and Neonatal Medicine LAC+USC Medical Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Istvan Seri
- Center of Excellence in Neonatology, Department of Pediatrics, Sidra Medical and Research Center and Weill Cornell Medical College, Doha, Qatar.
| |
Collapse
|
28
|
Sciaky-Tamir Y, Hershkovitz R, Mazor M, Shelef I, Erez O. The use of imaging technology in the assessment of the fetal inflammatory response syndrome-imaging of the fetal thymus. Prenat Diagn 2016; 35:413-9. [PMID: 25601186 DOI: 10.1002/pd.4560] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 12/07/2014] [Accepted: 01/09/2015] [Indexed: 12/28/2022]
Abstract
The fetal inflammatory response syndrome (FIRS) describes a state of extensive fetal multi organ involvement during chorioamnionitis, and is associated with grave implications on perinatal outcome. The syndrome has been linked to the preterm parturition syndrome and is associated with inflammation/infection processes in most of the fetal organs. The fetal thymus, a major organ in the developing immune system involutes during severe neonatal disease and has been shown to be smaller in fetuses with FIRS. Various methods for imaging of the fetal thymus and measurement are described. Currently the only method to diagnose FIRS prenatally is through amniocentesis. We suggest that women who are admitted with preterm labor with intact membranes and those with PPROM should have a detailed sonographic examination of the fetal thymus as a surrogate marker of fetal involvement in intrauterine infection/inflammation processes.
Collapse
Affiliation(s)
- Yael Sciaky-Tamir
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
29
|
Abstract
Cardiovascular disease continues to be the leading cause of global morbidity and mortality. Traditional risk factors account for only part of the attributable risk. The origins of atherosclerosis are in early life, a potential albeit largely unrecognized window of opportunity for early detection and treatment of subclinical cardiovascular disease. There are robust epidemiological data indicating that poor intrauterine growth and/or prematurity, and perinatal factors such as maternal hypercholesterolaemia, smoking, diabetes and obesity, are associated with adverse cardiovascular intermediate phenotypes in childhood and adulthood. Many of these early-life risk factors result in a heightened inflammatory state. Inflammation is a central mechanism in the development of atherosclerosis and cardiovascular disease, but few studies have investigated the role of overt perinatal infection and inflammation (chorioamnionitis) as a potential contributor to cardiovascular risk. Limited evidence from human and experimental models suggests an association between chorioamnionitis and cardiac and vascular dysfunction. Early life inflammatory events may be an important mechanism in the early development of cardiovascular risk and may provide insights into the associations between perinatal factors and adult cardiovascular disease. This review aims to summarise current data on the early life origins of atherosclerosis and cardiovascular disease, with particular focus on perinatal inflammation.
Collapse
|
30
|
Kim CJ, Romero R, Chaemsaithong P, Chaiyasit N, Yoon BH, Kim YM. Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am J Obstet Gynecol 2015; 213:S29-52. [PMID: 26428501 PMCID: PMC4774647 DOI: 10.1016/j.ajog.2015.08.040] [Citation(s) in RCA: 631] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/12/2015] [Accepted: 08/16/2015] [Indexed: 12/25/2022]
Abstract
Acute inflammatory lesions of the placenta consist of diffuse infiltration of neutrophils at different sites in the organ. These lesions include acute chorioamnionitis, funisitis, and chorionic vasculitis and represent a host response (maternal or fetal) to a chemotactic gradient in the amniotic cavity. While acute chorioamnionitis is evidence of a maternal host response, funisitis and chorionic vasculitis represent fetal inflammatory responses. Intraamniotic infection generally has been considered to be the cause of acute chorioamnionitis and funisitis; however, recent evidence indicates that "sterile" intraamniotic inflammation, which occurs in the absence of demonstrable microorganisms induced by "danger signals," is frequently associated with these lesions. In the context of intraamniotic infection, chemokines (such as interleukin-8 and granulocyte chemotactic protein) establish a gradient that favors the migration of neutrophils from the maternal or fetal circulation into the chorioamniotic membranes or umbilical cord, respectively. Danger signals that are released during the course of cellular stress or cell death can also induce the release of neutrophil chemokines. The prevalence of chorioamnionitis is a function of gestational age at birth, and present in 3-5% of term placentas and in 94% of placentas delivered at 21-24 weeks of gestation. The frequency is higher in patients with spontaneous labor, preterm labor, clinical chorioamnionitis (preterm or term), or ruptured membranes. Funisitis and chorionic vasculitis are the hallmarks of the fetal inflammatory response syndrome, a condition characterized by an elevation in the fetal plasma concentration of interleukin-6, and associated with the impending onset of preterm labor, a higher rate of neonatal morbidity (after adjustment for gestational age), and multiorgan fetal involvement. This syndrome is the counterpart of the systemic inflammatory response syndrome in adults: a risk factor for short- and long-term complications (ie, sterile inflammation in fetuses, neonatal sepsis, bronchopulmonary dysplasia, periventricular leukomalacia, and cerebral palsy). This article reviews the definition, pathogenesis, grading and staging, and clinical significance of the most common lesions in placental disease. Illustrations of the lesions and diagrams of the mechanisms of disease are provided.
Collapse
Affiliation(s)
- Chong Jai Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI.
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Noppadol Chaiyasit
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Bo Hyun Yoon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yeon Mee Kim
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
31
|
Tang X, Hernandez-Andrade E, Ahn H, Garcia M, Saker H, Korzeniewski SJ, Tarca AL, Yeo L, Hassan SS, Romero R. Intermediate Diastolic Velocity as a Parameter of Cardiac Dysfunction in Growth-Restricted Fetuses. Fetal Diagn Ther 2015; 39:28-39. [PMID: 26279291 DOI: 10.1159/000431321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/07/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To evaluate the intermediate intracardiac diastolic velocities in fetuses with growth restriction. METHODS Doppler waveforms of the two atrioventricular valves were obtained. Peak velocities of the E (early) and A (atrial) components, and the lowest intermediate velocity (IDV) between them, were measured in 400 normally grown and in 100 growth-restricted fetuses. The prevalence of abnormal IDV, E/IDV, and A/IDV ratios in fetuses presenting with perinatal death or acidemia at birth (pH ≤7.1) was estimated. RESULTS IDV was significantly lower and E/IDV ratios significantly higher in the two ventricles of growth-restricted fetuses with reduced diastolic velocities in the umbilical artery (p < 0.05). In 13 fetuses presenting with perinatal death or acidemia at birth, 11 (85%) had either an E/IDV or A/IDV ratio >95th percentile, whereas 5 (38%) showed absent or reversed atrial velocities in the ductus venosus (DV-ARAV; p < 0.04). Fetuses without DV-ARAV but with elevated E/IDV ratios in either ventricle were nearly 7-fold more likely to have perinatal demise or acidemia at birth (OR 6.9, 95% CI 1.4-34) than those with E/IDV ratios <95th percentile. CONCLUSION The E/IDV and A/IDV ratios in the two cardiac ventricles might provide information about the risk of perinatal demise or acidemia in growth-restricted fetuses.
Collapse
Affiliation(s)
- Xiangna Tang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Md. and Detroit, Mich., USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Romero R, Miranda J, Chaemsaithong P, Chaiworapongsa T, Kusanovic JP, Dong Z, Ahmed AI, Shaman M, Lannaman K, Yoon BH, Hassan SS, Kim CJ, Korzeniewski SJ, Yeo L, Kim YM. Sterile and microbial-associated intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2015; 28:1394-409. [PMID: 25190175 PMCID: PMC5371030 DOI: 10.3109/14767058.2014.958463] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The objectives of this study were to: (1) determine the amniotic fluid (AF) microbiology of patients with preterm prelabor rupture of membranes (PROM); and (2) examine the relationship between intra-amniotic inflammation with and without microorganisms (sterile inflammation) and adverse pregnancy outcomes in patients with preterm PROM. METHODS AF samples obtained from 59 women with preterm PROM were analyzed using cultivation techniques (for aerobic and anaerobic bacteria as well as genital mycoplasmas) and with broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry (PCR/ESI-MS). AF concentration of interleukin-6 (IL-6) was determined using ELISA. Results of both tests were correlated with AF IL-6 concentrations and the occurrence of adverse obstetrical/perinatal outcomes. RESULTS (1) PCR/ESI-MS, AF culture, and the combination of these two tests each identified microorganisms in 36% (21/59), 24% (14/59) and 41% (24/59) of women with preterm PROM, respectively; (2) the most frequent microorganisms found in the amniotic cavity were Sneathia species and Ureaplasma urealyticum; (3) the frequency of microbial-associated and sterile intra-amniotic inflammation was overall similar [ 29% (17/59)]: however, the prevalence of each differed according to the gestational age when PROM occurred; (4) the earlier the gestational age at preterm PROM, the higher the frequency of both microbial-associated and sterile intra-amniotic inflammation; (5) the intensity of the intra-amniotic inflammatory response against microorganisms is stronger when preterm PROM occurs early in pregnancy; and (6) the frequency of acute placental inflammation (histologic chorioamnionitis and/or funisitis) was significantly higher in patients with microbial-associated intra-amniotic inflammation than in those without intra-amniotic inflammation [93.3% (14/15) versus 38% (6/16); p = 0.001]. CONCLUSIONS (1) The frequency of microorganisms in preterm PROM is 40% using both cultivation techniques and PCR/ESI-MS; (2) PCR/ESI-MS identified microorganisms in the AF of 50% more women with preterm PROM than AF culture; and (3) sterile intra-amniotic inflammation was present in 29% of these patients, and it was as or more common than microbial-associated intra-amniotic inflammation among those presenting after, but not before, 24 weeks of gestation.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Jezid Miranda
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Juan P. Kusanovic
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Sótero del Río Hospital, Santiago, Chile
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Zhong Dong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
| | - Ahmed I. Ahmed
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Majid Shaman
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kia Lannaman
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chong J. Kim
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yeon Mee Kim
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA
- Department of Pathology, College of Medicine Inje University, Haeundae Paik Hospital, Seoul, Korea
| |
Collapse
|
33
|
Abehsera Davó D, Alcedo Olea R, Vegas García de Yébenes G, Sancha Naranjo M, Magdaleno Dans F, González González A. Corioamnionitis subclínica: un reto diagnóstico. A propósito de un caso. CLINICA E INVESTIGACION EN GINECOLOGIA Y OBSTETRICIA 2014. [DOI: 10.1016/j.gine.2013.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
34
|
Kim EN, Kim CJ, Park JW, Yoon BH. Acute funisitis is associated with distinct changes in fetal hematologic profile. J Matern Fetal Neonatal Med 2014; 28:588-93. [PMID: 24863633 DOI: 10.3109/14767058.2014.927426] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Acute funisitis is a histologic hallmark of the fetal inflammatory response syndrome. This study was conducted to examine the hematologic profiles of preterm newborns with funisitis. METHODS The hematologic profiles of umbilical cord blood obtained at birth were compared according to the presence or absence of acute funisitis in 197 preterm neonates (gestational age before 34 weeks) born to mothers with preterm labor with intact membranes or premature rupture of membranes. RESULTS (1) Funisitis was identified in 22.3% (44/197) of patients; (2) newborns with funisitis had higher median leukocyte, neutrophil, monocyte counts (p < 0.005 for each), higher rate of neutrophilia (p < 0.05), higher proportion of neutrophils among leukocytes and lower proportion of lymphocytes among leukocytes than those without funisitis (p < 0.01 for each); (3) newborns with funisitis had a significantly lower median RBC count, hemoglobin concentration and hematocrit than those without funisitis (p < 0.05 for each); (4) there was no significant difference in the median lymphocyte, eosinophil, basophil, NRBC and platelet counts between the two groups (p > 0.1 for each). CONCLUSIONS The hematologic profiles of preterm newborns with funisitis are characterized by increased total white blood cell, neutrophil, and monocyte counts and decreased RBC count and hemoglobin concentration.
Collapse
Affiliation(s)
- Eun Na Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine , Seoul , Republic of Korea
| | | | | | | |
Collapse
|
35
|
Romero R, Kadar N, Miranda J, Korzeniewski SJ, Schwartz AG, Chaemsaithong P, Rogers W, Soto E, Gotsch F, Yeo L, Hassan SS, Chaiworapongsa T. The diagnostic performance of the Mass Restricted (MR) score in the identification of microbial invasion of the amniotic cavity or intra-amniotic inflammation is not superior to amniotic fluid interleukin-6. J Matern Fetal Neonatal Med 2014; 27:757-69. [PMID: 24028673 PMCID: PMC5881917 DOI: 10.3109/14767058.2013.844123] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Intra-amniotic infection/inflammation are major causes of spontaneous preterm labor and delivery. However, diagnosis of intra-amniotic infection is challenging because most are subclinical and amniotic fluid (AF) cultures take several days before results are available. Several tests have been proposed for the rapid diagnosis of microbial invasion of the amniotic cavity (MIAC) or intra-amniotic inflammation. The aim of this study was to examine the diagnostic performance of the AF Mass Restricted (MR) score in comparison with interleukin-6 (IL-6) and matrix metalloproteinase-8 (MMP-8) for the identification of MIAC or inflammation. METHODS AF samples were collected from patients with singleton gestations and symptoms of preterm labor (n = 100). Intra-amniotic inflammation was defined as >100 white blood cells/mm(3) (WBCs) in AF; MIAC was defined as a positive AF culture. AF IL-6 and MMP-8 were determined using ELISA. The MR score was obtained using the Surface-Enhanced Laser Desorption Ionization Time of Flight (SELDI-TOF) mass spectrometry. Sensitivity and specificity were calculated and logistic regression models were fit to construct receiver-operating characteristic (ROC) curves for the identification of each outcome. The McNemar's test and paired sample non-parametric statistical techniques were used to test for differences in diagnostic performance metrics. RESULTS (1) The prevalence of MIAC and intra-amniotic inflammation was 34% (34/100) and 40% (40/100), respectively; (2) there were no significant differences in sensitivity of the three tests under study (MR score, IL-6 or MMP-8) in the identification of either MIAC or intra-amniotic inflammation (using the following cutoffs: MR score >2, IL-6 >11.4 ng/mL, and MMP-8 >23 ng/mL); (3) there was no significant difference in the sensitivity among the three tests for the same outcomes when the false positive rate was fixed at 15%; (4) the specificity for IL-6 was not significantly different from that of the MR score in identifying either MIAC or intra-amniotic inflammation when using previously reported thresholds; and (5) there were no significant differences in the area under the ROC curve when comparing the MR score, IL-6 or MMP-8 in the identification of these outcomes. CONCLUSIONS IL-6 and the MR score have equivalent diagnostic performance in the identification of MIAC or intra-amniotic inflammation. Selection from among these three tests (MR score, IL-6 and MMP-8) for diagnostic purposes should be based on factors such as availability, reproducibility, and cost. The MR score requires a protein chip and a SELDI-TOF instrument which are not widely available or considered "state of the art". In contrast, immunoassays for IL-6 can be performed in the majority of clinical laboratories.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Nicholas Kadar
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
| | - Jezid Miranda
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Alyse G. Schwartz
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Wade Rogers
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eleazar Soto
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Francesca Gotsch
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Integrata Verona, Ostetricia Ginecologia, Azienda Ospedaliera Universitaria, Verona, Italy
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
36
|
Stampalija T, Chaiworapongsa T, Romero R, Tarca AL, Bhatti G, Chiang PJ, Than NG, Ferrazzi E, Hassan SS, Yeo L. Soluble ST2, a modulator of the inflammatory response, in preterm and term labor. J Matern Fetal Neonatal Med 2013; 27:111-21. [PMID: 23688338 DOI: 10.3109/14767058.2013.806894] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Intra-amniotic infection/inflammation (IAI) is causally linked with spontaneous preterm labor and delivery. The ST2L receptor and its soluble form (sST2) are capable of binding to interleukin (IL)-33, a member of the IL-1 superfamily. Members of this cytokine family have been implicated in the onset of spontaneous preterm labor in the context of infection. Soluble ST2 has anti-inflammatory properties, and plasma concentrations are elevated in systemic inflammation, such as sepsis, acute pyelonephritis in pregnancy and the fetal inflammatory response syndrome. The aims of this study were to examine: (1) whether amniotic fluid concentrations of sST2 change with IAI, preterm, and term parturition; and (2) if mRNA expression of ST2 in the chorioamniotic membranes changes with acute histologic chorioamnionitis in women who deliver preterm. METHOD A cross-sectional study was conducted to determine amniotic fluid concentrations of sST2 in: (1) women with preterm labor (PTL) who delivered at term (n=49); (2) women with PTL who delivered preterm without IAI (n=21); (3) women with PTL who delivered preterm with IAI (n=31); (4) term pregnancies not in labor (n=13); and (5) term pregnancies in labor (n=43). The amniotic fluid concentration of sST2 was determined by ELISA. The mRNA expression of ST2 in the chorioamniotic membranes of women who delivered preterm with (n=24), and without acute histologic chorioamnionitis (n=19) was determined by qRT-PCR. RESULTS (1) Patients with PTL who delivered preterm with IAI had a lower median amniotic fluid concentration of sST2 compared to those with PTL who delivered preterm without IAI [median 410 ng/mL, inter-quartile range (IQR) 152-699 ng/mL versus median 825 ng/mL, IQR 493-1216 ng/mL; p=0.0003] and those with PTL who delivered at term [median 410 ng/mL, IQR 152-699 ng/mL versus median 673 ng/mL, IQR 468-1045 ng/mL; p=0.0003]; (2) no significant differences in the median amniotic fluid concentration of sST2 were observed between patients with PTL who delivered at term and those who delivered preterm without IAI (p=0.4), and between women at term in labor and those at term not in labor (p=0.9); (3) the mean mRNA expression of ST2 was 4-fold lower in women who delivered preterm with acute histologic chorioamnionitis than in those without this lesion (p=0.008). CONCLUSIONS The median sST2 amniotic fluid concentration and mRNA expression of ST2 by chorioamniotic membranes is lower in PTL associated with IAI and acute histologic chorioamnionitis than in PTL without these conditions. Changes in the median amniotic fluid sST2 concentration are not observed in preterm and term parturition without IAI. Thus, amniotic fluid sST2 in the presence of IAI behaves differently when compared to sST2 in the plasma of individuals affected by fetal inflammatory response syndrome, acute pyelonephritis in pregnancy, and adult sepsis. Decreased concentrations of sST2 in IAI are likely to promote a pro-inflammatory response, which is important for parturition in the context of infection.
Collapse
Affiliation(s)
- Tamara Stampalija
- Perinatology Research Branch, NICHD/NIH/DHHS , Bethesda, Maryland, and Detroit, Michigan , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Musilova I, Kacerovsky M, Andrys C, Kostal M, Slaba K, Jacobsson B. The fetal splenic vein flow pattern and fetal inflammatory response in the preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2013; 27:770-4. [DOI: 10.3109/14767058.2013.843665] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
38
|
Lee J, Romero R, Chaiworapongsa T, Dong Z, Tarca AL, Xu Y, Chiang PJ, Kusanovic JP, Hassan SS, Yeo L, Yoon BH, Than NG, Kim CJ. Characterization of the fetal blood transcriptome and proteome in maternal anti-fetal rejection: evidence of a distinct and novel type of human fetal systemic inflammatory response. Am J Reprod Immunol 2013; 70:265-84. [PMID: 23905683 DOI: 10.1111/aji.12142] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/07/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The human fetus is able to mount a systemic inflammatory response when exposed to microorganisms. This stereotypic response has been termed the 'fetal inflammatory response syndrome' (FIRS), defined as an elevation of fetal plasma interleukin-6 (IL-6). FIRS is frequently observed in patients whose preterm deliveries are associated with intra-amniotic infection, acute inflammatory lesions of the placenta, and a high rate of neonatal morbidity. Recently, a novel form of fetal systemic inflammation, characterized by an elevation of fetal plasma CXCL10, has been identified in patients with placental lesions consistent with 'maternal anti-fetal rejection'. These lesions include chronic chorioamnionitis, plasma cell deciduitis, and villitis of unknown etiology. In addition, positivity for human leukocyte antigen (HLA) panel-reactive antibodies (PRA) in maternal sera can also be used to increase the index of suspicion for maternal anti-fetal rejection. The purpose of this study was to determine (i) the frequency of pathologic lesions consistent with maternal anti-fetal rejection in term and spontaneous preterm births; (ii) the fetal serum concentration of CXCL10 in patients with and without evidence of maternal anti-fetal rejection; and (iii) the fetal blood transcriptome and proteome in cases with a fetal inflammatory response associated with maternal anti-fetal rejection. METHOD OF STUDY Maternal and fetal sera were obtained from normal term (n = 150) and spontaneous preterm births (n = 150). A fetal inflammatory response associated with maternal anti-fetal rejection was diagnosed when the patients met two or more of the following criteria: (i) presence of chronic placental inflammation; (ii) ≥80% of maternal HLA class I PRA positivity; and (iii) fetal serum CXCL10 concentration >75th percentile. Maternal HLA PRA was analyzed by flow cytometry. The concentrations of fetal CXCL10 and IL-6 were determined by ELISA. Transcriptome analysis was undertaken after the extraction of total RNA from white blood cells with a whole-genome DASL assay. Proteomic analysis of fetal serum was conducted by two-dimensional difference gel electrophoresis. Differential gene expression was considered significant when there was a P < 0.01 and a fold-change >1.5. RESULTS (i) The frequency of placental lesions consistent with maternal anti-fetal rejection was higher in patients with preterm deliveries than in those with term deliveries (56% versus 32%; P < 0.001); (ii) patients with spontaneous preterm births had a higher rate of maternal HLA PRA class I positivity than those who delivered at term (50% versus 32%; P = 0.002); (iii) fetuses born to mothers with positive maternal HLA PRA results had a higher median serum CXCL10 concentration than those with negative HLA PRA results (P < 0.001); (iv) the median serum CXCL10 concentration (but not IL-6) was higher in fetuses with placental lesions associated with maternal anti-fetal rejection than those without such lesions (P < 0.001); (v) a whole-genome DASL assay of fetal blood RNA demonstrated differential expression of 128 genes between fetuses with and without lesions associated with maternal anti-fetal rejection; and (vi) comparison of the fetal serum proteome demonstrated 20 proteins whose abundance differed between fetuses with and without lesions associated with maternal anti-fetal rejection. CONCLUSION We describe a systemic inflammatory response in human fetuses born to mothers with evidence of maternal anti-fetal rejection. The transcriptome and proteome of this novel type of fetal inflammatory response were different from that of FIRS type I (which is associated with acute infection/inflammation).
Collapse
Affiliation(s)
- Joonho Lee
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Stampalija T, Romero R, Korzeniewski SJ, Chaemsaithong P, Miranda J, Yeo L, Dong Z, Hassan SS, Chaiworapongsa T. Soluble ST2 in the fetal inflammatory response syndrome: in vivo evidence of activation of the anti-inflammatory limb of the immune response. J Matern Fetal Neonatal Med 2013; 26:1384-93. [PMID: 23488731 DOI: 10.3109/14767058.2013.784258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Inflammation is a mechanism of host response to infection, which can be harmful when inappropriately modulated. Soluble ST2 (sST2) is a decoy receptor of interleukin (IL)-33, and this complex modulates the balance in the Th1/Th2 immune response. Moreover, sST2 inhibits the production of pro-inflammatory cytokines in cooperation with an anti-inflammatory cytokine, IL-10. The objectives of this study were to: (1) determine whether umbilical cord plasma sST2 concentration differs between preterm neonates with and without funisitis and between those with and without the fetal inflammatory response syndrome (FIRS); and (2) evaluate the relationship between sST2 and IL-10 among neonates with funisitis and/or FIRS. METHODS Umbilical cord plasma was collected from neonates delivered prematurely due to preterm labor or preterm prelabor rupture of membranes with (n = 36), and without funisitis (n = 30). FIRS (umbilical cord IL-6 concentration ≥ 17.5 pg/mL) was identified in 29 neonates. Plasma sST2 and IL-10 concentrations were determined by enzyme linked immune sorbent assay. RESULTS The median umbilical cord plasma sST2 concentration was 6.7-fold higher in neonates with FIRS than in those without FIRS (median 44.6 ng/mL, interquartile range (IQR) 13.8-80.3 ng/mL versus median 6.7 ng/mL, IQR 5.6-20.1 ng/mL; p < 0.0001). Similarly, the median umbilical cord plasma sST2 concentration was 2.7-fold higher in neonates with funisitis than in those without funisitis (median 19.1 ng/mL; IQR 7.1-75.0 ng/mL versus median 7.2 ng/mL; IQR 5.9-23.1 ng/mL; p = 0.008). There was a strong positive correlation between sST2 and IL-10 in neonates with funisitis and/or FIRS (Spearman's Rho = 0.7, p < 0.0001). CONCLUSION FIRS and funisitis are associated with an elevation of umbilical cord plasma concentrations of soluble ST2. This protein represents an important mediator of the immune response in neonates diagnosed with FIRS by promoting an anti-inflammatory effect in association with IL-10.
Collapse
Affiliation(s)
- Tamara Stampalija
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD and Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tsiartas P, Kacerovsky M, Musilova I, Hornychova H, Cobo T, Sävman K, Jacobsson B. The association between histological chorioamnionitis, funisitis and neonatal outcome in women with preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2013; 26:1332-6. [PMID: 23489073 DOI: 10.3109/14767058.2013.784741] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To determine the impact of histological chorioamnionitis (HCA) and funisitis on neonatal outcome in preterm prelabor rupture of membranes (PPROM) pregnancies. METHODS Women with PPROM between 24 + 0 to 36 + 6 weeks of gestation, admitted to the Department of Obstetrics and Gynecology at the University Hospital Hradec Kralove in the Czech Republic, between July 2008 and October 2010, were enrolled in the study (n = 231). RESULTS The incidence of early-onset sepsis (EOS) differed significantly in neonates born to women with and without HCA, after adjustment for gestational age (11% versus 1%, p = 0.011). The incidence of EOS in neonates was also significantly different, after adjustment for gestational age, in cases with and without funisitis (18% versus 4%, p = 0.002). The same was also found for retinopathy of prematurity (ROP) cases with and without funisitis (23% versus 4%, p = 0.014), after adjustment for gestational age. CONCLUSIONS HCA and funisitis increase the risk of adverse perinatal outcome in PPROM pregnancies.
Collapse
Affiliation(s)
- Panagiotis Tsiartas
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
41
|
The consequences of chorioamnionitis: preterm birth and effects on development. J Pregnancy 2013; 2013:412831. [PMID: 23533760 PMCID: PMC3606792 DOI: 10.1155/2013/412831] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/28/2013] [Accepted: 02/10/2013] [Indexed: 11/18/2022] Open
Abstract
Preterm birth is a major cause of perinatal mortality and long-term morbidity. Chorioamnionitis is a common cause of preterm birth. Clinical chorioamnionitis, characterised by maternal fever, leukocytosis, tachycardia, uterine tenderness, and preterm rupture of membranes, is less common than subclinical/histologic chorioamnionitis, which is asymptomatic and defined by inflammation of the chorion, amnion, and placenta. Chorioamnionitis is often associated with a fetal inflammatory response. The fetal inflammatory response syndrome (FIRS) is defined by increased systemic inflammatory cytokine concentrations, funisitis, and fetal vasculitis. Clinical and epidemiological studies have demonstrated that FIRS leads to poor cardiorespiratory, neurological, and renal outcomes. These observations are further supported by experimental studies that have improved our understanding of the mechanisms responsible for these outcomes. This paper outlines clinical and experimental studies that have improved our current understanding of the mechanisms responsible for chorioamnionitis-induced preterm birth and explores the cellular and physiological mechanisms underlying poor cardiorespiratory, neural, retinal, and renal outcomes observed in preterm infants exposed to chorioamnionitis.
Collapse
|
42
|
Hamill N, Romero R, Hassan S, Lee W, Myers SA, Mittal P, Kusanovic JP, Balasubramaniam M, Chaiworapongsa T, Vaisbuch E, Espinoza J, Gotsch F, Goncalves LF, Mazaki-Tovi S, Erez O, Hernandez-Andrade E, Yeo L. The fetal cardiovascular response to increased placental vascular impedance to flow determined with 4-dimensional ultrasound using spatiotemporal image correlation and virtual organ computer-aided analysis. Am J Obstet Gynecol 2013; 208:153.e1-13. [PMID: 23220270 DOI: 10.1016/j.ajog.2012.11.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/05/2012] [Accepted: 11/05/2012] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We sought to determine if increased placental vascular impedance to flow is associated with changes in fetal cardiac function using spatiotemporal image correlation and virtual organ computer-aided analysis. STUDY DESIGN A cross-sectional study was performed in fetuses with umbilical artery pulsatility index >95th percentile (abnormal [ABN]). Ventricular volume (end-systole, end-diastole), stroke volume, cardiac output (CO), adjusted CO, and ejection fraction were compared to those of 184 normal fetuses. RESULTS A total of 34 fetuses were evaluated at a median gestational age of 28.3 (range, 20.6-36.9) weeks. Mean ventricular volumes were lower for ABN than normal cases (end-systole, end-diastole) with a proportionally greater decrease for left ventricular volume (vs right). Mean left and right stroke volume, CO, and adjusted CO were lower for ABN (vs normal) cases. Right ventricular volume, stroke volume, CO, and adjusted CO exceeded the left in ABN fetuses. Mean ejection fraction was greater for ABN than normal cases. Median left ejection fraction was greater (vs right) in ABN fetuses. CONCLUSION Increased placental vascular impedance to flow is associated with changes in fetal cardiac function.
Collapse
|
43
|
Musilova I, Kacerovsky M, Hornychova H, Kostal M, Jacobsson B. Pulsation of the fetal splenic vein--a potential ultrasound marker of histological chorioamnionitis and funisitis in women with preterm prelabor rupture of membranes. Acta Obstet Gynecol Scand 2012; 91:1119-23. [PMID: 22574855 DOI: 10.1111/j.1600-0412.2012.01450.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The fetal spleen is involved in the response to intrauterine infection and inflammation. The flow pattern of its vein is not pulsatile in normal conditions. The aim of the study was to determine whether the presence of histological chorioamnionitis and funisitis is associated with a continuous or pulsatile flow pattern in the fetal splenic vein. We performed a prospective study including 79 women with preterm prelabor rupture of membranes. We found a relation between pulsation in the splenic vein and histological chorioamnionitis (likelihood ratio 13.2), as well as funisitis (likelihood ratio 5.7). Ultrasound evaluation of the splenic vein could be a non-invasive tool for the prediction of these inflammatory complications.
Collapse
Affiliation(s)
- Ivana Musilova
- Department of Obstetrics and Gynecology, Faculty of Medicine Hradec Kralove, Charles University in Prague, University Hospital Hradec Kralove, Czech Republic
| | | | | | | | | |
Collapse
|
44
|
Savasan ZA, Chaiworapongsa T, Romero R, Hussein Y, Kusanovic JP, Xu Y, Dong Z, Kim CJ, Hassan SS. Interleukin-19 in fetal systemic inflammation. J Matern Fetal Neonatal Med 2012; 25:995-1005. [PMID: 21767236 PMCID: PMC3383927 DOI: 10.3109/14767058.2011.605917] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The fetal inflammatory response syndrome (FIRS) is considered the fetal counterpart of the systemic inflammatory response syndrome (SIRS), which can be caused by infection and non-infection-related insults. Although the initial response is mediated by pro-inflammatory signals, the control of this response is achieved by anti-inflammatory mediators which are essential for the successful outcome of the affected individual. Interleukin (IL)-19 is capable of stimulating the production of IL-10, a major anti-inflammatory cytokine, and is a potent inducer of the T-helper 2 (Th2) response. The aim of this study was to determine if there is a change in umbilical cord plasma IL-19 and IL-10 concentrations in preterm neonates with and without acute funisitis, the histologic counterpart of FIRS. METHODS A case-control study was conducted including 80 preterm neonates born after spontaneous labor. Neonates were classified according to the presence (n = 40) or absence of funisitis (n = 40), which is the pathologic hallmark of FIRS. Neonates in each group were also matched for gestational age. Umbilical cord plasma IL-19 and IL-10 concentrations were determined by ELISA. RESULTS 1) The median umbilical cord plasma IL-19 concentration was 2.5-fold higher in neonates with funisitis than in those without funisitis (median 87 pg/mL; range 20.6-412.6 pg/mL vs. median 37 pg/mL; range 0-101.7 pg/mL; p < 0.001); 2) newborns with funisitis had a significantly higher median umbilical cord plasma IL-10 concentration than those without funisitis (median 4 pg/mL; range 0-33.5 pg/mL vs. median 2 pg/mL; range 0-13.8 pg/mL; p < 0.001); and 3) the results were similar when we included only patients with funisitis who met the definition of FIRS by umbilical cord plasma IL-6 concentrations ≥ 17.5 pg/mL (p < 0.001). CONCLUSION IL-19 and IL-10 are parts of the immunologic response of FIRS. A subset of fetuses with FIRS had high umbilical cord plasma IL-19 concentrations. In utero exposure to high systemic concentrations of IL-19 may reprogram the immune response.
Collapse
Affiliation(s)
- Zeynep Alpay Savasan
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
| | - Youssef Hussein
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile and Center for Perinatal Research, Sótero del Río Hospital, Santiago, Chile
| | - Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
| | - Zhong Dong
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
- Department of Pathology, Wayne State University, Detroit, MI, United States
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, Michigan, United States
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| |
Collapse
|
45
|
Hernandez-Andrade E, Benavides-Serralde JA, Cruz-Martinez R, Welsh A, Mancilla-Ramirez J. Evaluation of conventional Doppler fetal cardiac function parameters: E/A ratios, outflow tracts, and myocardial performance index. Fetal Diagn Ther 2012; 32:22-9. [PMID: 22677618 DOI: 10.1159/000330792] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 07/09/2011] [Indexed: 12/27/2022]
Abstract
Structural evaluation of the fetal heart is well established. Functional evaluation using pulsed-wave Doppler may also be performed. E/A ratios express the relationship between the maximal velocities of the E and A waveforms of ventricular filling. In normal fetuses, E/A ratios are usually <1 but show a constant increment during gestation, mainly related to the increment of the E wave. In intrauterine growth restriction (IUGR) fetuses, E/A ratios are lower compared to values in normally grown fetuses at the same gestational age. Cardiac outflows provide information on the time-velocity integral that, combined with the vessel area, allows calculation of the left and right cardiac outputs. In normal fetuses there is a predominance of the right ventricle (55-60%) in contributing to the combined cardiac output. In IUGR fetuses this predominance shifts to the left ventricle in order to increase the flow to the upper part of the fetal body and brain. The myocardial performance index (MPI) also provides information on systolic and diastolic cardiac function. The MPI is an early and consistent marker of cardiac dysfunction which becomes altered in early stages of chronic hypoxia or in cases with cardiac overload such as in twin-to-twin transfusion syndrome.
Collapse
|
46
|
Crispi F, Gratacós E. Fetal cardiac function: technical considerations and potential research and clinical applications. Fetal Diagn Ther 2012; 32:47-64. [PMID: 22614129 DOI: 10.1159/000338003] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/07/2012] [Indexed: 12/12/2022]
Abstract
Fetal echocardiography was initially used to detect structural anomalies but has more recently also been proposed to assess fetal cardiac function. This review summarizes technical issues and limitations in fetal cardiac function evaluation, as well as its potential research and clinical applications. Functional echocardiography has been demonstrated to select high-risk populations and to be associated with outcome in several fetal conditions including intrauterine growth restriction, twin-to-twin transfusion syndrome, maternal diabetes, and congenital diaphragmatic hernia. Fetal heart evaluation is challenging due to the smallness and high heart rate of the fetus and restricted access to the fetus far from the transducer. Due to these limitations and differences in cardiac function which are related to fetal maturation, cardiovascular parameters should be validated in the fetus and used with caution. Despite these precautions, in expert hands and with appropriate ultrasound equipment, evaluation of cardiac function is feasible in most fetuses. Functional fetal echocardiography is a promising tool that may soon be incorporated into clinical practice. Research is warranted to further refine the contribution of fetal cardiac assessment to the diagnosis, monitoring, or prediction of outcomes in various fetal conditions.
Collapse
Affiliation(s)
- Fatima Crispi
- Department of Maternal-Fetal Medicine, Institut Clínic de Ginecologia, Obstetrícia i Neonatologia, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain.
| | | |
Collapse
|
47
|
Kim SY, Romero R, Tarca AL, Bhatti G, Kim CJ, Lee J, Elsey A, Than NG, Chaiworapongsa T, Hassan SS, Kang GH, Kim JS. Methylome of fetal and maternal monocytes and macrophages at the feto-maternal interface. Am J Reprod Immunol 2012; 68:8-27. [PMID: 22385097 DOI: 10.1111/j.1600-0897.2012.01108.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 01/19/2012] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Decidual macrophages (dMφ) of the mother and placental macrophages (Hofbauer cells, HC) of the fetus are deployed at a critical location: the feto-maternal interface. This study was conducted to compare the DNA methylome of maternal and fetal monocytes, dMφ, and HC and thereby to determine the immunobiological importance of DNA methylation in pregnancy. METHOD OF STUDY Paired samples were obtained from normal pregnant women at term not in labor and their neonates. Maternal monocytes (MMo) and fetal monocytes (FMo) were isolated from the peripheral blood of mothers and fetal cord blood, respectively. dMφ and HC were obtained from the decidua of fetal membranes and placentas, respectively. DNA methylation profiling was performed using the Illumina Infinium Human Methylation27 BeadChip. Quantitative real-time PCR and Western Blot were performed for validation experiments. RESULTS (i) Significant differences in DNA methylation were found in each comparison (MMo versus FMo, 65 loci; dMφ versus HC, 266 loci; MMo versus dMφ, 199 loci; FMo versus HC, 1030 loci). (ii) Many of the immune response-related genes were hypermethylated in fetal cells (FMo and HC) compared to maternal cells (MMo and dMφ). (iii) Genes encoding markers of classical macrophage activation were hypermethylated, and genes encoding alternative macrophage activation were hypomethylated in dMφ and HC compared to MMo and FMo, respectively. (iv) mRNA expressions of DNMT1, DNMT3A, and DNMT3B were significantly lower in dMφ than in HC. (v) 5-azacytidine treatment increased expression of INCA1 in dMφ. CONCLUSIONS The findings herein indicate that DNA methylation patterns change during monocyte-macrophage differentiation at the feto-maternal interface. It is also suggested that DNA methylation is an important component of the biological machinery conferring an anti-inflammatory phenotype to macrophages at the feto-maternal interface.
Collapse
Affiliation(s)
- Sun Young Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Seehase M, Gantert M, Ladenburger A, Garnier Y, Kunzmann S, Thomas W, Wirbelauer J, Speer CP, Kramer BW. Myocardial response in preterm fetal sheep exposed to systemic endotoxinaemia. Pediatr Res 2011; 70:242-6. [PMID: 21629153 DOI: 10.1203/pdr.0b013e318225fbcb] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Exposure of the fetus to antenatal inflammation can occur from chorioamnionitis, which may progress to a fetal inflammatory response syndrome (FIRS) and to fetal sepsis. We tested whether the fetal myocardium responded to systemic Gram-negative endotoxinaemia. We hypothesized that the myocardium would respond to inflammation by changes in hypoxia-inducible factor-α (HIF-1α), inducible NO-synthase (iNOS), Toll-like receptors 2 and 4 (TLR2 and TLR4), IL-6, and phosphorylated signal transducer and activator of transcription-3 (pSTAT3). To model systemic endotoxinaemia, fetal sheep were exposed to Gram-negative endotoxin or saline i.v. 3 d before preterm delivery at 113 d of gestation (term = 147 d). All endotoxin-exposed animals developed cardiac dysfunction within these 72 h. Cardiac mRNA and protein levels of HIF-1α and TLR2 and TLR4 mRNA increased, whereas STAT3 phosphorylation decreased significantly. IL-6 and iNOS mRNA remained unchanged. Fetal systemic endotoxinaemia induced myocardial inflammation by activating TLR2 and 4. The following cardiac dysfunction seems not to be mediated via cardiac iNOS.
Collapse
Affiliation(s)
- Matthias Seehase
- Department of Paediatrics, Maastricht University Medical Center, Maastricht NL-6202, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Myocardial dysfunction in fetuses exposed to intraamniotic infection: new insights from tissue Doppler and strain imaging. Am J Obstet Gynecol 2010; 203:459.e1-7. [PMID: 20691411 DOI: 10.1016/j.ajog.2010.06.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 05/02/2010] [Accepted: 06/15/2010] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The purpose of this study was to assess myocardial function of fetuses who were exposed to intraamniotic infection compared with fetuses of uncomplicated pregnancies by the application of tissue Doppler and strain rate (SR) imaging. STUDY DESIGN We evaluated the right ventricular function of fetuses with preterm premature rupture of membranes and proven intraamniotic infection (n = 12 fetuses) and healthy fetuses (n = 27). Tissue Doppler velocities during early diastolic relaxation (E(m)) and atrial contraction (A(m)) and early diastolic SR were measured as indices of diastolic function, whereas the peak systolic strain and SR were used as parameters of systolic function. RESULTS Fetuses with intraamniotic infection exhibit impairment in both diastolic and systolic performance, which was characterized by increased diastolic compliance (increased E(m)/A(m) ratio, increased early diastolic SR compared with the control fetuses), decreased systolic contractile function (reduced systolic strain and SR), and longitudinal myocardial dyskinesia. CONCLUSION New echocardiographic tools suggest that fetal heart is a target organ in the context of intraamniotic infection.
Collapse
|
50
|
Randis TM. Progress toward improved understanding of infection-related preterm birth. Clin Perinatol 2010; 37:677-88. [PMID: 20813278 DOI: 10.1016/j.clp.2010.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Current strategies to prevent infection-related preterm birth and its associated neonatal morbidities have had limited success. Improved understanding of the pathogen-host interactions underlying altered colonization of the lower genital tract is necessary before significant progress can be made. The application of novel diagnostic techniques such as broad range PCR and proteomic analysis contribute to our knowledge of the diversity and abundance of microbial species invading the amniotic cavity as well as the resultant inflammatory response. Preterm infants delivered following intrauterine infection may respond differently to subsequent infectious challenges in the neonatal intensive care unit.
Collapse
Affiliation(s)
- Tara M Randis
- Division of Neonatology, Columbia University Medical Center, 3959 Broadway, CHN 1201, New York, NY 10032, USA.
| |
Collapse
|